Hostname: page-component-76fb5796d-qxdb6 Total loading time: 0 Render date: 2024-04-29T08:46:23.175Z Has data issue: false hasContentIssue false

Canadian Consensus Guidelines for the Diagnosis and Treatment of Autoimmune Encephalitis in Adults

Published online by Cambridge University Press:  05 February 2024

Christopher Hahn*
Affiliation:
Department of Clinical Neurosciences, University of Calgary, Calgary, AB, Canada
Adrian Budhram
Affiliation:
Clinical Neurological Sciences, London Health Sciences Centre, London, ON, Canada Department of Pathology and Laboratory Medicine, Western University, London Health Sciences Centre, London, ON, Canada
Katayoun Alikhani
Affiliation:
Department of Clinical Neurosciences, University of Calgary, Calgary, AB, Canada
Nasser AlOhaly
Affiliation:
Division of Neurology, University of Toronto, Toronto, ON, Canada
Grayson Beecher
Affiliation:
Division of Neurology, University of Alberta, Edmonton, AB, Canada
Gregg Blevins
Affiliation:
Division of Neurology, University of Alberta, Edmonton, AB, Canada
John Brooks
Affiliation:
Division of Neurology, University of Toronto, Toronto, ON, Canada
Robert Carruthers
Affiliation:
Division of Neurology, University of British Columbia, Vancouver, BC, Canada
Jacynthe Comtois
Affiliation:
Neurosciences, Universite de Montreal Faculte de Medecine, Montreal, QC, Canada
Juthaporn Cowan
Affiliation:
Division of Infectious Diseases, Department of Medicine Ottawa Hospital, Ottawa, ON, Canada
Paula de Robles
Affiliation:
Department of Clinical Neurosciences, University of Calgary, Calgary, AB, Canada Department of Oncology, University of Calgary, Calgary, AB, Canada
Julien Hébert
Affiliation:
Division of Neurology, University of Toronto, Toronto, ON, Canada
Ronak K. Kapadia
Affiliation:
Department of Clinical Neurosciences, University of Calgary, Calgary, AB, Canada
Sarah Lapointe
Affiliation:
Neurosciences, Universite de Montreal Faculte de Medecine, Montreal, QC, Canada
Aaron Mackie
Affiliation:
Department of Psychiatry, University of Calgary, Calgary, AB, Canada
Warren Mason
Affiliation:
Division of Neurology, University of Toronto, Toronto, ON, Canada
Brienne McLane
Affiliation:
Department of Clinical Neurosciences, University of Calgary, Calgary, AB, Canada Department of Psychiatry, University of Calgary, Calgary, AB, Canada
Alexandra Muccilli
Affiliation:
Division of Neurology, University of Toronto, Toronto, ON, Canada
Ilia Poliakov
Affiliation:
Division of Neurology, University of Saskatchewan College of Medicine, Saskatoon, SK, Canada
Penelope Smyth
Affiliation:
Division of Neurology, University of Alberta, Edmonton, AB, Canada
Kimberly G. Williams
Affiliation:
Department of Psychiatry, University of Calgary, Calgary, AB, Canada
Christopher Uy
Affiliation:
Division of Neurology, University of British Columbia, Vancouver, BC, Canada
Jennifer A. McCombe
Affiliation:
Division of Neurology, University of Alberta, Edmonton, AB, Canada
*
Corresponding author: Christopher Hahn. E-mail: chahn@ucalgary.ca
Rights & Permissions [Opens in a new window]

Abstract

Autoimmune encephalitis is increasingly recognized as a neurologic cause of acute mental status changes with similar prevalence to infectious encephalitis. Despite rising awareness, approaches to diagnosis remain inconsistent and evidence for optimal treatment is limited. The following Canadian guidelines represent a consensus and evidence (where available) based approach to both the diagnosis and treatment of adult patients with autoimmune encephalitis. The guidelines were developed using a modified RAND process and included input from specialists in autoimmune neurology, neuropsychiatry and infectious diseases. These guidelines are targeted at front line clinicians and were created to provide a pragmatic and practical approach to managing such patients in the acute setting.

Résumé

RÉSUMÉ

Consensus canadien en ce qui a trait aux lignes directrices pour le diagnostic et le traitement de l’encéphalite auto-immune.

L’encéphalite auto-immune (EAI) est de plus en plus reconnue comme une cause neurologique de modifications aiguës de l’état mental dont la prévalence est semblable à celle de l’encéphalite infectieuse. Malgré une sensibilisation accrue, les approches diagnostiques demeurent incohérentes et les preuves garantissant un traitement optimal sont limitées. Les lignes directrices canadiennes représentent une approche consensuelle fondée sur des données probantes (lorsque ces dernières sont disponibles) en vue du diagnostic et du traitement de patients adultes atteints d’EAI. Elles ont été élaborées selon un processus RAND modifié et ont bénéficié de l’apport de spécialistes en neurologie auto-immune, en neuropsychiatrie et en maladies infectieuses. Ces lignes directrices s’adressent aux cliniciens de première ligne et ont été créées pour offrir une approche pragmatique et pratique de prise en charge des patients dans un contexte de soins aigus.

Type
Review Article
Creative Commons
Creative Common License - CCCreative Common License - BY
This is an Open Access article, distributed under the terms of the Creative Commons Attribution licence (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted re-use, distribution and reproduction, provided the original article is properly cited.
Copyright
© The Author(s), 2024. Published by Cambridge University Press on behalf of Canadian Neurological Sciences Federation

Introduction

Autoimmune encephalitis (AIE) is an inflammatory disease of the brain that classically presents with subacute-onset neurological symptoms. The symptoms associated with specific AIE entities are reflective of selective involvement of central nervous system (CNS) structures by the inflammatory process. AIE has been demonstrated to be at least equal in prevalence to infectious encephalitis with an incidence of 0.2–0.8 per 100,000-person years. Reference Dubey, Pittock and Kelly1,Reference Hébert, Riche and Vogrig2,Reference Gable, Sheriff, Dalmau, Tilley and Glaser3 AIE is increasingly being considered in the differential diagnosis of patients presenting with subacute onset of psychiatric symptoms, cognitive decline, and seizures, although misdiagnosis remains common Reference Flanagan, Geschwind and Lopez-Chiriboga4 .

Unfortunately, there is a paucity of clinical trial data to guide the diagnosis and treatment of AIE, and a lack of formally trained autoimmune neurologists both globally and in Canada. As a result, challenges persist in ensuring patients are accurately diagnosed in a timely manner, undergo complete evaluation, and are rapidly initiated on appropriate therapy when indicated. International best practice recommendations have been published to help address this gap, Reference Abboud, Probasco and Irani5 but lack in regional specificity and contain more detail than may be relevant for non-neuroimmunologists. The following guideline document has been developed as a practical and rapid reference for front line neurologists, internal medicine physicians, psychiatrists and other health care workers caring for patients with AIE in hospitals in Canada, while emphasizing throughout that input from a specialist in autoimmune neurology is valuable and should be sought when possible. Additionally, whenever applicable, this document has been tailored to reflect the reality in Canada around access to testing and treatment.

Patients with AIE can consume a disproportionate amount of healthcare resources. Reference Cohen, Sotoca and Gandhi6 As with stroke, we believe that early and effective treatment gives patients the best possible chance of recovery and early data suggests that delays in immunotherapy treatment result in a poor prognosis. Reference Balu, McCracken, Lancaster, Graus, Dalmau and Titulaer7Reference Titulaer, McCracken and Gabilondo9 Neurologists, in conjunction with patients and their families, can play an important role when advocating for prompt access to treatments.

Methodology

Canadian Consensus Guidelines for the Diagnosis and Treatment of Autoimmune Encephalitis in Adults were developed with input from neurologists and psychiatrists with interest and/or training in the field of neuroimmunology from across Canada, as well as an infectious disease physician where relevant. Physicians were identified through the Anti-NMDA Receptor Encephalitis Foundation Directory and Canadian Consortium of MS Clinics. Participants were divided into primary and secondary panels with the primary panel responsible for the comprehensive literature review and drafting of each section and the secondary panel providing initial section review. For diagnosis and management decisions that lack evidence, consensus was attempted using a modified RAND method (see appendix). Areas where consensus was not obtainable are indicated. The guidelines were additionally reviewed by a member of the public previously diagnosed with AIE and endorsed by the leadership of the Anti-NMDA Receptor Encephalitis Foundation.

Diagnosis of autoimmune encephalitis

Epidemiologic considerations

The prevalence of AIE is increasing, likely related to increasing recognition of the diagnosis. Reference Dubey, Pittock and Kelly1,Reference Hébert, Riche and Vogrig2 AIE occurs in people of all ages; however, there are antibody-specific age predilections. For example, N-methyl-d-aspartate receptor (NMDAR) antibody encephalitis occurs more frequently in children and young women Reference Hébert, Riche and Vogrig2,Reference Gable, Sheriff, Dalmau, Tilley and Glaser3 while leucine-rich glioma-inactivated 1 (LGI1) antibody encephalitis predominates in older men. Reference Gable, Sheriff, Dalmau, Tilley and Glaser3,Reference Irani, Alexander and Waters10 A personal or family history of autoimmune disease may also be a predisposing factor. Reference Mittal, Rabinstein, Hocker, Pittock, Wijdicks and McKeon12 Other known risk factors for AIE include preceding herpes simplex encephalitis and other forms of infectious encephalitis. Reference Armangue, Spatola and Vlagea13Reference Predkele and Mednieks17 Immune checkpoint inhibitors (ICIs), used in the treatment of a growing number of malignancies, have also been identified as conferring increased risk for diverse neurological autoimmune complications, including AIE. Reference Marini, Bernardini and Gigli18

Presenting symptoms

AIE typically presents with a subacute onset of symptoms, however, certain neural antibody-associated syndromes may have a more indolent course, such as LGI1 antibody, contactin-associated protein-like 2 (CASPR2) antibody, glutamic acid decarboxylase-65 (GAD65) antibody and dipeptidyl-peptidase-like protein 6 (DPPX) antibody encephalitis. Reference Abboud, Rossman and Mealy19Reference van Sonderen, Thijs and Coenders22 An infectious prodrome often occurs days to weeks preceding neuropsychiatric manifestations. Other non-specific prodromal symptoms can include headache, fever, fatigue, sleep disturbance, weight loss, and early psychiatric manifestations. Reference Titulaer, McCracken and Gabilondo9

A detailed history and physical examination are critical to achieving a timely diagnosis of AIE. In one retrospective series examining diagnoses at hospital admission in patients subsequently confirmed as having AIE, only a third were initially thought to have encephalitis; more than 50% of those were initially felt to be of infectious origin. Reference Baumgartner, Rauer and Hottenrott23 This is reflective of the heterogeneity of clinical presentations of AIE. Misdiagnosis is also not uncommon; a recent study of patients referred to AIE specialty clinics reported a misdiagnosis rate of 27% among patients with an original referral diagnosis of AIE. Reference Flanagan, Geschwind and Lopez-Chiriboga4 Factors associated with misdiagnosis include overreliance on isolated serum antibody positivity, and classification of encephalopathy based on non-specific cognitive, psychiatric and/or functional symptoms. Reference Flanagan, Geschwind and Lopez-Chiriboga4

The most common presenting symptoms of AIE are reflected by diagnostic criteria (see below) and consist of acute mental status changes (including cognitive decline and working memory deficits), seizures, and psychiatric symptoms (commonly paranoia, agitation, personality changes and/or hallucinations). Reference Kayser, Titulaer, Gresa-Arribas and Dalmau24Reference Uy, Binks and Irani26 Other symptoms may be seen depending on the phenotype/antibody association and common phenotypes of AIE include, but are not limited to, limbic encephalitis, cerebellar or brainstem syndromes, and encephalomyelitis. Reference Uy, Binks and Irani26 While significant overlap exists in the diverse phenotypes of AIE, certain features should prompt suspicion for a particular antibody. Faciobrachial dystonic seizures, for example, are strongly associated with LGI1 antibody encephalitis. Reference Irani, Michell and Lang27

Diagnostic criteria

Diagnostic criteria for AIE were first proposed by Graus et al. in 2016 (Figure 1). Reference Graus, Titulaer and Balu28 An AIE diagnosis can be classified according to the degree of diagnostic certainty including possible, probable, and definite. While the diagnostic criteria can be helpful in identifying patients in whom a diagnosis of AIE should be considered, they also serve to identify those patients in whom AIE is less likely. In cases where criteria are met only for “possible” AIE after investigations including neural antibody testing are complete, early involvement of a specialist in autoimmune neurology is important. This is especially true if considering a trial of immunotherapy.

Figure 1: Diagnostic criteria for possible autoimmune encephalitis.

Based on the above diagnostic criteria, transition from “possible” to “probable” AIE can be achieved by meeting more specific phenotypic criteria for NMDAR antibody encephalitis or antibody-negative AIE. A diagnosis of definite AIE requires neural specific antibody positivity or meeting specific criteria for limbic encephalitis/acute disseminated encephalomyelitis (ADEM)/Bickerstaff’s encephalitis in addition to the criteria for “possible” AIE (Figure 2).

Figure 2: Diagnostic progression in autoimmune encephalitis (excluding ADEM, Bickerstaff’s). ADEM = Acute disseminated encephalomyelitis; CNS = central nervous system; CSF = cerebral spinal fluid; EEG = electroencephalography; GAD65 = glutamic acid decarboxylase-65; IgG = immunoglobulin G; MRI = magnetic resonance imaging (of the brain); OGBs = oligoclonal bands; PET = positron emission tomography (of the brain).

While psychiatric symptoms are common in AIE Reference Titulaer, McCracken and Gabilondo9 caution must be exercised in cases of isolated psychiatric manifestations. Some studies report the identification of NMDAR antibody in patients with schizophrenia and psychotic disorders, as well as in healthy control subjects; however, antibody detection in these series typically relied on serum testing alone or evaluated antibody subtypes/sub-classes which are not known to be pathogenic. Reference Jernbom Falk, Galletly and Just29Reference Luo, Li and Jiang31 More recent studies have confirmed that NMDAR antibodies are highly specific for AIE when detected in CSF in addition to serum. Reference Guasp, Giné-Servén and Maudes32 In recognition of this, while balancing the desire to not miss any patient with an autoimmune cause of psychiatric symptoms, it has been suggested that certain factors should prompt further workup in patients with a first psychotic episode. This includes rapid progression, the presence of concurrent neurological symptoms, fluctuating catatonia, refractoriness to antipsychotic medications, or abnormal paraclinical investigations. Reference Herken and Prüss33

Practical Tips on Diagnosis:

  1. 1. AIE can have variable clinical presentations, and a thorough history and physical exam are essential in making an accurate diagnosis.

  2. 2. Caution must be taken when undertaking treatment in patients who only meet criteria for “Possible AIE” despite completion of investigations.

Initial investigations

Once AIE is suspected, clinicians need to: 1) work towards obtaining evidence supporting this diagnosis and; 2) exclude mimics. This is an evolving process in which tests are processed in parallel with decisions on acute therapies. Recommended for all patients are cerebrospinal fluid (CSF) examination, brain magnetic resonance imaging (MRI), electroencephalography (EEG), and panel-based neural antibody testing. Reference Abboud, Probasco and Irani5,Reference Graus, Titulaer and Balu28 Other paraclinical tests should also be considered in certain cases as detailed below.

Laboratory testing

Cerebrospinal fluid and serum

Please see Table 1 for an outline of suggested CSF and serum investigations for AIE. Reference Abboud, Probasco and Irani5,Reference Graus, Titulaer and Balu28,Reference Au, Latonas, Shameli, Auer and Hahn34 CSF examination is of vital importance in the workup of AIE. Infectious pathogens should be ruled out. Evidence of inflammation can be found by evaluating the protein, cellular profile, oligoclonal banding, and IgG index. Paired CSF and serum samples must be tested to identify CSF-specific oligoclonal bands. CSF findings most consistent with AIE include mild to moderate lymphocytic leukocytosis (generally <100 white blood cells), presence of CSF-specific oligoclonal bands, increased IgG index, and/or elevated protein. Reference Blinder and Lewerenz35 A significant proportion of people with AIE have normal CSF studies Reference Escudero, Guasp and Ariño36 and inclusion of testing for oligoclonal bands significantly increases sensitivity. Reference Hébert, Gros and Lapointe37 Glucose is typically normal in AIE although low glucose can be seen in autoimmune glial fibrillary acidic protein (GFAP) astrocytopathy. Reference Lan, Li, Ai and Luo38,Reference Zhu, Sun, Yang, Yu and Wang39 Testing for CJD should be sent only if there is high clinical suspicion. Repeat CSF examination can be considered if diagnostic uncertainty remains, as CSF findings may evolve with evolution of AIE. Reference Dalmau, Lancaster, Martinez-Hernandez, Rosenfeld and Balice-Gordon40

Table 1: Initial laboratory investigations of suspected AIE. Tests in bold are strongly recommended to establish the diagnosis and exclude common mimics. Optional tests are also listed if clinically indicated by presenting history

AFB = acid-fast bacillus; ANA = anti-nuclear antibody; ANCA = anti-nuclear cytoplasmic antibody; C3 = complement component 3; C4 = complement component 4; CMV = cytomegalovirus; CRP = C-reactive protein; CSF = cerebrospinal fluid; dsDNA = double stranded DNA; EBV = Epstein-Barr virus; GAD65 = glutamic acid decarboxylase-65; GFAP = glial fibrillary acidic glycoprotein; HHV-6 = human herpesvirus-6; HIV = human immunodeficiency virus; HSV = herpes simplex virus; IgG = immunoglobulin G; IgLON5 = IgLON Family Member 5; JCV = John Cunningham virus; MOG = myelin oligodendrocyte glycoprotein; PCR = polymerase chain reaction; SSA = Sjögren’s-syndrome-related antigen A; SSB = Sjögren’s-syndrome-related antigen B; TB = tuberculosis; TPO = thyroid peroxidase; TSH = thyroid stimulating hormone; VDRL = venereal disease research laboratory; VZV = varicella zoster virus.

**Hepatitis and tuberculosis testing is important for future treatment planning and should be obtained before use of steroid or IVIg whenever possible.

***Yield of flow cytometry without a history of hematological malignancy nor leptomeningeal gadolinium enhancement has been shown to be extremely low. Reference Au, Latonas, Shameli, Auer and Hahn34

Serum screening should incorporate a broad differential, excluding infectious, metabolic, and systemic autoimmune causes of encephalopathy. Of note, some systemic clues may exist to suggest underlying cause; for example, hyponatremia can be seen in 65% of cases of LGI1 antibody encephalitis. Reference van Sonderen, Thijs and Coenders22

Neural antibody testing

Neural antibody testing in patients with suspected AIE is invaluable to patient diagnosis and management. Reference Graus, Titulaer and Balu28,Reference Budhram, Dubey and Sechi41,Reference Ropper, Dalmau and Graus42 While rare clinical features are virtually pathognomonic for an individual neural antibody, many clinical and neuroimaging features of AIE can occur with various neural antibodies (e.g., memory deficits, psychiatric symptoms, seizures, medial temporal lobe T2-hyperintensities on MRI brain). Reference Abboud, Probasco and Irani5,Reference Irani, Michell and Lang27,Reference Graus, Titulaer and Balu28,Reference Budhram, Leung, Nicolle and Burneo43 Because of the potential for phenotypic overlap across antibody-associated presentations, comprehensive panel-based neural antibody testing in patients with suspected AIE is generally recommended over sequential antibody testing to maximize sensitivity and facilitate prompt diagnosis.

Sensitivity of serum is higher than CSF for some antibodies (e.g., LGI1 and CASPR2 antibodies) while sensitivity/specificity of CSF is higher than serum for others (e.g., NMDAR and GFAP antibodies). Reference Budhram, Yang, Bhayana, Mills and Dubey44Reference Gresa-Arribas, Titulaer and Torrents47 Furthermore, for rare/novel antibodies, the preferred specimen to test may not be well-established. For these reasons, testing both serum and CSF optimizes sensitivity and specificity. Assays commonly used for neural antibody detection include tissue indirect immunofluorescence/immunohistochemistry (TIIF/IHC), immunoblots, cell-based assays, radioimmunoassays and enzyme-linked immunosorbent assays (Table 2). Incorporation of TIIF/IHC is recommended to optimize sensitivity and specificity of neural antibody testing for patients with suspected AIE and should be considered standard of practice. Reference Graus, Vogrig and Muñiz-Castrillo63

Table 2: Test methodologies employed for neural antibody detection in patients with suspected autoimmune encephalitis

ADEM = acute disseminated encephalomyelitis; AMPAR = α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor; CASPR2 = contactin-associated protein-like 2; CRMP5 = collapsin response-mediator protein 5; DNER = delta/Notch-like epidermal growth factor-related receptor; DPPX = dipeptidyl-peptidase-like protein 6; FLAMES = FLAIR-hyperintense Lesions in Anti-MOG-associated Encephalitis with Seizures; GABA(B)R = γ-aminobutyric acid type B receptor; GAD65 = glutamic acid decarboxylase-65; GlyR = glycine receptor; IGLON5 = IgLON Family Member 5; LGI1 = leucine-rich glioma-inactivated 1; MOG = myelin oligodendrocyte glycoprotein; NMDAR = N-methyl-D-aspartate receptor; PERM = progressive encephalomyelitis with rigidity and myoclonus; SOX1 = SRY-box transcription factor 1; VGKC = voltage-gated potassium channel.

In Canada however, many centers rely on commercial laboratories that do not incorporate TIIF/IHC in their testing algorithms. As such, the results reported are not confirmed with 2 testing methodologies, which may result in a high proportion of false-positive results. Reference Budhram, Nicolle and Yang60Reference Ruiz-García, Martínez-Hernández, Saiz, Dalmau and Graus62,Reference Budhram, Mirian, McFadden, Edmond, Bhayana and Yang65 Similarly, more limited testing can miss antibodies that are not included in, or are sub-optimally detected by, standard commercially available panels; such antibodies may only be detected if TIIF/IHC along with confirmatory assays available at specialized centers are used. Reference McCracken, Zhang and Greene81,Reference Sabater, Saiz, Dalmau and Graus82 As such, it is highly recommended to involve a specialist in autoimmune neurology when a positive neural antibody result is found in a patient with an atypical clinical presentation or when no antibody is detected in a patient in whom AIE is strongly suspected, so they can review results and direct further testing at specialized laboratories.

Importantly, awaiting neural antibody results should not delay consideration of empiric immunotherapy if there is a high index of suspicion for AIE and reasonable exclusion of alternative diagnoses. Once available, a positive neural antibody result can serve to confirm the diagnosis, inform malignancy screening, and support escalation of immunotherapy in cases where there is incomplete response.

Special mention must be given to testing for thyroid antibodies, which are found in 11% of healthy controls and as high as 20% of adults over 60 years. Reference Hollowell, Staehling and Flanders83 Accordingly, testing for thyroglobulin antibodies or thyroid peroxidase antibodies is of limited value in the evaluation of AIE, may contribute to AIE misdiagnosis, and should not be routinely performed. Reference Flanagan, Geschwind and Lopez-Chiriboga4,Reference Mattozzi, Sabater and Escudero84,Reference Valencia-Sanchez, Pittock and Mead-Harvey85 Similarly, detection of voltage-gated potassium channel (VGKC) antibodies without LGI1 or CASPR2 antibodies lacks specificity for neurologic autoimmunity, so testing of VGKC antibodies is not recommended. Reference Lang, Makuch and Moloney79,Reference Michael, Waters and Irani80

Imaging

Magnetic resonance imaging

MRI brain with and without gadolinium should be obtained for all patients with suspected AIE. Imaging can reveal evidence of focal or multifocal involvement. Subtypes of AIE can preferentially affect certain anatomical structures, aiding in classifying type of AIE, in addition to directing a more focused differential diagnosis. Reference Kelley, Patel, Marin, Corrigan, Mitsias and Griffith86,Reference Heine, Prüss, Bartsch, Ploner, Paul and Finke87 With respect to particular MRI findings, T2/FLAIR hyperintensities restricted to the medial temporal lobes are classical of autoimmune limbic encephalitis and are central to its diagnostic criteria Reference Graus, Titulaer and Balu28 . Cortical-subcortical T2/FLAIR-hyperintense lesions are characteristic of GABA(A)R antibody encephalitis, and have also been reported with several other neural antibodies including those targeting Hu. Reference Petit-Pedrol, Armangue and Peng88Reference Chang, Nouri, Mirsattari, Burneo and Budhram93 Unilateral cortical T2/FLAIR-hyperintense lesions with hypointensity of the adjacent subcortical white matter have been described in patients with encephalitic presentations of MOG antibody-associated disease. Reference Ogawa, Nakashima and Takahashi73,Reference Budhram, Mirian, Le, Hosseini-Moghaddam, Sharma and Nicolle74,Reference Fujimori, Ogawa, Murata, Takai, Misu and Nakashima94,Reference Valencia-Sanchez, Guo and Krecke95 T2/FLAIR hyperintensities restricted to the claustrum have been reported to be useful markers of seizures related to autoimmune encephalitis. Reference Steriade, Tang-Wai, Krings and Wennberg96 Importantly, however, these findings have varying specificity for AIE and similar neuroimaging abnormalities may be observed in patients with a broad array of alternative diagnoses, underscoring the need to interpret their presence in the clinical context of the patient. Additionally, MRI can be normal in AIE, particularly when imaging is obtained early. Reference Bordonne, Chawki and Doyen97Reference Piao, Bao and Yang100 Imaging changes can develop over time, therefore repeat imaging should be considered when the diagnosis is unclear. Reference Bordonne, Chawki and Doyen97Reference Piao, Bao and Yang100 MRI of the spinal cord should be obtained in cases with symptoms and signs localizing to the spinal cord. MRI can also be utilized for anatomy-specific malignancy screening as appropriate (e.g., pelvic MRI for ovarian teratoma).

Fluorodeoxyglucose (FDG)-Positron Emission Tomography (PET) imaging

There is increasing evidence for the utility of FDG-PET in the diagnosis of AIE. FDG-PET is more sensitive than MRI (87% and 25-50% respectively) and shows changes earlier. Reference Bordonne, Chawki and Doyen97 Medial temporal lobe hypermetabolism has been reported in patients with limbic encephalitis and normal-appearing temporal lobes on MRI, indicating a higher sensitivity of FDG-PET for active inflammation that may be of benefit to the diagnosis and monitoring of patients with this condition. Reference Graus, Titulaer and Balu28,Reference Baumgartner, Rauer, Mader and Meyer101 Various other metabolic patterns have been described, involving regions correlating to clinical findings. For example, NMDAR antibody encephalitis can demonstrate an anteroposterior gradient with frontotemporal hypermetabolism and parieto-occipital hypometabolism; but not in all cases. Reference Bordonne, Chawki and Doyen97 Findings should be interpreted with caution though, as PET findings, especially hypometabolism, are influenced by medications or concurrent neurodegeneration and the specificity of PET findings in the context of suspected AIE is unknown. Reference Bordonne, Chawki and Doyen97,Reference Heine, Prüss, Bartsch, Ploner, Paul and Finke98

FDG-PET should not be used alone for diagnosis, rather to complement other evidence of AIE inflammation and notably is not relied upon in commonly used diagnostic criteria and consensus statements. Reference Abboud, Probasco and Irani5,Reference Graus and Dalmau102 Additionally, PET scans are not widely available in all Canadian provinces and use may be restricted to oncologic indications only.

Whole-body FDG-PET is a sensitive test for occult malignancy, Reference McKeon, Apiwattanakul and Lachance103 with particularly high yield in AIE associated with neural antibodies that are intermediate-risk or high-risk for underlying tumor. Reference Sundermann, Schröder and Warnecke104,Reference Younes-Mhenni, Janier and Cinotti105

Electroencephalography (EEG)

EEG can provide evidence of focality when MRI is normal, occasionally shows findings that may help identifying specific subtypes of AIE and is part of the diagnostic criteria for definite limbic AIE Reference Graus, Titulaer and Balu28,Reference Moise, Karakis and Herlopian106,Reference Wennberg, Steriade, Chen and Andrade107 (Figure 2). Common findings include slowing and epileptiform changes, but these are not specific. Reference Carvajal-González, Leite and Waters70,Reference Elkhider, Sharma, Kapoor, Vattoth and Shihabuddin108 Extreme delta brush is seen in up to 30% of cases of NMDAR antibody encephalitis but is not unique to this disorder. Reference Moise, Karakis and Herlopian106,Reference Nathoo, Anderson and Jirsch109,Reference Baykan, Gungor Tuncer and Vanli-Yavuz110 Hyperventilation induced focal seizures may also be a marker of AIE. Reference Wennberg111

Other ancillary tests

Biopsy is not typically necessary in AIE, and the presence of inflammatory cells alone is not specific. Reference Dalmau, Lancaster, Martinez-Hernandez, Rosenfeld and Balice-Gordon40 The utility of brain biopsy is for confirmation of alternate pathologies. Nerve conduction studies and electromyography can be helpful in cases of CASPR2 antibody encephalitis, Bickerstaff’s encephalitis, and stiff person spectrum disorders. Reference Alberti, Povedano, Montero and Casasnovas112,Reference De Wel and Claeys113

Practical Tips on Investigations:

  1. 1. Due to phenotypic overlap, neural antibody testing for suspected AIE should generally be performed using comprehensive panels instead of sequential single-antibody testing to maximize sensitivity and facilitate prompt diagnosis.

  2. 2. Paired CSF/serum neural antibody testing should be performed, when feasible, to optimize sensitivity and specificity.

  3. 3. Incorporation of TIIF/IHC in neural antibody testing optimizes sensitivity and specificity, and is considered standard of laboratory practice in many regions around the world; neural antibody testing should be performed at laboratories with expertise in TIIF/IHC whenever possible.

  4. 4. When encountering unexpectedly positive or negative neural antibody results, contacting the testing laboratory to review test methodology (e.g., use of comprehensive panel-based testing, confirmatory testing with TIIF/IHC) as well as discussion with a specialist in autoimmune neurology is recommended.

  5. 5. Follow-up MRI of brain should be considered in cases with an initial negative MRI and a strong suspicion for a diagnosis of AIE.

  6. 6. Brain PET is more sensitive than MRI but specificity of findings is unknown; as such PET findings alone should not be used to make a diagnosis of AIE.

  7. 7. Thyroglobulin antibody and Thyroid peroxidase antibody testing are of limited value in the evaluation of patients meeting criteria for autoimmune encephalitis and should not be routinely performed.

First-line treatment

Defining severity of AIE

Defining the clinical severity of autoimmune encephalitis can be helpful in guiding treatment. While clinical scales do exist (e.g., Clinical Assessment Scale in Encephalitis Reference Lim, Lee and Moon114 ) we propose a rapid and practical approach to stratify disease severity based on functional impairment.

  1. i. Severe – All patients requiring non-elective admission (including ICU) or any patient with significant and/or progressive functional impairment defined as inability to perform basic ADLs.

  2. ii. Mild/Moderate – all others (mostly applicable to outpatients).

Recommendations for first-line treatment

The treatment of autoimmune encephalitis (AIE) is predominantly based on case series/retrospective data Reference Nosadini, Eyre and Molteni115Reference Thaler, Zimmermann and Kammermeier117 and expert consensus apart from one positive randomized controlled trial for intravenous immunoglobulin (IVIg) in LGI1/CASPR2 antibody seropositive patients. Reference Dubey, Britton and McKeon118 A clinical presentation consistent with the diagnosis of AIE with exclusion of an infectious cause are the key elements to initiate first-line treatment without delay. Studies suggest that early immunotherapy is associated with better outcome. Reference Balu, McCracken, Lancaster, Graus, Dalmau and Titulaer7Reference Titulaer, McCracken and Gabilondo9 Early tumor removal in paraneoplastic encephalitis is crucial to consider in parallel with first-line therapeutic agents.

For patients who have acute presentations (like are typical of viral encephalitides) or subacute/chronic presentations with clinical or ancillary test findings concerning for infection (e.g., bacterial, fungal or parasitic infections of the CNS), antimicrobial treatment for a possible infectious cause of encephalitis as well as infectious disease consultation as appropriate should be pursued prior to empiric administration of immunotherapy.

We recommend that patients with severe AIE be treated with a combination of high dose steroids (1 g IV methylprednisolone daily for 5 days, or equivalent), AND IVIg (2 g/kg over 2–5 days) or plasma exchange (PLEX) (5 exchanges every other day over 10 days) (Figure 3). Treatment with steroid monotherapy can be considered in mild/moderate cases, such as in patients with anti-LGI1 encephalitis who are assessed in the outpatient setting with seizures in relative isolation, but input from a specialist in autoimmune neurology is recommended. If an associated neoplasm is found, treatment of the neoplasm must be part of the first-line protocol.

Figure 3: Proposed treatment algorithm for AIE. PLEX = plasma exchange; IVIg = intravenous immunoglobulin; GAD = glutamic acid decarboxylase; GFAP = glial fibrillary acidic protein.

Although some studies suggest that PLEX might induce a more rapid effect by removing autoantibodies and other inflammatory substances in the plasma. Reference Zhang, Liu, Jiang, Chen and Su119 there is currently no definite evidence to suggest the superiority of IVIg versus PLEX. Additionally, there are some concerns that antibody mediated diseases associated with pathogenic IgG4 antibodies (e.g., LGI1, CASPR2) may not respond as well to IVIg. Reference Dalakas120 It should be noted, that treatment with IVIg can affect autoantibody testing in serum and may result in a CSF pleocytosis and increased CSF IgG, Reference Shayan, Adamiak, Choe, Relkin and Lee121,Reference Yelehe-Okouma, Czmil-Garon, Pape, Petitpain and Gillet122 and so completion of investigations is necessary prior to initiation of IVIg.

In addition to the recommendation for early and appropriate treatment, we recommend early consideration for the transfer of any patient with moderate to severe AIE to a center with experience in the management of such patients and access to PLEX.

Use of corticosteroids after initial treatment

Current evidence for or against the use of corticosteroid taper is limited, although use of a taper may help prevent early relapse. Reference Abboud, Probasco and Irani5,Reference Alkabie and Budhram123,Reference Nosadini, Eyre and Giacomini124 We recommend that corticosteroid taper be considered in most patients treated with any first-line treatment protocol which includes high dose IV steroids, particularly in patients with severe disease, unless a clear medical contraindication exists (including the risk of exacerbating neuropsychiatric symptoms). This is in keeping with other expert recommendations. Reference Abboud, Probasco and Irani5,Reference López-Chiriboga and Flanagan125

If a steroid taper is used, we recommend 0.5–1 mg/kg/day of oral prednisone, tapering over 4–12 weeks depending on risk of side effects and comorbidities. An alternate (and possibly better tolerated regimen) of pulse steroids (1 g IV methylprednisolone or PO equivalent) weekly initially for 6 weeks followed by every 2 weeks for the next 6 weeks may also be considered.

Longer tapers may be helpful in some diseases (e.g., LGI1 antibody encephalitis). Reference Alkabie and Budhram123 For NMDAR antibody encephalitis, if ongoing corticosteroids are administered in the first months of disease then IV pulses rather than oral taper may be preferable, although guidance for adult cases is limited. Reference Nosadini, Thomas and Eyre126 Input from a specialist in autoimmune neurology may be helpful in guiding taper.

Treatment trials in patients meeting criteria only for “Possible” AIE

Patients meeting only possible AIE criteria after completion of investigations merit special consideration. Importantly, these were not intended to be used as standalone criteria, but rather as the minimum requirements to suspect AIE. Reference Dalmau and Graus127 In patients who only meet criteria for possible AIE but in whom alternative diagnoses have reasonably been excluded, a controlled immunotherapy trial with predefined distinct and objective treatment measures of efficacy can be considered with involvement of a specialist in autoimmune neurology.

Defining treatment failure

There is no widely accepted definition of treatment failure or refractory disease in autoimmune encephalitis. Second-line therapies are felt to be relatively safe and effective in managing AIE. Reference Titulaer, McCracken and Gabilondo9 They are also associated with better outcomes when compared to those who receive additional first-line treatment or no further treatment. Reference Titulaer, McCracken and Gabilondo9 Accordingly, we suggest a low threshold to escalate immunotherapy, particularly in patients with severe AIE.

We define treatment failure in patients with AIE as lack of improvement or worsening 5–10 days after initiation of first-line treatment in severe AIE and 2–4 weeks after initiation of first-line treatment in mild/moderate AIE. The shorter time period in severe AIE is to encourage earlier consideration of second-line therapy (discussed below). Whenever possible, lack of improvement or worsening of objective measures (e.g., seizure frequency, cognitive testing scores) should be used to define treatment failure.

In patients who experience treatment failure, particularly those who are antibody-negative, care should be taken to ensure that the diagnosis of autoimmune encephalitis is correct.

Second-line treatment

Second-line therapy is recommended for anyone who fails first-line treatment as defined above. Choice of second-line therapy is influenced by antibody associations and presence of neoplasm. Involvement of a specialist in autoimmune neurology is recommended in patients undergoing consideration for second-line therapy.

Acute second-line treatment in patients with cell surface antibodies or negative antibodies

Patients with autoimmune encephalitis who have antibodies targeting cell surface antigens (Table 3) should preferentially receive rituximab as second-line therapy due to possible better efficacy and more favorable safety profile Reference Abboud, Probasco and Irani5,Reference Shin, Lee and Park128 (Figure 3). In antibody-negative AIE, rituximab is preferable over cyclophosphamide due to its more favorable safety profile. Reference Abboud, Probasco and Irani5,Reference Shin, Lee and Park128,Reference Ledingham, Plant, Mustafa and Patil129 In general, however, input from a specialist in autoimmune neurology is recommended prior to administering second-line immunotherapies given their prolonged immunosuppressive effect and unique toxicities (e.g., infertility with cyclophosphamide, discussed later on). For ease of administration, rituximab may be administered as two intravenous infusions of 1 g, two weeks apart, rather than four 375 mg/m2 infusions for 4 weeks. Reference Shin, Lee and Park128 Administration of monthly cyclophosphamide infusions of 600–1000 mg/m2 for 3–6 months have commonly been described. Reference Shin, Lee and Park128 Myeloablative cyclophosphamide protocols can be considered in severe disease with input from a specialist in autoimmune neurology with experience in their use.

Table 3: Anti-neural antibodies associated with encephalitis

AMPAR = α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor; ANNA = antineuronal nuclear antibody; CASPR2 = contactin-associated protein-like 2; CRMP5 = collapsin response-mediator protein 5; DNER = Delta/notch-like epidermal growth factor-related receptor; DPPX = dipeptidyl-peptidase-like protein; GABAAR = gamma-aminobutyric acid-a receptor; GABABR = gamma-aminobutyric acid-b receptor; GAD = glutamic acid decarboxylase; GFAP = glial fibrillary acidic protein; GlyR = glycine receptor; LGI1 = leucine-rich glioma-inactivated protein 1; LKLHL11 = Kelch-like protein 11; mGluR1 = metabotropic glutamate receptor type 1; mGluR5 = metabotropic glutamate receptor type 5; NMDAR = N-methyl-D-aspartate receptor; PCA-1 = Purkinje cell cytoplasmic antibody type 1; PCA-2 = Purkinje cell cytoplasmic antibody type 2; References: 63,130.

Consideration should also be given to repeated high dose pulse steroids, concurrent steroid taper, and repeated PLEX/IVIg (if initial PLEX/IVIg was effective) or initiation of PLEX/IVIg (if one of these were not applied during first-line treatment). Reference Abboud, Probasco and Irani5 This should be strongly considered when there is potential for delays in treatment, for example if insurance/administrative approval is needed for use of rituximab.

Acute second-line treatment in patients with intracellular antibodies

In confirmed or probable paraneoplastic cases, treatment and/or tumor resection is strongly recommended in all patients in consultation with oncology and/or neuro-oncology (Figure 3). Paraneoplastic encephalitis with autoantibodies against intracellular targets may be immunotherapy-resistant, in contrast to autoimmune encephalitis with autoantibodies against extracellular targets. Reference Ledingham, Plant, Mustafa and Patil129 However, this is not universal and so trials of immunotherapy (both first line and second line if initial treatment fails) should still be considered in these cases. Reference Vedeler, Antoine and Giometto130 Patients with encephalitis associated with antibodies targeting intracellular antigens, and in particular those with high-risk paraneoplastic antibodies (e.g., anti-Hu, anti-Yo), should preferentially receive cyclophosphamide due to its broader immunosuppressive effect. Reference Binks, Uy, Honnorat and Irani131,Reference Madondo, Quinn and Plebanski132 There is little evidence regarding optimum cyclophosphamide therapy or dosing regimens in this setting.

Despite B-cell depletion being the main mechanism of action for rituximab, there is also indirect suppression of T-cell activity through changes in B- and T-cell signaling pathways. Reference Ramwadhdoebe, van Baarsen and Boumans133 Rituximab can be considered in circumstances where contraindications to cyclophosphamide exist.

For patients with encephalitis associated with antibodies targeting intracellular antigens that have a lower cancer association (e.g., anti-GFAP, anti-GAD65), either cyclophosphamide or rituximab may be considered for second-line immunotherapy.

Additionally, if they have not been given previously, trial of IVIg or PLEX may be considered when other immunotherapies are unavailable or contraindicated, given their relatively low risk and rare reports of benefit attributed to their use even in patients with AIE who harbor antibodies against intracellular targets. Reference Fumal, Jobe and Pepin134Reference Hu, Shang, Liu and Yuan136 It should be acknowledged, however, that in general their efficacy is likely to be lower than in those with AIE in the setting of extracellular antibodies, which are considered to be pathogenic, as opposed to intracellular antibodies, which are not considered to be pathogenic.

Second-line therapy in patients with neoplasm

Second-line therapy can be undertaken in concert with treatment of the underlying neoplasm. Guidance from oncology in cases where surgery is not expected to be curative is essential and delaying second-line therapy may or may not be necessary based on oncology input.

Choice of second-line therapy should be guided by antibody type as above.

Treatment failure after second-line therapy

Defining treatment failure after second-line therapy is challenging due to variations in onset of action of second-line therapies depending on dose/regimen used as well as the co-administration of PLEX/IVIg/repeat IV steroids. As a result, we suggest the determination of treatment failure after second-line therapy be done in consultation with a specialist in autoimmune neurology.

Third-line/alternative immunotherapy

There are a number of third-line and experimental immunotherapies (tocilizumab, bortezomib) which have been employed in cases refractory to second-line immunotherapy. Reference Abboud, Probasco and Irani5,Reference Shin, Lee and Park128,Reference Lee, Lee and Moon137,Reference Turnbull, Siegel, Becker, Stephens, Lopez-Chiriboga and Freeman138 Involvement of a specialist in autoimmune neurology is essential to discuss additional therapeutic options in cases when a patient fails second-line therapy.

Special considerations for refractory NMDAR-IgG AIE

NMDAR antibody encephalitis is associated with underlying teratoma in 11–36% of cases. Reference Titulaer, McCracken and Gabilondo9,Reference Bost, Chanson and Picard139Reference Xu, Lu and Huang141 Microteratomas (occult or imaging negative) are rarely reported Reference Boeck, Logemann and Krauß142,Reference Cirkel, Cirkel and Royl143 and true incidence is unknown.

In cases of severe refractory NMDAR antibody encephalitis, evidence for bilateral “blind” oophorectomy for removal of potential occult ovarian teratoma is limited. Reference Iyengar, Hébert and Climans144 The existing evidence is predominantly based on case series/case reports, many of which were published before options for third-line treatment were available. Given the permanent effects on fertility/iatrogenic menopause and the fact that most patients with refractory NMDAR antibody encephalitis would be unable to provide informed consent due to their underlying disease, such procedures should only be undertaken as a last resort and with informed consent through a surrogate decision maker.

Accordingly, we strongly recommend these procedures be limited to patients with persistently life-threatening disease who fail second (rituximab and/or cyclophosphamide), and third-line therapy (tocilizumab or bortezomib) and who have had multiple imaging modalities to investigate for teratoma including endovaginal ultrasound and MRI pelvis as well as CSF confirmation of NMDAR antibodies. Given the poor level of evidence for “blind” oophorectomy and permanent effects on fertility some authors involved in this guideline strongly believe that this procedure should not be offered.

Teratomas are rare in patients younger than 18 or older than 45 Reference Titulaer, McCracken and Gabilondo9 and blind oophorectomy is not recommended outside of this age range.

Involvement of specialists in autoimmune neurology and gynecology is essential in such cases given permanent impacts to fertility and iatrogenic menopause.

Immune checkpoint inhibitor related autoimmune encephalitis

A detailed discussion of treatment of ICI related AIE is outside the scope of this article and oncology guidelines exist elsewhere. Reference Schneider, Naidoo and Santomasso145 In general, first-line treatment remain similar to other forms of AIE. Rituximab may be used in antibody positive cases although concerns exist around persistent immunosuppression in active malignancy, and oncology input is essential. Reference Schneider, Naidoo and Santomasso145,Reference Haanen, Carbonnel and Robert146

Practical Tips on Treatment:

  1. 1. Early initiation of appropriate treatment is essential to optimize outcomes.

  2. 2. Awaiting neural antibody results should not delay consideration of empiric immunotherapy if there is a high index of suspicion for AIE, after reasonable exclusion of alternative diagnoses.

  3. 3. All patients with severe AIE should receive high dose corticosteroids with IVIg or PLEX as initial therapy; treatment with steroid monotherapy can be considered in mild/moderate cases but input from a specialist in autoimmune neurology is recommended (Figure 3).

  4. 4. Treatment of underlying neoplasm (if found) should occur in concert with first-line treatment when possible.

  5. 5. Second-line therapy should be offered to all patients with severe AIE who fail to improve or worsen 5-10 days after initiation of first-line therapy (2-4 weeks for mild/moderate cases).

  6. 6. Proposed differences in the approach to treatment of patients with severe AIE compared to those with mild/moderate disease include the routine initiation of dual first-line immunotherapies and the more rapid determination of treatment failure, to encourage earlier escalation to second-line immunotherapy in those with severe disease.

  7. 7. Early involvement of a specialist in autoimmune neurology is strongly recommended for all patients who fail first-line treatment.

Screening for neoplasm

While initially described as consequences of paraneoplastic syndromes, AIE has been increasingly recognized as a non-paraneoplastic phenomenon. Reference Abboud, Probasco and Irani5 Still, all subtypes of AIE may be associated with an underlying neoplasm at varying frequencies. Reference Abboud, Probasco and Irani5,Reference Titulaer, Soffietti and Dalmau147 A paraneoplastic AIE cannot be ruled out clinically; hence, neoplasm screening is recommended for all initial adult AIE presentations and should also be considered at the time of AIE relapse. Reference Abboud, Probasco and Irani5,Reference Abboud, Probasco and Irani148Reference Zuliani, Nosadini and Gastaldi150

Initial neoplasm screening is imaging based and can be formulated as a three-step process, terminated early if neoplasm is identified or if three steps are exhausted without a neoplasm identified. Reference Abboud, Probasco and Irani148 Conventional CT imaging of the body, focused sex-specific imaging (ectopic germ cell tumors would be covered by the prior step), and finally a whole-body PET scan (in patients with intermediate or high-risk antibodies or some cases of antibody-negative AIE – see below) comprise the three imaging modalities recommended for occult cancer screening in cases of AIE. First-line use of whole-body PET CT can be considered in cases where there is a strong antibody-neoplasm association. This may not be feasible in all areas of Canada and we do not encourage delaying neoplasm screening if there is a prolonged wait for antibody testing results. Of note, sensitivity of PET for ovarian teratoma is felt to be low, and pelvic US or MRI is preferred. Reference Saleh, Bhosale and Menias151

Optimal screening for antibody-negative AIE is unknown. Some authors propose considering definite LE as a higher risk phenotype meriting PET scan and regular follow-up screening similar to intermediate and/or high-risk antibodies. Reference Abboud, Probasco and Irani148 This can also be considered in patients with antibody-negative AIE and poor response to immunotherapy or other significant risk factors for malignancy.

Imaging is recommended even in patients with a known neoplasm that is not strongly associated with AIE, as it may identify a second, more likely relevant neoplasm. Reference Lucchinetti, Kimmel and Lennon152 In cases with a strong paraneoplastic association, directed testing can be started immediately (e.g., immediate ovarian ultrasound for NMDAR antibody encephalitis in a young woman to rule out a teratoma, testicular ultrasound in a man with KLHL11 antibody encephalitis to rule out testicular neoplasm). Screening for classic tumor markers (e.g., carcinoembryonic antigen (CEA)) is of little benefit in identifying neoplasm in patients with AIE. Reference Trevisiol, Cani, Fabricio, Gion, Giometto and De Massis153 A neoplasm screening flowchart is shown in Figure 4 Reference Abboud, Probasco and Irani5 .

Figure 4: Neoplasm screening protocol. AIE: autoimmune encephalitis, CT: computed tomography, MRI: magnetic resonance imaging, PET: positron emission tomography. *Can be considered in a relapse, especially in patients with intermediate or high-risk antibodies. **For benign neoplasms oncology involvement may not be necessary as treatment is typically surgical.

If a neoplasm is identified, anti-neoplastic therapy and follow-up monitoring are dependent on neoplasm type and should be directed by an oncologist. In cases of benign neoplasm, oncologist involvement may not be needed as surgical removal may be the only treatment required. Paraneoplastic AIE is most commonly associated with neoplasms of the lung (especially SCLC), breast, thymus, ovaries or testicles, as well as neuroblastoma and Hodgkin’s lymphoma. Reference Abboud, Probasco and Irani5 Paraneoplastic antibodies do not correlate with tumor bulk, and so do not reflect neoplasm treatment response, which should be dictated by neoplasm specific guidelines. Reference Trevisiol, Cani, Fabricio, Gion, Giometto and De Massis153

If no neoplasm is found, the need for follow-up oncologic screening is most dependent on the presence of a high or intermediate-risk antibodies, and to a lesser extent high-risk phenotype (LE, encephalomyelitis with peripheral involvement), age, incomplete treatment response, smoking status, and other cancer risk factors. Reference Zekeridou, Kryzer and Guo48,Reference Titulaer, Soffietti and Dalmau147,Reference Rosenfeld and Dalmau154 A list of high, intermediate, and low-risk antibodies associated with AIE is in Table 4. Reference Graus, Vogrig and Muñiz-Castrillo63,Reference Marsili, Marcucci, LaPorta, Chirra, Espay and Colosimo155

Table 4: Antibody type and malignancy risk

*CASPR2 is only considered intermediate risk when in the context of Morvan syndrome.

AMPAR = α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor; ANNA = antineuronal nuclear antibody; CASPR2 = contactin-associated protein-like 2; CRMP5 = collapsin response-mediator protein 5; DNER = Delta/notch-like epidermal growth factor-related receptor; DPPX = dipeptidyl-peptidase-like protein; GABAAR = gamma-aminobutyric acid-a receptor; GABABR = gamma-aminobutyric acid-b receptor; GAD = glutamic acid decarboxylase; GFAP = glial fibrillary acidic protein; GlyR = glycine receptor; LGI1 = leucine-rich glioma-inactivated protein 1; LKLHL11 = Kelch-like protein 11; MAP1B = microtubule-associated protein 1B; mGluR1 = metabotropic glutamate receptor type 1; mGluR5 = metabotropic glutamate receptor type 5; MOG = myelin oligodendrocyte glycoprotein; NMDAR = N-methyl-D-aspartate receptor; PCA-1 = Purkinje cell cytoplasmic antibody type 1; References: 63,155.

While paraneoplastic neurological disorders may precede a neoplasm diagnosis, a tumor is found within 1 year of presentation in >90% of cases with solid tumors. Reference Rosenfeld and Dalmau154 In cases associated with intermediate and high-risk antibodies, repeat screening every 3–6 months for at least 2 years is recommended. Reference Abboud, Probasco and Irani5,Reference Titulaer, Soffietti and Dalmau147-Reference Pollak, Lennox and Müller149,Reference Rosenfeld and Dalmau154,Reference Rössling and Prüss156,Reference Toledano and Pittock157 While there is no firm consensus on the exact frequency and duration of neoplasm surveillance, one should consider frequent and prolonged screening for cases with multiple risk factors. Patients with antibody-negative disease and high-risk phenotypes such as LE, poor response to immunotherapy, relapsing disease and/or significant risk factors for malignancy should also be considered for follow-up neoplasm screening as defined in these guidelines.

Practical Tips on Neoplasm Screening

  1. 1. All adult patients presenting with AIE should undergo screening for malignancy at the time of diagnosis.

  2. 2. Initial screening should not be delayed while awaiting neural antibody test results.

  3. 3. Sex-specific testing should be undertaken if the initial screen is negative (Figure 4).

  4. 4. Follow-up screening is not necessary in patients with a low-risk antibody and initial negative screening but should be done in patients with intermediate or high-risk antibodies.

  5. 5. For antibody-negative patients, the optimal approach to follow-up screening is unknown but repeat screening should be considered in certain patients (e.g., those with high-risk phenotypes such as LE, refractory/relapsing disease, and/or significant risk factors for malignancy)

Treatment of other disease manifestations

Seizure management

The International League Against Epilepsy nomenclature of “acute symptomatic seizures secondary to autoimmune encephalitis” (ASSAE) and “autoimmune-associated epilepsy” are helpful conceptually when classifying patients AIE with seizures, Reference Steriade, Britton and Dale158 although the modified term “autoimmune encephalitis-associated epilepsy” (AEAE) serves to make explicit the link between AIE and seizures in all cases. Reference Budhram and Burneo159 The proportion of patients with AIE who develop seizures has been estimated at 50–70%. Reference Budhram and Burneo159 AIE associated with autoantibodies targeting extracellular antigens – especially GABAAR, Reference Petit-Pedrol, Armangue and Peng59 GABABR, Reference van Coevorden-Hameete, de Bruijn and de Graaff161 NMDAR, Reference Dalmau, Gleichman and Hughes162 and LGI1 Reference Gadoth, Pittock and Dubey46 – are a more commonly encountered cause of seizures than AIE associated with those targeting intracellular antigens. GAD65 antibody is a notable exception. Reference Budhram and Burneo159,Reference Budhram, Sechi and Flanagan77,Reference Steriade, Gillinder and Rickett163 There is a paucity of data on the overall incidence and risk factors associated with the development of AEAE. Relatively small retrospective cohort studies of patients treated for seizures related to AIE reported that, at a two-year follow-up, 2-42% of patients developed AEAE. Reference Casciato, Morano and Fattouch164Reference Steriade, Moosa, Hantus, Prayson, Alexopoulos and Rae-Grant166

We recommend a combined initial treatment approach with anti-seizure medications (ASMs) and immunosuppression for the treatment of seizures related to AIE. Some patients (in particular those with antibodies against extracellular targets) may respond to immunosuppression alone, indicating ASSAE. Reference Dubey, Britton and McKeon118,Reference Abboud, Probasco and Irani148,Reference de Bruijn, van Sonderen and van Coevorden-Hameete165,Reference Feyissa, Lamb and Pittock167 Patients who receive immunosuppression, and who receive it quickly, have a higher chance of achieving seizure freedom. Reference Dubey, Britton and McKeon118,Reference de Bruijn, van Sonderen and van Coevorden-Hameete165,Reference Feyissa, Lamb and Pittock167,Reference Bozzetti, Rossini and Ferrari168 Sodium-channel blockers are possibly more effective for the treatment of seizures related to AIE, especially in patients with LGI1 antibody encephalitis, but are associated with higher risks of side effects. Reference Gong, Chen and Liu140,Reference Feyissa, Lamb and Pittock167,Reference Cabezudo-García, Mena-Vázquez, Villagrán-García and Serrano-Castro169 Enzyme-inducing ASMs should be avoided in patients treated with chemotherapy due to risk of lowering drug levels. Reference Gonzalez Castro and Milligan170 In the patient who is no longer in the acute phase of AIE and has become seizure-free a gradual wean from ASMs can be considered after discussing the importance of vigilance for signs or symptoms of seizure recurrence that would merit prompt clinical re-assessment. Reference Abboud, Probasco and Irani148,Reference de Bruijn, van Sonderen and van Coevorden-Hameete165

Neuropsychiatric symptoms

Neuropsychiatric symptoms are common in AIE and are expected to improve with first- and second-line treatments. Reference Hébert, Day, Steriade, Wennberg and Tang-Wai8,Reference Loughan, Allen, Perna and Malkin171 To our knowledge, there are no randomized controlled trials investigating treatment of neuropsychiatric symptoms in patients with AIE; management therefore relies on observational studies and expert opinion, usually with conventional psychiatric treatments. Reference Oldham172 Psychiatric symptoms in patients with AIE improve less frequently with symptomatic treatments compared to patients with psychiatric conditions, and patients are more likely to experience side effects. Reference Marinas, Matveychuk, McCombe, Tymchuk, Dursun and Baker173Reference Zhang, Cai and Zheng175

Regarding acute agitation and psychosis, special caution is required with antipsychotic medications. Reference Marinas, Matveychuk, McCombe, Tymchuk, Dursun and Baker173,Reference Dale, Gorman and Lim176 Patients face a higher risk of drug induced movement disorders, worsening agitation, rhabdomyolysis, and neuroleptic malignant syndrome, especially with medications with high affinity for the D2 dopamine receptor, such as haloperidol. Reference Liu, Tian and Guo177 If required, atypical antipsychotics are preferred. Reference Oldham172,Reference Coffey and Cooper178 Options are detailed in Table 5 including strategies to target sleep disturbance.

Table 5: Recommended agents for neuropsychiatric symptoms

Electroconvulsive therapy has been used for severe psychosis and catatonia in AIE Reference Loughan, Allen, Perna and Malkin171Reference Marinas, Matveychuk, McCombe, Tymchuk, Dursun and Baker173,Reference Dhossche and Wachtel179 with some success, especially when the catatonia is treatment resistant, and immunotherapy has failed. Reference Tanguturi, Cundiff and Fuchs180 Catatonia can be treated with benzodiazepines, with caution in patients with hypoventilation and decreased arousal. Reference Coffey and Cooper178

Even with treatment, recovery can take many years and is often incomplete with cognitive, and neuropsychiatric symptoms persisting in 50% of patients. Reference Zhang, Cai and Zheng175,Reference Huang, Wang, Feng, Wang and Li181Reference Seery, Butzkueven, O’Brien and Monif183 Behavioral agitation, emotional lability, posttraumatic stress disorder symptoms, anxiety, and depression are common. Selective serotonin reuptake inhibitors, pindolol and valproic acid have been used for agitation and disinhibition Reference Marinas, Matveychuk, McCombe, Tymchuk, Dursun and Baker173 in those with chronic symptoms.

Persistent cognitive symptoms include deficits in attention, memory, executive function, and processing speed, and are associated with poor psychosocial outcomes. Reference Marinas, Matveychuk, McCombe, Tymchuk, Dursun and Baker173 Supervision, rehabilitation, and family-centered care are important. Reference Loughan, Allen, Perna and Malkin171,Reference Scheer and John184 Non-pharmacological management borrowed from the TBI literature includes reducing overstimulation, clustering care, minimizing physical restraint as well as assessing the environment and sleep wake cycles. Reference Vova185

Rehabilitation/prognosis for recovery

Prognostication in AIE is challenging due to lack of long-term evidence, and variability in outcomes based on specific AIE subtype and associated cancer. Reference Deng, Qiu, Liu, Wu, Lei and Lu186,Reference Broadley, Seneviratne and Beech187 For example, 75–85% of patients with NMDAR antibody encephalitis achieve good functional outcomes (modified Rankin Score of 0-2) Reference Gong, Chen and Liu140,Reference Dalmau, Gleichman and Hughes162 compared with only 43% of patients with GAD65 antibody encephalitis. Reference Budhram, Sechi and Flanagan77 Approximately half of patients with AIE may continue to experience cognitive impairment after the acute onset of disease. Reference Hébert, Day, Steriade, Wennberg and Tang-Wai8 Earlier initiation of treatment has been associated with improved long-term functional, Reference Deng, Qiu, Liu, Wu, Lei and Lu186,Reference Broadley, Seneviratne and Beech187 cognitive Reference Hébert, Day, Steriade, Wennberg and Tang-Wai8 and psychiatric Reference Nguyen, Yang, Goyal, Irani and Graves188 outcomes across a variety of AIE subtypes. Once patients are no longer in the acute phase of AIE, they should be routinely screened for residual neuropsychiatric impairment and be offered a combination of psychiatric care Reference Blum, Tomlinson, Jetté, Kwon, Easton and Yeshokumar189 and neuropsychological rehabilitation. Reference Abboud, Probasco and Irani148,Reference Blum, Tomlinson, Jetté, Kwon, Easton and Yeshokumar189

Practical Tips on Secondary Management

  1. 1. Patients who receive prompt immunosuppression are more likely to obtain seizure freedom.

  2. 2. A combined initial approach of immunosuppression and use of anti-seizure medications is strongly recommended for most patients with AIE and seizures.

  3. 3. Neuropsychiatric symptoms are common in AIE and are expected to improve with first- and second-line treatments but symptomatic therapy may still be required (Table 5).

Chronic management

Relapse in autoimmune encephalitis

Relapse in AIE is not uncommon, and characteristics demonstrate significant variability between AIE subtypes. Reference Titulaer, McCracken and Gabilondo9,Reference van Sonderen, Ariño and Petit-Pedrol21,Reference van Sonderen, Thijs and Coenders22,Reference Ropper, Dalmau and Graus42,Reference Shin, Lee and Park128,Reference Smith, Dubey, Liebo, Flanagan and Britton190 Retrospective observational studies of NMDAR, LGI1, and CASPR2 antibody encephalitis indicate relapse rates ranging from 10 to 41% and symptoms may be identical to the index episode (e.g., LGI1 antibody encephalitis re-presenting with seizures), similar but milder (e.g., NMDAR antibody encephalitis presenting with behavioral symptoms after previous severe ICU admission) or a different distinct core clinical syndrome (e.g., CASPR2 antibody encephalitis presenting with cerebellar ataxia after previous seizure presentation). Reliable data is however hard to obtain as rates and characteristics are confounded by small cohort sizes with limited follow-up duration and a lack of consensus on the definition of relapse in autoimmune encephalitis. Resultantly, relapse attributes are not fully known for all AIE subtypes (e.g., the relapse rate for antibody-negative AIE is unknown).

For the purposes of these guidelines, we define relapse in AIE as a clear objective worsening of symptoms after initial improvement or plateau, preferably supported by evidence of inflammation on MRI or in CSF, and usually occurring at a minimum of 2–3 months from original presentation. PET scan findings are often non-specific Reference Graus and Dalmau102 and isolated PET abnormalities should be interpreted with caution if they are the only objective marker of relapse. Antibody titers are typically not helpful in defining relapse due to their imperfect correlation with disease activity. Reference Gresa-Arribas, Titulaer and Torrents47,Reference Di Giacomo, Deleo and Pastori191

For acute treatment of relapse, we suggest following the first-line and/or second-line treatment algorithm as appropriate unless a patient is known to have failed a particular therapy in the past. Long-term immunosuppression may be appropriate and consultation with a specialist in autoimmune neurology is recommended in cases of relapse both to confirm relapse and guide long-term treatment.

Long-term immunosuppression

Data to guide long-term immunosuppression decisions in AIE are lacking. Given uncertainty and variability of relapse rates, long-term therapy is not routinely recommended for all patients at first presentation of AIE, although we acknowledge substantial variability in practice even among experts. One to two years of immunotherapy may be justified for patients with more severe disease. In relapsing forms of AIE, three or more years of maintenance immunotherapy has been proposed. Reference Abboud, Probasco and Irani148 We recommend involvement of a specialist in autoimmune neurology when considering initiation of long-term immunosuppression.

Practical Tips on Chronic Management:

  1. 1. Risk of relapse in AIE differs depending on specific antibody positivity and ranges from 10-41%.

  2. 2. Long-term immunosuppressive therapy is not routinely recommended for all patients at first presentation of AIE, although we acknowledge there is substantial variability in practice even among experts.

  3. 3. Relapses should be treated according to the 1st/2nd line treatment algorithms as above unless a particular therapy is known to be ineffective for the patient.

Mitigation of risks of immunosuppression

Latent infection screening

Although risks of recrudescent infection vary by pathogen and immunotherapy, early broad testing to expedite appropriate prophylactic or definitive treatment is advised. Screening should routinely include testing for VZV, HIV, Hepatitis B/C, and tuberculosis. When abnormalities are found, immunotherapy can often be started soon after implementing appropriate monitoring/treatment (Table 6). When active or latent infections are identified, consultation with an infectious disease specialist is advised.

Table 6: Overview of screening response by pathogen

†Rituximab and cyclophosphamide can be used to treat the immunological complications of Hepatitis C but a risk of severe hepatitis exists beyond risk posed by hepatitis C.

‡VZV testing is to ascertain if sufficient immunity remains to reduce the risk of emergent singles.

Ab: Antibody, Ag: Antigen, cART: combined antiretroviral therapy, CD: Cell differentiation factor, CNS: Central Nervous System, CSF: Cerebrospinal Fluid, DNA: Deoxyribonucleic Acid, esp: especially, HB: Hepatitis B, HC: Hepatitis C, HIV: Human Immunodeficiency Virus, HBs Ag: Hepatitis B surface antigen, HBc Ab: Hepatitis B core antibody, ID: Infectious disease.

Vaccination

Age appropriate and risk context appropriate vaccinations (e.g., meningococcal vaccination in military recruits) should be considered for all patients embarking on immunosuppression (Table 7). Reference Mejia and Nutman200,203206 Although delaying immunotherapy 4–6 weeks after a vaccine series is completed is typically recommended, this is not practical in acute autoimmune encephalitis presentations. Often vaccination may be delayed pending treatment conclusion (Table 8) 207,Reference Rubin, Levin and Ljungman208 or, if therapy is prolonged, be taken during treatment with non-live vaccines, recognizing efficacy of vaccines may be reduced.

Table 7: Vaccination recommendations – adapted from CDC 2022 and Canadian guidelines (Rubin et al. 2014; Government of Canada 2021; CDC 2022)

HPV: Human Papilloma Virus; InV: Inactivated vaccine; LV: Live vaccine; MMR: Measles, Mumps, Rubella; PCV15: 15-valent pneumococcal conjugate vaccine; PCV20: 20-valent pneumococcal conjugate vaccine; Pneumococcal polysaccharide vaccine (PPSV23); RIV4: recombinant influenza vaccine; LAIV4: live attenuated influenza vaccine; RV: Recombinant Vaccine; RZV: Recombinant Varicella Vaccine; SRPCV: Subunit, recombinant, polysaccharide, and conjugate vaccines; TD: Tetanus, Diphtheria; Tdap: Tetanus, Diphtheria, Acellular Pertussis, Tox: Toxoid; IIV4: inactivated influenza vaccine.

Table 8: Recommendations for treatment related vaccine delays

Reproductive health/pregnancy risks

First-line immunotherapies are generally safe in pregnancy. 211 Steroids can be administered, including high dose IV pulse therapy. Reference Bandoli, Palmsten, Forbess Smith and Chambers212Reference Hviid and Mølgaard-Nielsen214 When used, decision makers should be advised of mild increased infection risk including those more relevant to pregnancy (e.g., endometritis). Treatment with IVIg and PLEX are considered safe in pregnancy although additional hypercoagulability should be discussed. Reference Achiron, Kishner and Dolev215,Reference Watson, Katz and Bowes216

Some second-line agents can also be used safely. Guidelines for rituximab vary with regards to advisable delay of 6 to 12 months between treatment and conception/delivery. Reference Chakravarty, Murray, Kelman and Farmer217 Reports have demonstrated safety when treatment occurs within 6 months of delivery even if an ability to cross the placenta routinely leads to transient CD19/20 depletion in the infant. Reference Das, Damotte and Gelfand218,Reference Smith, Hellwig, Fink, Lyell, Piehl and Langer-Gould219 Cyclophosphamide should never be used in pregnancy and its use in men or women requires discussion of infertility risk, and potential gamete harvesting/storage (acuity permitting) should be explored. Involvement of a specialist in autoimmune neurology in such cases is strongly recommended.

Pneumocystis jirovecii (PJP) prophylaxis

PJP prophylaxis is crucial but precise application is debated for those on immunotherapy. Guidelines exist for those with rheumatologic/hematologic conditions but relevance in neurological illness is unknown.

Trimethoprim-sulfamethoxazole (TMP-SMX) is the gold standard for PJP prophylaxis. If a prior minor reaction to TMP-SMX is reported (i.e., no respiratory or mucosal membrane involvement or fever) then desensitization and subsequent use is preferable to therapy with atovaquone, dapsone, or pentamidine. Reference Cooley, Dendle and Wolf220,Reference Urbancic, Ierino, Phillips, Mount, Mahony and Trubiano221

Overall, we recommend providing PJP prophylaxis in the following scenarios Reference Cooley, Dendle and Wolf220,Reference Park, Curtis and Jun222 .

  1. 1. All patients on high dose corticosteroids (20 mg of prednisone for 2 weeks or more, alone or in combination with other medications).

  2. 2. Overlapping rituximab/cyclophosphamide therapy until the resolution of either CD19/20 cell counts (if rituximab is discontinued) or lymphopenia/neutropenia (if cyclophosphamide is discontinued).

  3. 3. Patients on 5 mg prednisone equivalent or more in combination with cyclophosphamide.

Practical Tips on Mitigation of Risks of Immunosuppression:

  1. 1. All patients with AIE should be routinely screened for VZV, HIV, Hepatitis B/C, and tuberculosis, ideally prior to initiation of immunosuppression.

  2. 2. IVIg can result in false-positive Hepatitis B core antibody results leading to unnecessary use of anti-viral medications.

  3. 3. First-line therapies (IVIg, PLEX, corticosteroids) are generally considered safe in pregnancy.

  4. 4. All patients receiving 20 mg prednisone equivalent or more for more than 2 weeks should receive PJP prophylaxis.

Complications of corticosteroid use

Corticosteroids infrequently cause lasting side effects during brief courses (e.g., gastric ulcer/bleeding, avascular necrosis), but the consequences of long-term use can undermine therapeutic efforts. Common issues include worsening metabolic syndrome (i.e., increased body mass index, hypertension, hyperlipidemia, glucose intolerance), osteoporosis, infections, and impaired wound healing/skin thinning. Reference Miloslavsky, Naden and Bijlsma223Reference Kersey and Broadway225 Neuropsychiatric manifestations are perhaps the most problematic as they can overlap with encephalitic symptomatology. If side effects are extreme, more rapid tapering/discontinuation may be required however several mitigation strategies exist (Table 9). Reference Liu, Ahmet and Ward226,Reference Liu, Reidy, Saatee and Collard227

Table 9: Steroid side effect mitigation Reference Liu, Ahmet and Ward226

BMD: Bone mineral density; IOP: Intraocular pressure; FRAX: Fracture risk assessment tool; MRI: Magnetic resonance imaging.

Considerations for clinical and multidisciplinary care

Given the complexities in both the acute and chronic management of AIE, early referral to a center with expertise in managing AIE and access to PLEX is recommended, especially in patients who do not respond to first-line therapy. A link to an updated list of centers and clinicians with interest and expertise in AIE is provided in the appendix. Early involvement of psychiatry in patients with prominent and/or persistent neuropsychiatric symptoms is also warranted. Rheumatology, oncology and infectious disease consultations should be considered in patients with features of systemic autoimmune disease, known malignancy or immunocompromised state and/or features suggestive of CNS infection respectively.

Conclusion

Autoimmune encephalitis has been increasingly recognized as a cause of subacute neurological symptoms and mental status changes. Nonetheless, high quality evidence for diagnosis and management remains lacking. In this context, these consensus guidelines may be helpful in aiding front line clinicians to recognize, appropriately investigate, and treat patients with AIE with the aim of standardizing care. Gaps in care exist and will continue to exist in Canada; not only around access to first- and second-line treatments (notably PLEX and rituximab) but also in access to clinicians and centers with expertise in diagnosing and managing AIE. Additionally, the authors hope that establishment of guidelines may be helpful when advocating for equitable access to treatment at the provincial and national level.

Increased recognition has fortunately also resulted in the recent development of clinical trials and therapeutics specific to AIE. These developments provide hope for new evidence-based treatments for AIE and ongoing improvement in outcomes for patients.

Supplementary material

The supplementary material for this article can be found at https://doi.org/10.1017/cjn.2024.16.

Acknowledgements

We would like to thank Dr Alexandra Olmos-Perez for her valuable input in the ethical considerations around blind oophorectomy as well as Brooke Hahn for her help with the graphic design of the figures and tables.

We would also like to thank Gary Sarohia and Nesrin Shaheen for their input and review from the patient/family member perspective along with the support of the anti-NMDA Receptor Encephalitis Foundation.

Author contribution

CH was responsible for the overall design of the guidelines and administration of the RAND process.

CH and JM provided overall editorial review and editing of the draft manuscript.

AB additionally provided overall editorial review.

The primary panel members included NA, AB, JB, JC, RC, JH, AM, BM, IP, KW and CU.

Secondary panel members were KA, GB, GB, JB, JC, PD, RK, SL, AM, WM and PS.

RAND Process voting participants were KA, NA, AB, GB, JB, RC, JH, RK, JM, IP, PS, KW and CU.

Funding statement

This manuscript was unfunded.

Competing interests

AB reports that he holds the London Health Sciences Centre and London Health Sciences Foundation Chair in Neural Antibody Testing for Neuro-Inflammatory Diseases, and receives support from the Opportunities Fund of the Academic Health Sciences Centre Alternative Funding Plan of the Academic Medical Organization of Southwestern Ontario.

AM reports honoraria from Alexion, Biogen, EMD Serono, and Novartis.

CH reports grant funding from the Alberta Neurosciences, Rehabilitation and Vision Strategic Clinical Network as well as honoraria from Alnylam, Akcea and Pfizer.

CU reports a paid committee position on the British Columbia Provincial Blood Coordinating Board “IVIG for Central Nervous System Indications Task Force” and speaker’s fee from the Northwest Rheumatism Society.

IP reports honoraria from Biogen, Novartis, Roche and EMD Serono.

JB reports remuneration from Alexion, Atara Phramaceutics, Bayer Healthcare, Beigene, BMS (Celgene), EMD Inc., Hoffmann-La Roche, Jansen (J&J), Merck Serono, Novartis, Sanofi-Genzyme, and Teva Canada innovation for work as a clinical trial EDSS rater.

JC reports educational honoraria from Takeda

JH received salary support during his work on this manuscript through a grant from the American Epilepsy Society.

JM reports grant funding from the University of Alberta Department of Medicine, consulting fees from Novartis, educational honoraria from the American Academy of Neurology and participation in an advisory board for Horizon

KM reports educational funding from the University of Calgary, membership on the executive of the Federation of Medical Women of Canada, membership on the Nominations Committee of the Royal College of Physicians and Surgeons of Canada and is the co-founder of Kolabo.

NA reports educational honoraria from Biogen

PS received speaking honoraria and honoraria from serving on the scientific advisory boards of: Biogen-Idec Pharmaceuticals; Roche Pharmaceuticals; EMD Serono Canada Pharmaceuticals; Novartis Pharmaceuticals, and honoraria for serving as an expert reviewer for the Short Term Exceptional Drug Therapy program in Alberta, Canada. She is a co-investigator in receiving an unrestricted research grant for a prospective pregnancy registry cohort project in Canada from Biogen-Idec Canada.

RC reports grants for Roche/Genentech, Roche and Teva Innovation, consulting fees from Roche, Serono, Sanofi, Biogen, Novartis, Alexion and Teva and travel support from Serono.

RK reports honoraria from Horizon Therapeutics

SL reports consulting fees from Bayers, educational honoraria from UdeM Neuro-Oncology Course, Medlink, Le Medecin du Quebec and Neurodiem, payment for expert testimony from MD Analytics, as well as participation in scientific advisory board meetings for Alexion and Novocure

WM Reports consulting fees from Servier, Novocure, AnHHeart and Boeringher Inggelheim as well as participation as the chair of the data monitoring committee for Ono Therapeutics.

The remaining authors have no competing interests to declare.

References

Dubey, D, Pittock, SJ, Kelly, CR, et al. Autoimmune encephalitis epidemiology and a comparison to infectious encephalitis. Ann Neurol. 2018;83:166–77. DOI: 10.1002/ana.25131.Google Scholar
Hébert, J, Riche, B, Vogrig, A, et al. Epidemiology of paraneoplastic neurologic syndromes and autoimmune encephalitides in France. Neurol Neuroimmunol Neuroinflamm. 2020;7:e883. DOI: 10.1212/NXI.0000000000000883.Google Scholar
Gable, MS, Sheriff, H, Dalmau, J, Tilley, DH, Glaser, CA. The frequency of autoimmune N-methyl-D-aspartate receptor encephalitis surpasses that of individual viral etiologies in young individuals enrolled in the California Encephalitis Project. Clin Infect Dis. 2012;54:899904. DOI: 10.1093/cid/cir1038.CrossRefGoogle ScholarPubMed
Flanagan, EP, Geschwind, MD, Lopez-Chiriboga, AS, et al. Autoimmune encephalitis misdiagnosis in adults. JAMA Neurol. 2023;80:30–9. DOI: 10.1001/jamaneurol.2022.4251.CrossRefGoogle ScholarPubMed
Abboud, H, Probasco, JC, Irani, S, et al. Autoimmune encephalitis: proposed best practice recommendations for diagnosis and acute management. J Neurol Neurosurg Psychiatry. 2021;92:757–68. DOI: 10.1136/jnnp-2020-325300.Google Scholar
Cohen, J, Sotoca, J, Gandhi, S, et al. Autoimmune encephalitis: a costly condition. Neurology. 2019;92:e964e72. DOI: 10.1212/WNL.0000000000006990.CrossRefGoogle ScholarPubMed
Balu, R, McCracken, L, Lancaster, E, Graus, F, Dalmau, J, Titulaer, MJ. A score that predicts 1-year functional status in patients with anti-NMDA receptor encephalitis. Neurology. 2019;92:e244e52. DOI: 10.1212/WNL.0000000000006783.CrossRefGoogle ScholarPubMed
Hébert, J, Day, GS, Steriade, C, Wennberg, RA, Tang-Wai, DF. Long-term cognitive outcomes in patients with autoimmune encephalitis. Can J Neurol Sci. 2018;45:540–4. DOI: 10.1017/cjn.2018.33.CrossRefGoogle ScholarPubMed
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;12:157–65. DOI: 10.1016/S1474-4422(12)70310-1.Google Scholar
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 Sep;133:2734–48. DOI: 10.1093/brain/awq213.Google Scholar
Muñiz-Castrillo, S, Haesebaert, J, Thomas, L, et al. Clinical and prognostic value of immunogenetic characteristics in anti-LGI1 encephalitis. Neurol Neuroimmunol Neuroinflamm. 2021;8: e974. DOI: 10.1212/NXI.0000000000000974.CrossRefGoogle ScholarPubMed
Mittal, MK, Rabinstein, AA, Hocker, SE, Pittock, SJ, Wijdicks, EF, McKeon, A. Autoimmune encephalitis in the ICU: analysis of phenotypes, serologic findings, and outcomes. Neurocrit Care. 2016;24:240–50. DOI: 10.1007/s12028-015-0196-8.Google Scholar
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;17:760–72. DOI: 10.1016/S1474-4422(18)30244-8.Google Scholar
Hou, R, Wu, J, He, D, Yan, Y, Li, L. Anti-N-methyl-D-aspartate receptor encephalitis associated with reactivated Epstein-Barr virus infection in pediatric patients: three case reports. Medicine (Baltimore). 2019;98:e15726. DOI: 10.1097/MD.0000000000015726.Google Scholar
Prakash, PA, Jin, J, Matharu, K, et al. Anti-NMDAR encephalitis with concomitant varicella zoster virus detection and nonteratomatous malignancy. Neurol Neuroimmunol Neuroinflamm. 2019;6:e537. DOI: 10.1212/NXI.0000000000000537.CrossRefGoogle ScholarPubMed
Prüss, H, Dalmau, J, Harms, L, et al. Retrospective analysis of NMDA receptor antibodies in encephalitis of unknown origin. Neurology. 2010;75:1735–9. DOI: 10.1212/WNL.0b013e3181fc2a06.Google Scholar
Predkele, N, Mednieks, J. Lyme disease and associated NMDAR encephalitis: a case report and literature review. Neurol Int. 2021;13:487–96. DOI: 10.3390/neurolint13040048.Google Scholar
Marini, A, Bernardini, A, Gigli, GL, et al. Neurologic adverse events of immune checkpoint inhibitors: a systematic review. Neurology. 2021;96:754–66. DOI: 10.1212/WNL.0000000000011795.Google Scholar
Abboud, H, Rossman, I, Mealy, MA, et al. Neuronal autoantibodies: differentiating clinically relevant and clinically irrelevant results. J Neurol. 2017;264:2284–92. DOI: 10.1007/s00415-017-8627-4.CrossRefGoogle ScholarPubMed
Hara, M, Ariño, H, Petit-Pedrol, M, et al. DPPX antibody-associated encephalitis: main syndrome and antibody effects. Neurology. 2017;88:1340–8. DOI: 10.1212/WNL.0000000000003796.Google Scholar
van Sonderen, A, Ariño, H, Petit-Pedrol, M, et al. The clinical spectrum of Caspr2 antibody-associated disease. Neurology. 2016;87:521–8. DOI: 10.1212/WNL.0000000000002917.Google Scholar
van Sonderen, A, Thijs, RD, Coenders, EC, et al. Anti-LGI1 encephalitis: clinical syndrome and long-term follow-up. Neurology. 2016;87:1449–56. DOI: 10.1212/WNL.0000000000003173.Google Scholar
Baumgartner, A, Rauer, S, Hottenrott, T, et al. Admission diagnoses of patients later diagnosed with autoimmune encephalitis. J Neurol. 2019;266:124–32. DOI: 10.1007/s00415-018-9105-3.CrossRefGoogle ScholarPubMed
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;70:1133–9. DOI: 10.1001/jamaneurol.2013.3216.CrossRefGoogle ScholarPubMed
Schieveld, JNM, Strik, JJMH, van Kraaij, S, Nicolai, J. Psychiatric manifestations and psychopharmacology of autoimmune encephalitis: a multidisciplinary approach. Handb Clin Neurol. 2019;165:285307. DOI: 10.1016/B978-0-444-64012-3.00017-4.Google Scholar
Uy, CE, Binks, S, Irani, SR. Autoimmune encephalitis: clinical spectrum and management. Pract Neurol. 2021;21:412–23. DOI: 10.1136/practneurol-2020-002567.Google Scholar
Irani, SR, Michell, AW, Lang, B, et al. Faciobrachial dystonic seizures precede Lgi1 antibody limbic encephalitis. Ann Neurol. 2011;69:892900. DOI: 10.1002/ana.22307.CrossRefGoogle ScholarPubMed
Graus, F, Titulaer, MJ, Balu, R, et al. A clinical approach to diagnosis of autoimmune encephalitis. Lancet Neurol. 2016;15:391404. DOI: 10.1016/S1474-4422(15)00401-9.Google Scholar
Jernbom Falk, A, Galletly, C, Just, D, et al. Autoantibody profiles associated with clinical features in psychotic disorders. Transl Psychiatry. 2021;11:474. DOI: 10.1038/s41398-021-01596-0.Google Scholar
Lang, K, Prüss, H. Frequencies of neuronal autoantibodies in healthy controls: estimation of disease specificity. Neurol Neuroimmunol Neuroinflamm. 2017;4:e386. DOI: 10.1212/NXI.0000000000000386.Google Scholar
Luo, Y, Li, J, Jiang, F, et al. Autoimmune encephalitis with psychotic manifestations and cognitive impairment presenting as schizophrenia: case report and literature review. Front Psychiatry. 2022;13:827138. DOI: 10.3389/fpsyt.2022.827138.Google Scholar
Guasp, M, Giné-Servén, E, Maudes, E, et al. Neuroimmunologic, and CSF investigations in first episode psychosis. Neurology. 2021;97:e61e75. DOI: 10.1212/WNL.0000000000012191.CrossRefGoogle ScholarPubMed
Herken, J, Prüss, H. Red flags: clinical signs for identifying autoimmune encephalitis in psychiatric patients. Front Psychiatry. 2017;8:25. DOI: 10.3389/fpsyt.2017.00025.CrossRefGoogle ScholarPubMed
Au, KLK, Latonas, S, Shameli, A, Auer, I, Hahn, C. Cerebrospinal fluid flow cytometry: utility in central nervous system lymphoma diagnosis. Can J Neurol Sci. 2020;47:382–8. DOI: 10.1017/cjn.2020.22.CrossRefGoogle ScholarPubMed
Blinder, T, Lewerenz, J. Cerebrospinal fluid findings in patients with autoimmune encephalitis-A systematic analysis. Front Neurol. 2019;10:804. DOI: 10.3389/fneur.2019.00804.CrossRefGoogle ScholarPubMed
Escudero, D, Guasp, M, Ariño, H, et al. Antibody-associated CNS syndromes without signs of inflammation in the elderly. Neurology. 2017;89:1471–5. DOI: 10.1212/WNL.0000000000004541.CrossRefGoogle Scholar
Hébert, J, Gros, P, Lapointe, S, et al. Searching for autoimmune encephalitis: beware of normal CSF. J Neuroimmunol. 2020;345:577285. DOI: 10.1016/j.jneuroim.2020.577285.Google Scholar
Lan, W, Li, J, Ai, P, Luo, W. Autoimmune glial fibrillary acidic protein astrocytopathy: clinical analysis and review of 15 cases. Acta Neurol Belg. 2023;123:1465–79. DOI: 10.1007/s13760-023-02268-0.Google Scholar
Zhu, B, Sun, M, Yang, T, Yu, H, Wang, L. Clinical, imaging features and outcomes of patients with anti-GFAP antibodies: a retrospective study. Front Immunol. 2023;14:1106490. DOI: 10.3389/fimmu.2023.1106490.CrossRefGoogle ScholarPubMed
Dalmau, J, Lancaster, E, Martinez-Hernandez, E, Rosenfeld, MR, Balice-Gordon, R. Clinical experience and laboratory investigations in patients with anti-NMDAR encephalitis. Lancet Neurol. 2011;10:6374. DOI: 10.1016/S1474-4422(10)70253-2.Google Scholar
Budhram, A, Dubey, D, Sechi, E, et al. Neural antibody testing in patients with suspected autoimmune encephalitis. Clin Chem. 2020;66:1496–509. DOI: 10.1093/clinchem/hvaa254.Google Scholar
Ropper, AH, Dalmau, J, Graus, F. Antibody-mediated encephalitis. N Engl J Med. 2018;378:840–51. DOI: 10.1056/NEJMra1708712.Google Scholar
Budhram, A, Leung, A, Nicolle, MW, Burneo, JG. Diagnosing autoimmune limbic encephalitis. CMAJ. 2019;191:E529E34. DOI: 10.1503/cmaj.181548.CrossRefGoogle ScholarPubMed
Budhram, A, Yang, L, Bhayana, V, Mills, JR, Dubey, D. SClinical sensitivity, specificity, and predictive value of neural antibody testing for autoimmune encephalitis. J Appl Lab Med. 2022;7:350–6. DOI: 10.1093/jalm/jfab127.Google Scholar
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;81:298309. DOI: 10.1002/ana.24881.CrossRefGoogle ScholarPubMed
Gadoth, A, Pittock, SJ, Dubey, D, et al. Expanded phenotypes and outcomes among 256 LGI1/CASPR2-IgG-positive patients. Ann Neurol. 2017;82:7992. DOI: 10.1002/ana.24979.Google Scholar
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;13:167–77. DOI: 10.1016/S1474-4422(13)70282-5.Google Scholar
Zekeridou, A, Kryzer, T, Guo, Y, et al. Phosphodiesterase 10A IgG: a novel biomarker of paraneoplastic neurologic autoimmunity. Neurology. 2019;93:e815e22.Google Scholar
Mandel-Brehm, C, Dubey, D, Kryzer, TJ, et al. Kelch-like protein 11 antibodies in seminoma-associated paraneoplastic encephalitis. N Engl J Med. 2019;381:4754.CrossRefGoogle ScholarPubMed
Muñiz-Castrillo, S, Hedou, JJ, Ambati, A, et al. Distinctive clinical presentation and pathogenic specificities of anti-AK5 encephalitis. Brain. 2021;144:2709–21.Google Scholar
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:e916. DOI: 10.1212/NXI.0000000000000916 Google Scholar
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:e658. DOI: 10.1212/NXI.0000000000000658.Google Scholar
Landa, J, Guasp, M, Míguez-Cabello, F, et al. Encephalitis with autoantibodies against the glutamate kainate receptors GluK2. Ann Neurol. 2021;90:101–17.Google Scholar
Valencia-Sanchez, C, Knight, AM, Hammami, MB, et al. Characterisation of TRIM46 autoantibody-associated paraneoplastic neurological syndrome. J Neurol Neurosurg Psychiatry. 2022;93:196200.Google Scholar
Zekeridou, A, Yang, B, Lennon, VA, et al. Anti-neuronal nuclear antibody 3 autoimmunity targets dachshund homolog 1. Ann Neurol. 2022;91:670–5.Google Scholar
Shelly, S, Kryzer, TJ, Komorowski, L, et al. Neurochondrin neurological autoimmunity. Neurol Neuroimmunol Neuroinflamm. 2019;6:e612. DOI: 10.1212/NXI.0000000000000612.CrossRefGoogle ScholarPubMed
Basal, E, Zalewski, N, Kryzer, TJ, et al. Paraneoplastic neuronal intermediate filament autoimmunity. Neurology. 2018;91:e1677e89.Google Scholar
Fang, B, McKeon, A, Hinson, SR, et al. Autoimmune glial fibrillary acidic protein astrocytopathy: a novel meningoencephalomyelitis. JAMA Neurol. 2016;73:1297–307.Google Scholar
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;13:276–86. DOI: 10.1016/S1474-4422(13)70299-0.Google Scholar
Budhram, A, Nicolle, MW, Yang, L. The positive predictive value of onconeural antibody testing: a retrospective review. Can J Neurol Sci. 2018;45:577–9. DOI: 10.1017/cjn.2018.74.Google Scholar
Déchelotte, B, Muñiz-Castrillo, S, Joubert, B, et al. Diagnostic yield of commercial immunodots to diagnose paraneoplastic neurologic syndromes. Neurol Neuroimmunol Neuroinflamm. 2020;7:e701. DOI: 10.1212/NXI.0000000000000701 Google Scholar
Ruiz-García, R, Martínez-Hernández, E, Saiz, A, Dalmau, J, Graus, F. The diagnostic value of onconeural antibodies depends on how they are tested. Front Immunol. 2020;11:1482. DOI: 10.3389/fimmu.2020.01482.Google Scholar
Graus, F, Vogrig, A, Muñiz-Castrillo, S, et al. Updated diagnostic criteria for paraneoplastic neurologic syndromes. Neurol Neuroimmunol Neuroinflamm. 2021;8:e1014. DOI: 10.1212/NXI.0000000000001014.Google Scholar
Graus, F, Saiz, A, Dalmau, J. GAD antibodies in neurological disorders - insights and challenges. Nat Rev Neurol. 2020;16:353–65.Google Scholar
Budhram, A, Mirian, A, McFadden, S, Edmond, P, Bhayana, V, Yang, L. Neural antibody testing for autoimmune encephalitis: a Canadian single-centre experience. Can J Neurol Sci. 2021;48:859–63. DOI: 10.1017/cjn.2021.23.Google Scholar
Bien, CG, Mirzadjanova, Z, Baumgartner, C, et al. Anti-contactin-associated protein-2 encephalitis: relevance of antibody titres, presentation and outcome. Eur J Neurol. 2017;24:175–86.Google Scholar
Garrido Sanabria, ER, Zahid, A, Britton, J, et al. CASPR2-IgG-associated autoimmune seizures. Epilepsia. 2022;63:709–22.Google Scholar
Sechi, E, Buciuc, M, Pittock, SJ, et al. Positive predictive value of myelin oligodendrocyte glycoprotein autoantibody testing. JAMA Neurol. 2021;78:741–6.Google Scholar
Held, F, Kalluri, SR, Berthele, A, Klein, AK, Reindl, M, Hemmer, B. Frequency of myelin oligodendrocyte glycoprotein antibodies in a large cohort of neurological patients. Mult Scler J Exp Transl Clin. 2021;7:20552173211022767.Google Scholar
Carvajal-González, A, Leite, MI, Waters, P, et al. Glycine receptor antibodies in PERM and related syndromes: characteristics, clinical features and outcomes. Brain. 2014;137:2178–92.Google Scholar
Hinson, SR, Lopez-Chiriboga, AS, Bower, JH, et al. Glycine receptor modulating antibody predicting treatable stiff-person spectrum disorders. Neurol Neuroimmunol Neuroinflamm. 2018;5:e438.Google Scholar
O’Connor, KC, McLaughlin, KA, De Jager, PL, et al. Self-antigen tetramers discriminate between myelin autoantibodies to native or denatured protein. Nat Med. 2007;13:211–7.CrossRefGoogle ScholarPubMed
Ogawa, R, Nakashima, I, Takahashi, T, et al. MOG antibody-positive, benign, unilateral, cerebral cortical encephalitis with epilepsy. Neurol Neuroimmunol Neuroinflamm. 2017;4:e322.Google Scholar
Budhram, A, Mirian, A, Le, C, Hosseini-Moghaddam, SM, Sharma, M, Nicolle, MW. Unilateral cortical FLAIR-hyperintense lesions in anti-MOG-associated encephalitis with seizures (FLAMES): characterization of a distinct clinico-radiographic syndrome. J Neurol. 2019;266:2481–7.CrossRefGoogle ScholarPubMed
Walikonis, JE, Lennon, VA. Radioimmunoassay for glutamic acid decarboxylase (GAD65) autoantibodies as a diagnostic aid for stiff-man syndrome and a correlate of susceptibility to type 1 diabetes mellitus. Mayo Clin Proc. 1998;73:1161–6.Google Scholar
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;131:2553–63.Google Scholar
Budhram, A, Sechi, E, Flanagan, EP, et al. Clinical spectrum of high-titre GAD65 antibodies. J Neurol Neurosurg Psychiatry. 2021;92:645654. DOI: 10.1136/jnnp-2020-325275.Google Scholar
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:e696. DOI: 10.1212/NXI.0000000000000696.CrossRefGoogle ScholarPubMed
Lang, B, Makuch, M, Moloney, T, et al. Intracellular and non-neuronal targets of voltage-gated potassium channel complex antibodies. J Neurol Neurosurg Psychiatry. 2017;88:353–61. DOI: 10.1136/jnnp-2016-314758.Google Scholar
Michael, S, Waters, P, Irani, SR. Stop testing for autoantibodies to the VGKC-complex: only request LGI1 and CASPR2. Pract Neurol. 2020;20:377–84. DOI: 10.1136/practneurol-2019-002494.Google Scholar
McCracken, L, Zhang, J, Greene, M, et al. Improving the antibody-based evaluation of autoimmune encephalitis. Neurol Neuroimmunol Neuroinflamm. 2017;4:e404. DOI: 10.1212/NXI.0000000000000404.Google Scholar
Sabater, L, Saiz, A, Dalmau, J, Graus, F. Pitfalls in the detection of CV2 (CRMP5) antibodies. J Neuroimmunol. 2016;15:80–3. DOI: 10.1016/j.jneuroim.2015.11.009.Google Scholar
Hollowell, JG, Staehling, NW, Flanders, WD, et al. T(4), and thyroid antibodies in the United States population (1988 to 1994): National Health and Nutrition Examination Survey (NHANES III). J Clin Endocrinol Metab. 2002;87:489–99. DOI: 10.1210/jcem.87.2.8182.Google Scholar
Mattozzi, S, Sabater, L, Escudero, D, et al. Hashimoto encephalopathy in the 21st century. Neurology. 2019;94:e217e24. DOI: 10.1212/WNL.0000000000008785.Google Scholar
Valencia-Sanchez, C, Pittock, SJ, Mead-Harvey, C, et al. Brain dysfunction and thyroid antibodies: autoimmune diagnosis and misdiagnosis. Brain Commun. 2021;3:fcaa233. DOI: 10.1093/braincomms/fcaa233.Google Scholar
Kelley, BP, Patel, SC, Marin, HL, Corrigan, JJ, Mitsias, PD, Griffith, B. Autoimmune encephalitis: pathophysiology and imaging review of an overlooked diagnosis. AJNR Am J Neuroradiol. 2017;38:1070–8. DOI: 10.3174/ajnr.A5086.CrossRefGoogle ScholarPubMed
Heine, J, Prüss, H, Bartsch, T, Ploner, CJ, Paul, F, Finke, C. Imaging of autoimmune encephalitis--relevance for clinical practice and hippocampal function. Neuroscience. 2015;309:6883. DOI: 10.1016/j.neuroscience.2015.05.037.Google Scholar
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;13:276–86. DOI: 10.1016/S1474-4422(13)70299-0.CrossRefGoogle Scholar
Spatola, M, Petit-Pedrol, M, Simabukuro, MM, et al. Investigations in GABAA receptor antibody-associated encephalitis. Neurology. 2017;88:1012–20. DOI: 10.1212/WNL.0000000000003713.Google Scholar
Deng, B, Cai, M, Qiu, Y, et al. MRI characteristics of autoimmune encephalitis with autoantibodies to GABAA receptor: a case series. Neurol Neuroimmunol Neuroinflamm. 2022;9:e1158. DOI: 10.1212/NXI.0000000000001158.Google Scholar
O’Connor, K, Waters, P, Komorowski, L, et al. GABAA receptor autoimmunity: a multicenter experience. Neurol Neuroimmunol Neuroinflamm. 2019;6:e552. DOI: 10.1212/NXI.0000000000000552.Google Scholar
Budhram, A, Sharma, M, Young, GB. Seizures in anti-Hu-associated extra-limbic encephalitis: characterization of a unique disease manifestation. Epilepsia. 2022;63:e172e177. DOI: 10.1111/epi.17437.Google Scholar
Chang, YC, Nouri, MN, Mirsattari, S, Burneo, JG, Budhram, A. Obvious” indications for neural antibody testing in epilepsy or seizures: the ONES checklist. Epilepsia. 2022;63:1658–70. DOI: 10.1111/epi.17238.Google Scholar
Fujimori, J, Ogawa, R, Murata, T, Takai, Y, Misu, T, Nakashima, I. Decreased subcortical T2 FLAIR signal with cortical T2 FLAIR hyperintense lesions in unilateral cerebral cortical encephalitis with myelin oligodendrocyte glycoprotein antibody. Neuroimmunol Rep. 2022;2:100096. DOI: 10.1016/j.nerep.2022.100096.Google Scholar
Valencia-Sanchez, C, Guo, Y, Krecke, KN, et al. Cerebral cortical encephalitis in myelin oligodendrocyte glycoprotein antibody-associated disease. Ann Neurol. 2023;93:297302. DOI: 10.1002/ana.26549.Google Scholar
Steriade, C, Tang-Wai, DF, Krings, T, Wennberg, R. Claustrum hyperintensities: a potential clue to autoimmune epilepsy. Epilepsia Open. 2017;2:476–80. DOI: 10.1002/epi4.12077.Google Scholar
Bordonne, M, Chawki, MB, Doyen, M, et al. Brain 18F-FDG PET for thediagnosis of autoimmune encephalitis: a systematic review and a meta-analysis. Eur J Nucl Med Mol Imaging. 2021;48:3847–58. DOI: 10.1007/s00259-021-05299-y.Google Scholar
Heine, J, Prüss, H, Bartsch, T, Ploner, CJ, Paul, F, Finke, C. Imaging of autoimmune encephalitis--relevance for clinical practice and hippocampal function. Neuroscience. 2015;309:6883. DOI: 10.1016/j.neuroscience.2015.05.037.Google Scholar
Neo, S, Yeo, T, Chen, Z, et al. Acute radiological features facilitate diagnosis and prognosis of anti-N-methyl-d-aspartate receptor (NMDAR) and anti-voltage-gated potassium channel (VGKC) encephalitis in adults. J Neurol Sci. 2020;419:117216. DOI: 10.1016/j.jns.2020.117216.Google Scholar
Piao, S, Bao, Y, Yang, L, et al. Brain MRI features of anti-N-methyl-D-aspartate (anti-NMDA) receptor encephalitis secondary to central nervous system infection in adult patients. Acta Radiol. 2022;64:760768. DOI: 10.1177/02841851221091443.Google Scholar
Baumgartner, A, Rauer, S, Mader, I, Meyer, PT. Cerebral FDG-PET and MRI findings in autoimmune limbic encephalitis: correlation with autoantibody types. J Neurol. 2013;260:2744–53. DOI: 10.1007/s00415-013-7048-2.Google Scholar
Graus, F, Dalmau, J. Role of (18)F-FDG-PET imaging in the diagnosis of autoimmune encephalitis - authors’ reply. Lancet Neurol. 2016;15:1010. DOI: 10.1016/S1474-4422(16)30130-2.Google Scholar
McKeon, A, Apiwattanakul, M, Lachance, DH, et al. Positron emission tomography-computed tomography in paraneoplastic neurologic disorders: systematic analysis and review. Arch Neurol. 2010;67:322–9. DOI: 10.1001/archneurol.2009.336.CrossRefGoogle Scholar
Sundermann, B, Schröder, JB, Warnecke, T, et al. Imaging workup of suspected classical paraneoplastic neurological syndromes: a systematic review and retrospective analysis of. Acad Radiol. 2017;24:1195–202. DOI: 10.1016/j.acra.2017.03.022.Google Scholar
Younes-Mhenni, S, Janier, MF, Cinotti, L, et al. FDG-PET improves tumour detection in patients with paraneoplastic neurological syndromes. Brain. 2004;127:2331–8. DOI: 10.1093/brain/awh247.Google Scholar
Moise, AM, Karakis, I, Herlopian, A, et al. Continuous EEG findings in autoimmune encephalitis. J Clin Neurophysiol. 2021;38:124–9. DOI: 10.1097/WNP.0000000000000654.Google Scholar
Wennberg, R, Steriade, C, Chen, R, Andrade, D. Frontal infraslow activity marks the motor spasms of anti-LGI1 encephalitis. Clin Neurophysiol. 2018;129:5968. DOI: 10.1016/j.clinph.2017.10.014.Google Scholar
Elkhider, H, Sharma, R, Kapoor, N, Vattoth, S, Shihabuddin, B. Autoimmune encephalitis and seizures, cerebrospinal fluid, imaging, and EEG findings: a case series. Neurol Sci. 2022;43:2669–80. DOI: 10.1007/s10072-021-05617-0.Google Scholar
Nathoo, N, Anderson, D, Jirsch, J. Extreme delta brush in anti-NMDAR encephalitis correlates with poor functional outcome and death. Front Neurol. 2021;12:686521. DOI: 10.3389/fneur.2021.686521.Google Scholar
Baykan, B, Gungor Tuncer, O, Vanli-Yavuz, EN, et al. Delta brush pattern is not unique to NMDAR encephalitis: evaluation of two independent long-term EEG cohorts. Clin EEG Neurosci. 2018;49:278–84. DOI: 10.1177/1550059417693168.Google Scholar
Wennberg, RA. Hyperventilation-induced focal seizures in adults: think autoimmune encephalitis. Ann Clin Transl Neurol. 2022;9:877–81. DOI: 10.1002/acn3.51566.Google Scholar
Alberti, MA, Povedano, M, Montero, J, Casasnovas, C. Early electrophysiological findings in Fisher-Bickerstaff syndrome. Neurologia (Engl Ed). 2020;35:40–5. DOI: 10.1016/j.nrl.2017.05.012.Google Scholar
De Wel, B, Claeys, KG. Neuromuscular hyperexcitability syndromes. Curr Opin Neurol. 2021;34:714–20. DOI: 10.1097/WCO.0000000000000963.Google Scholar
Lim, JA, Lee, ST, Moon, J, et al. Development of the clinical assessment scale in autoimmune encephalitis. Ann Neurol. 2019;85:352–8. DOI: 10.1002/ana.25421.Google Scholar
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;78:1333–44. DOI: 10.1001/jamaneurol.2021.3188.Google Scholar
Thompson, J, Bi, M, Murchison, AG, et al. The importance of early immunotherapy in patients with faciobrachial dystonic seizures. Brain. 2018;141:348–56. DOI: 10.1093/brain/awx323.Google Scholar
Thaler, FS, Zimmermann, L, Kammermeier, S, et al. German network for research on autoimmune encephalitis (GENERATE). Rituximab treatment and long-term outcome of patients with autoimmune encephalitis: real-world evidence from the GENERATE registry. Neurol Neuroimmunol Neuroinflamm. 2021;8:e1088. DOI: 10.1212/NXI.0000000000001088.Google Scholar
Dubey, D, Britton, J, McKeon, A, et al. Randomized placebo-controlled trial of intravenous immunoglobulin in autoimmune LGI1/CASPR2 epilepsy. Ann Neurol. 2020;87:313–23. DOI: 10.1002/ana.25655.Google Scholar
Zhang, Y, Liu, G, Jiang, M, Chen, W, Su, Y. Efficacy of therapeutic plasma exchange in patients with severe refractory anti-NMDA receptor encephalitis. Neurotherapeutics. 2019;16:828–37. DOI: 10.1007/s13311-019-00725-4.Google Scholar
Dalakas, MC. IgG4-mediated neurologic autoimmunities: understanding the pathogenicity of IgG4, ineffectiveness of IVIg, and long-lasting benefits of anti-B cell therapies. Neurol Neuroimmunol Neuroinflamm. 2022;9:e1116. DOI: 10.1212/NXI.0000000000001116.Google Scholar
Shayan, G, Adamiak, B, Choe, LH, Relkin, N, Lee, KH. Longitudinal effects of intravenous immunoglobulin on Alzheimer’s cerebrospinal fluid proteome. Electrophoresis. 2014;35:1821–7. DOI: 10.1002/elps.201300609.Google Scholar
Yelehe-Okouma, M, Czmil-Garon, J, Pape, E, Petitpain, N, Gillet, P. Drug-induced aseptic meningitis: a mini-review. Fundam Clin Pharmacol. 2018;32:252–60. DOI: 10.1111/fcp.12349.CrossRefGoogle ScholarPubMed
Alkabie, S, Budhram, A. Prolonged corticosteroids without maintenance immunotherapy for treatment of anti-LGI1 encephalitis: analysis of outcomes and relapse rate. Neurol Neuroimmunol Neuroinflamm. 2023;10. DOI: 10.1212/NXI.0000000000200115.Google Scholar
Nosadini, M, Eyre, M, Giacomini, T, et al. Early immunotherapy and longer corticosteroid treatment are associated with lower risk of relapsing disease course in pediatric MOGAD. Neurol Neuroimmunol Neuroinflamm. 2023;10:e200065. DOI: 10.1212/NXI.0000000000200065.Google Scholar
López-Chiriboga, AS, Flanagan, EP. Diagnostic and therapeutic approach to autoimmune neurologic disorders. Semin Neurol. 2018;38:392402. DOI: 10.1055/s-0038-1660819.Google Scholar
Nosadini, M, Thomas, T, Eyre, M, et al. International consensus recommendations for the treatment of pediatric NMDAR antibody encephalitis. Neurol Neuroimmunol Neuroinflamm. 2021;8:e1052. DOI: 10.1212/NXI.0000000000001052.Google Scholar
Dalmau, J, Graus, F. Diagnostic criteria for autoimmune encephalitis: utility and pitfalls for antibody-negative disease. Lancet Neurol. 2023;22:529–40. DOI: 10.1016/S1474-4422(23)00083-2.Google Scholar
Shin, YW, Lee, ST, Park, KI, et al. Treatment strategies for autoimmune encephalitis. Ther Adv Neurol Disord. 2018;11:1756285617722347. DOI: 10.1177/1756285617722347.CrossRefGoogle ScholarPubMed
Ledingham, D, Plant, M, Mustafa, F, Patil, B. Preserving fertility: using cyclophosphamide and other cytotoxics in young people. Pract Neurol. 2020;20:148–53. DOI: 10.1136/practneurol-2019-002247.Google Scholar
Vedeler, CA, Antoine, JC, Giometto, B, et al. Management of paraneoplastic neurological syndromes: report of an EFNS Task Force. Eur J Neurol. 2006;13:682–90. DOI: 10.1111/j.1468-1331.2006.01266.x.Google Scholar
Binks, S, Uy, C, Honnorat, J, Irani, SR. Paraneoplastic neurological syndromes: a practical approach to diagnosis and management. Pract Neurol. 2022;22:1931. DOI: 10.1136/practneurol-2021-003073.Google Scholar
Madondo, MT, Quinn, M, Plebanski, M. Low dose cyclophosphamide: mechanisms of T cell modulation. Cancer Treat Rev. 2016;42:39. DOI: 10.1016/j.ctrv.2015.11.005.Google Scholar
Ramwadhdoebe, TH, van Baarsen, LGM, Boumans, MJH, et al. Effect of rituximab treatment on T and B cell subsets in lymph node biopsies of patients with rheumatoid arthritis. Rheumatology (Oxford). 2019;58:1075–85. DOI: 10.1093/rheumatology/key428.Google Scholar
Fumal, A, Jobe, J, Pepin, JL, et al. Intravenous immunoglobulins in paraneoplastic brainstem encephalitis with anti-Ri antibodies. J Neurol. 2006;253:1360–1. DOI: 10.1007/s00415-006-0199-7.Google Scholar
van Vuurden, DG, Plötz, FB, de Jong, M, Bokenkamp, A, van Wijk, JA. Therapeutic total plasma exchange in a child with neuroblastoma-related anti-Hu syndrome. Pediatr Nephrol. 2005;20:1655–6. DOI: 10.1007/s00467-005-2004-8.Google Scholar
Hu, FQ, Shang, FR, Liu, JJ, Yuan, H. Plasma exchange for treating anti-Yo-associated paraneoplastic cerebellar degeneration: case report and literature review. Medicine (Baltimore). 2020;99:e21760. DOI: 10.1097/MD.0000000000021760.Google Scholar
Lee, WJ, Lee, ST, Moon, J, et al. Tocilizumab in autoimmune encephalitis refractory to rituximab: an institutional cohort study. Neurotherapeutics. 2016;13:824–32. DOI: 10.1007/s13311-016-0442-6.Google Scholar
Turnbull, MT, Siegel, JL, Becker, TL, Stephens, AJ, Lopez-Chiriboga, AS, Freeman, WD. Early bortezomib therapy for refractory anti-NMDA receptor encephalitis. Front Neurol. 2020;11:188. DOI: 10.3389/fneur.2020.00188.Google Scholar
Bost, C, Chanson, E, Picard, G, et al. Malignant tumors in autoimmune encephalitis with anti-NMDA receptor antibodies. J Neurol. 2018;265:2190–200. DOI: 10.1007/s00415-018-8970-0.Google Scholar
Gong, X, Chen, C, Liu, X, et al. Long-term functional outcomes and relapse of anti-NMDA receptor encephalitis: a cohort study in Western China. Neurol Neuroimmunol Neuroinflamm. 2021;8:e958. DOI: 10.1212/NXI.0000000000000958.Google Scholar
Xu, X, Lu, Q, Huang, Y, et al. Anti-NMDAR encephalitis: a single-center, longitudinal study in China. Neurol Neuroimmunol Neuroinflamm. 2020;7:e633. DOI: 10.1212/NXI.0000000000000633.Google Scholar
Boeck, AL, Logemann, F, Krauß, T, et al. Ovarectomy despite negative imaging in anti-NMDA receptor encephalitis: effective even late. Case Rep Neurol Med. 2013;2013:843192–3. DOI: 10.1155/2013/843192.Google Scholar
Cirkel, C, Cirkel, A, Royl, G, et al. On the quest for hidden ovarian teratomas in therapy-refractory anti-NMDA receptor encephalitis: a case report. Neurol Res Pract. 2022;4:15. DOI: 10.1186/s42466-022-00181-0.CrossRefGoogle ScholarPubMed
Iyengar, Y, Hébert, J, Climans, SA, et al. Ovarian resection in anti-N-methyl-D-aspartate receptor encephalitis: a comparison of surgical approaches. Front Neurol. 2022;13:1043785. DOI: 10.3389/fneur.2022.1043785.Google Scholar
Schneider, BJ, Naidoo, J, Santomasso, BD, et al. Management of immune-related adverse events in patients treated with immune checkpoint inhibitor therapy: ASCO guideline update. J Clin Oncol. 2021;39:4073–126. DOI: 10.1200/JCO.21.01440.Google Scholar
Haanen, JBAG, Carbonnel, F, Robert, C, et al. Management of toxicities from immunotherapy: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol. 2018;29:iv264iv6. DOI: 10.1093/annonc/mdy162.Google Scholar
Titulaer, MJ, Soffietti, R, Dalmau, J, et al. Screening for tumours in paraneoplastic syndromes: report of an EFNS task force. Eur J Neurol. 2011;18:19e3. DOI: 10.1111/j.1468-1331.2010.03220.x.Google Scholar
Abboud, H, Probasco, J, Irani, SR, et al. Autoimmune encephalitis: proposed recommendations for symptomatic and long-term management. J Neurol Neurosurg Psychiatry. 2021;92:897907. DOI: 10.1136/jnnp-2020-325302.Google Scholar
Pollak, TA, Lennox, BR, Müller, S, et al. Autoimmune psychosis: an international consensus on an approach to the diagnosis and management of psychosis of suspected autoimmune origin. Lancet Psychiatry. 2020;7:93108. DOI: 10.1016/S2215-0366(19)30290-1.Google Scholar
Zuliani, L, Nosadini, M, Gastaldi, M, et al. Management of antibody-mediated autoimmune encephalitis in adults and children: literature review and consensus-based practical recommendations. Neurol Sci. 2019;40:2017–30. DOI: 10.1007/s10072-019-03930-3.Google Scholar
Saleh, M, Bhosale, P, Menias, CO, et al. Ovarian teratomas: clinical features, imaging findings and management. Abdom Radiol (NY). 2021;46:2293–307. DOI: 10.1007/s00261-020-02873-0.Google Scholar
Lucchinetti, CF, Kimmel, DW, Lennon, VA. Paraneoplastic and oncologic profiles of patients seropositive for type 1 antineuronal nuclear autoantibodies. Neurology. 1998;50:652–7. DOI: 10.1212/wnl.50.3.652.Google Scholar
Trevisiol, C, Cani, I, Fabricio, ASC, Gion, M, Giometto, B, De Massis, P. Serum tumor markers in paraneoplastic neurologic syndromes: a systematic review of guidelines. Front Neurol. 2020;11:607553. DOI: 10.3389/fneur.2020.607553.Google Scholar
Rosenfeld, MR, Dalmau, J. Paraneoplastic neurologic syndromes. Neurol Clin. 2018;36:675–85. DOI: 10.1016/j.ncl.2018.04.015 2018.Google Scholar
Marsili, L, Marcucci, S, LaPorta, J, Chirra, M, Espay, AJ, Colosimo, C. Paraneoplastic neurological syndromes of the central nervous system: pathophysiology, diagnosis, and treatment. Biomedicines. 2023;11:1406. DOI: 10.3390/biomedicines11051406.Google Scholar
Rössling, R, Prüss, H. SOP: antibody-associated autoimmune encephalitis. Neurol Res Pract. 2020;2:1. DOI: 10.1186/s42466-019-0048-7.Google Scholar
Toledano, M, Pittock, SJ. Autoimmune epilepsy. Semin Neurol. 2015;35:245–58. DOI: 10.1055/s-0035-1552625.Google Scholar
Steriade, C, Britton, J, Dale, RC, et al. Acute symptomatic seizures secondary to autoimmune encephalitis and autoimmune-associated epilepsy: conceptual definitions. Epilepsia. 2020;61:1341–51. DOI: 10.1111/epi.16571.Google Scholar
Budhram, A, Burneo, JG. Acute symptomatic seizures, epilepsy, and autoimmune encephalitis: clarifying terminology in neural antibody-associated disease. Epilepsia. 2023;64:306–10. DOI: 10.1111/epi.17478.Google Scholar
Yeshokumar, AK, Coughlin, A, Fastman, J, et al. Seizures in autoimmune encephalitis-A systematic review and quantitative synthesis. Epilepsia. 2021;62:397407. DOI: 10.1111/epi.16807.Google Scholar
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;142:1631–43. DOI: 10.1093/brain/awz094.Google Scholar
Dalmau, J, Gleichman, AJ, Hughes, EG, et al. Anti-NMDA-receptor encephalitis: case series and analysis of the effects of antibodies. Lancet Neurol. 2008;7:1091–8. DOI: 10.1016/S1474-4422(08)70224-2.Google Scholar
Steriade, C, Gillinder, L, Rickett, K, et al. Discerning the role of autoimmunity and autoantibodies in epilepsy: a review. JAMA Neurol. 2021;78:1383–90. DOI: 10.1001/jamaneurol.2021.3113.Google Scholar
Casciato, S, Morano, A, Fattouch, J, et al. Factors underlying the development of chronic temporal lobe epilepsy in autoimmune encephalitis. J Neurol Sci. 2019;396:102–7. DOI: 10.1016/j.jns.2018.10.026.Google Scholar
de Bruijn, MAAM, van Sonderen, A, van Coevorden-Hameete, MH, et al. Evaluation of seizure treatment in anti-LGI1, anti-NMDAR, and anti-GABA. Neurology. 2019;92:e2185e96. DOI: 10.1212/WNL.0000000000007475.Google Scholar
Steriade, C, Moosa, ANV, Hantus, S, Prayson, RA, Alexopoulos, A, Rae-Grant, A. Electroclinical features of seizures associated with autoimmune encephalitis. Seizure. 2018;60:198204. DOI: 10.1016/j.seizure.2018.06.021.Google Scholar
Feyissa, AM, Lamb, C, Pittock, SJ, et al. Antiepileptic drug therapy in autoimmune epilepsy associated with antibodies targeting the leucine-rich glioma-inactivated protein 1. Epilepsia Open. 2018;3:348–56. DOI: 10.1002/epi4.12226.Google Scholar
Bozzetti, S, Rossini, F, Ferrari, S, et al. Epileptic seizures of suspected autoimmune origin: a multicentre retrospective study. J Neurol Neurosurg Psychiatry. 2020;91:1145–53. DOI: 10.1136/jnnp-2020-323841.Google Scholar
Cabezudo-García, P, Mena-Vázquez, N, Villagrán-García, M, Serrano-Castro, PJ. Efficacy of antiepileptic drugs in autoimmune epilepsy: a systematic review. Seizure. 2018;59:72–6. DOI: 10.1016/j.seizure.2018.05.004.Google Scholar
Gonzalez Castro, LN, Milligan, TA. Seizures in patients with cancer. Cancer. 2020;126:1379–89. DOI: 10.1002/cncr.32708.Google Scholar
Loughan, AR, Allen, A, Perna, R, Malkin, MG. Anti-N-methyl-D-aspartate receptor encephalitis: a review and neuropsychological case study. Clin Neuropsychol. 2016;30:150–63. DOI: 10.1080/13854046.2015.1132772.Google Scholar
Oldham, M. Autoimmune encephalopathy for psychiatrists: when to suspect autoimmunity and what to do next. Psychosomatics. 2017;58:228–44. DOI: 10.1016/j.psym.2017.02.014.Google Scholar
Marinas, JE, Matveychuk, D, McCombe, JA, Tymchuk, S, Dursun, SM, Baker, GB. Paraneoplastic and autoimmune encephalitis: alterations of mood and emotion. Handb Clin Neurol. 2021;183:221–34. DOI: 10.1016/B978-0-12-822290-4.00010-4.Google Scholar
Najjar, S, Pearlman, D, Devinsky, O, et al. Neuropsychiatric autoimmune encephalitis without VGKC-complex, NMDAR, and GAD autoantibodies: case report and literature review. Cogn Behav Neurol. 2013;26:3649. DOI: 10.1097/WNN.0b013e31828b6531.Google Scholar
Zhang, TY, Cai, MT, Zheng, Y, et al. Anti-alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor encephalitis: a review. Front Immunol. 2021;12:652820. DOI: 10.3389/fimmu.2021.652820.Google Scholar
Dale, RC, Gorman, MP, Lim, M. Autoimmune encephalitis in children: clinical phenomenology, therapeutics, and emerging challenges. Curr Opin Neurol. 2017;30:334–44. DOI: 10.1097/WCO.0000000000000443.Google Scholar
Liu, Y, Tian, Y, Guo, R, et al. Anti-NMDA receptor encephalitis: retrospective analysis of 15 cases. Literature review, and implications for gynecologists. J Healthc Eng. 2022;2022:4299791–9. DOI: 10.1155/2022/4299791.Google Scholar
Coffey, MJ, Cooper, JJ. Electroconvulsive therapy in Anti-N-methyl-D-aspartate receptor encephalitis: a case report and review of the literature. J ECT. 2016;32:225–9. DOI: 10.1097/YCT.0000000000000334.Google Scholar
Dhossche, DM, Wachtel, LE. Catatonia is hidden in plain sight among different pediatric disorders: a review article. Pediatr Neurol. 2010;43:307–15. DOI: 10.1016/j.pediatrneurol.2010.07.001.Google Scholar
Tanguturi, YC, Cundiff, AW, Fuchs, C. Anti-N-methyl d-aspartate receptor encephalitis and electroconvulsive therapy: literature review and future directions. Child Adolesc Psychiatr Clin N Am. 2019;28:7989. DOI: 10.1016/j.chc.2018.07.005.Google Scholar
Huang, A, Wang, J, Feng, DP, Wang, W, Li, X. Case analysis and literature review of thirteen patients with autoimmune encephalitis. Dis Markers. 2022;2022:4802480–7. DOI: 10.1155/2022/4802480.Google Scholar
Miya, K, Takahashi, Y, Mori, H. Anti-NMDAR autoimmune encephalitis. Brain Dev. 2014;36:645–52. DOI: 10.1016/j.braindev.2013.10.005.Google Scholar
Seery, N, Butzkueven, H, O’Brien, TJ, Monif, M. Contemporary advances in anti-NMDAR antibody (Ab)-mediated encephalitis. Autoimmun Rev. 2022;21:103057. DOI: 10.1016/j.autrev.2022.103057.Google Scholar
Scheer, S, John, RM. Anti-N-methyl-D-aspartate receptor encephalitis in children and adolescents. J Pediatr Health Care. 2016;30:347–58. DOI: 10.1016/j.pedhc.2015.09.004.Google Scholar
Vova, JA. A narrative review of pharmacologic approaches to symptom management of pediatric patients diagnosed with anti-NMDA receptor encephalitis. J Pediatr Rehabil Med. 2021;14:333–43. DOI: 10.3233/PRM-200677.Google Scholar
Deng, S, Qiu, K, Liu, H, Wu, X, Lei, Q, Lu, W. Clinical characteristics and short-term prognosis of autoimmune encephalitis: a single-center cohort study in Changsha, China. Front Neurol. 2019;10:539. DOI: 10.3389/fneur.2019.00539.Google Scholar
Broadley, J, Seneviratne, U, Beech, P, et al. Prognosticating autoimmune encephalitis: a systematic review. J Autoimmun. 2019;96:2434. DOI: 10.1016/j.jaut.2018.10.014.Google Scholar
Nguyen, L, Yang, JH, Goyal, S, Irani, N, Graves, JS. A systematic review and quantitative synthesis of the long-term psychiatric sequelae of pediatric autoimmune encephalitis. J Affect Disord. 2022;308:449–57. DOI: 10.1016/j.jad.2022.04.027.Google Scholar
Blum, RA, Tomlinson, AR, Jetté, N, Kwon, CS, Easton, A, Yeshokumar, AK. Assessment of long-term psychosocial outcomes in anti-NMDA receptor encephalitis. Epilepsy Behav. 2020;108:107088. DOI: 10.1016/j.yebeh.2020.107088.Google Scholar
Smith, KM, Dubey, D, Liebo, GB, Flanagan, EP, Britton, JW. Clinical course and features of seizures associated with LGI1-antibody encephalitis. Neurology. 2021;97:e1141e9. DOI: 10.1212/WNL.0000000000012465.Google Scholar
Di Giacomo, R, Deleo, F, Pastori, C, et al. Predictive value of high titer of GAD65 antibodies in a case of limbic encephalitis. J Neuroimmunol. 2019;337:577063. DOI: 10.1016/j.jneuroim.2019.577063.Google Scholar
Tsutsumi, Y, Yamamoto, Y, Ito, S, et al. Hepatitis B virus reactivation with a rituximab-containing regimen. World J Hepatol. 2015;7:2344–51.Google Scholar
Terrault, NA, Lok, ASF, McMahon, BJ, et al. Update on prevention, diagnosis, and treatment of chronic hepatitis B: AASLD. 2018 hepatitis B guidance. Hepatology. 2018;67:1560–99.Google Scholar
Foran, JM. Hepatitis C in the rituximab era. Blood. 2010;116:5081–2.Google Scholar
Zignego, AL, Gragnani, L, Giannini, C, Laffi, G. The hepatitis C virus infection as a systemic disease. Intern Emerg Med. 2012;7 Suppl 3:S201–8.Google Scholar
Ciuffreda, D, Pantaleo, G, Pascual, M. Effects of immunosuppressive drugs on HIV infection: implications for solid-organ transplantation. Transpl Int. 2007;20:649–58.Google Scholar
Heckmann, JM, Marais, S. Management issues in myasthenia gravis patients living with HIV: a case series and literature review. Front Neurol. 2020;11:775.Google Scholar
Diagnostic methods for the control of strongyloidiasis, Virtual meeting, 29 September 2020. Available from: Diagnostic methods for the control of strongyloidiasis, Virtual meeting, 29 September 2020 (who.int).Google Scholar
Schär, F, Trostdorf, U, Giardina, F, et al. Strongyloides stercoralis: global distribution and risk factors. PLoS Negl Trop Dis. 2013;7:e2288.Google Scholar
Mejia, R, Nutman, TB. Screening, prevention, and treatment for hyperinfection syndrome and disseminated infections caused by strongyloides stercoralis. Curr Opin Infect Dis. 2012;25:458–63. DOI: 10.1097/QCO.0b013e3283551dbd.Google Scholar
Roufosse, F. Management of hypereosinophilic syndromes. Immunol Allergy Clin North Am. 2015;35:561–75.Google Scholar
Hasan, T, Au, E, Chen, S, Tong, A, Wong, G. Screening and prevention for latent tuberculosis in immunosuppressed patients at risk for tuberculosis: a systematic review of clinical practice guidelines. BMJ Open. 2018;8:e022445.Google Scholar
Adult Immunization Schedule by Medical Condition and Other Indication CDC. Available from: http://www.cdc.gov/vaccines/schedules/hcp/imz/adult-conditions.html.Google Scholar
Canadian Immunization Guide Government of Canada. Available from: http://www.canada.ca/en/public-health/services/canadian-immunization-guide.html.Google Scholar
Prevention of Hepatitis B Reactivation During Immunosuppressive Therapy U.S. Department of Veterans Affairs. Available from: http://www.hepatitis.va.gov/hbv/reactivation-prevention.asp Google Scholar
Who to Vaccinate for Hepatitis B U.S. Department of Veterans Affairs [August 12, 2022]. Available from: http://www.hepatitis.va.gov/hbv/who-to-vaccinate.asp Google Scholar
Altered Immunocompetence General Best Practice Guidelines for Immunization: Best Practices Guidance of the Advisory Committee on Immunization Practices (ACIP) CDC. http://www.cdc.gov/vaccines/hcp/acip-recs/general-recs/immunocompetence.html.Google Scholar
Rubin, LG, Levin, MJ, Ljungman, P, et al. IDSA clinical practice guideline for vaccination of the immunocompromised host. Clin Infect Dis. 2013;58:309–18.Google Scholar
Curtis, JR, Johnson, SR, Anthony, DD, et al. American college of rheumatology guidance for COVID-19 vaccination in patients with rheumatic and musculoskeletal diseases: version 1. Arthritis Rheumatol. 2021;73:1093–107.Google Scholar
Tanrıöver, MD, Akar, S, Türkçapar, N, Karadağ, Ö., Ertenli, İ., Kiraz, S. Vaccination recommendations for adult patients with rheumatic diseases. Eur J Rheumatol. 2016;3:2935.Google Scholar
Drugs and Lactation Database NIH. Available from: http://www.ncbi.nlm.nih.gov/books/NBK501922/.Google Scholar
Bandoli, G, Palmsten, K, Forbess Smith, CJ, Chambers, CD. A review of systemic corticosteroid use in pregnancy and the risk of select pregnancy and birth outcomes. Rheum Dis Clin North Am. 2017;43:489502. DOI: 10.1016/j.rdc.2017.04.013.Google Scholar
Dobson, R, Dassan, P, Roberts, M, Giovannoni, G, Nelson-Piercy, C, Brex, PA. UK consensus on pregnancy in multiple sclerosis: ‘Association of British Neurologists’ guidelines. Pract Neurol. 2019;19:106–14. DOI: 10.1136/practneurol-2018-002060.Google Scholar
Hviid, A, Mølgaard-Nielsen, D. Corticosteroid use during pregnancy and risk of orofacial clefts. CMAJ. 2011;183:796804. DOI: 10.1503/cmaj.101063.Google Scholar
Achiron, A, Kishner, I, Dolev, M, et al. Effect of intravenous immunoglobulin treatment on pregnancy and postpartum-related relapses in multiple sclerosis. J Neurol. 2004;251:1133–7. DOI: 10.1007/s00415-004-0495-z.Google Scholar
Watson, WJ, Katz, VL, Bowes, WA. Plasmapheresis during pregnancy. Obstet Gynecol. 1990;76:451–7.Google Scholar
Chakravarty, EF, Murray, ER, Kelman, A, Farmer, P. Pregnancy outcomes after maternal exposure to rituximab. Blood. 2011;117:1499–506. DOI: 10.1182/blood-2010-07-295444.Google Scholar
Das, G, Damotte, V, Gelfand, JM, et al. Rituximab before and during pregnancy: a systematic review, and a case series in MS and NMOSD. Neurol Neuroimmunol Neuroinflamm. 2018;5:e453. DOI: 10.1212/NXI.0000000000000453.Google Scholar
Smith, JB, Hellwig, K, Fink, K, Lyell, DJ, Piehl, F, Langer-Gould, A. Rituximab, MS, and pregnancy. Neurol Neuroimmunol Neuroinflamm. 2020;7:e734. DOI: 10.1212/NXI.0000000000000734.Google Scholar
Cooley, L, Dendle, C, Wolf, J, et al. Consensus guidelines for diagnosis, prophylaxis and management of Pneumocystis jirovecii pneumonia in patients with haematological and solid malignancies, 2014. Intern Med J. 2014;44:1350–63. DOI: 10.1111/imj.12599.Google Scholar
Urbancic, KF, Ierino, F, Phillips, E, Mount, PF, Mahony, A, Trubiano, JA. Taking the challenge: a protocolized approach to optimize Pneumocystis pneumonia prophylaxis in renal transplant recipients. Am J Transplant. 2018;18:462–6. DOI: 10.1111/ajt.14498.Google Scholar
Park, JW, Curtis, JR, Jun, KI, et al. Primary prophylaxis for pneumocystis jirovecii pneumonia in patients receiving rituximab. Chest. 2022;161:1201–10. DOI: 10.1016/j.chest.2021.11.007.Google Scholar
Miloslavsky, EM, Naden, RP, Bijlsma, JW, et al. Development of a Glucocorticoid Toxicity Index (GTI) using multicriteria decision analysis. Ann Rheum Dis. 2017;76:543–6. DOI: 10.1136/annrheumdis-2016-210002.Google Scholar
Caplan, A, Fett, N, Rosenbach, M, Werth, VP, Micheletti, RG. Prevention and management of glucocorticoid-induced side effects: a comprehensive review: gastrointestinal and endocrinologic side effects. J Am Acad Dermatol. 2017;76:11–6. DOI: 10.1016/j.jaad.2016.02.1239.Google Scholar
Kersey, JP, Broadway, DC. Corticosteroid-induced glaucoma: a review of the literature. Eye (Lond). 2006;20:407–16. DOI: 10.1038/sj.eye.6701895.Google Scholar
Liu, D, Ahmet, A, Ward, L, et al. A practical guide to the monitoring and management of the complications of systemic corticosteroid therapy. Allergy Asthma Clin Immunol. 2013;9:30. DOI: 10.1186/1710-1492-9-30.Google Scholar
Liu, MM, Reidy, AB, Saatee, S, Collard, CD. Perioperative steroid management: approaches based on current evidence. Anesthesiology. 2017;127:166–72. DOI: 10.1097/ALN.0000000000001659.Google Scholar
Hamilton, DD, Cotton, BA. Cosyntropin as a diagnostic agent in the screening of patients for adrenocortical insufficiency. Clin Pharmacol. 2010;2:7782.Google Scholar
Kersey, JP, Broadway, DC. Corticosteroid-induced glaucoma: a review of the literature. Eye (Lond). 2006;20:407–16.Google Scholar
Caplan, A, Fett, N, Rosenbach, M, Werth, VP, Micheletti, RG. Prevention and management of glucocorticoid-induced side effects: a comprehensive review: gastrointestinal and endocrinologic side effects. J Am Acad Dermatol. 2017;76:11–6.Google Scholar
Allen, CS, Yeung, JH, Vandermeer, B, Homik, J. Bisphosphonates for steroid-induced osteoporosis. Cochrane Database Syst Rev. 2016;10:CD001347.Google Scholar
Huynh, G, Reinert, JP. Pharmacological management of steroid-induced psychosis: a review of patient cases. J Pharm Technol. 2021;37:120–6.Google Scholar
Warrington, TP, Bostwick, JM. Psychiatric adverse effects of corticosteroids. Mayo Clin Proc. 2006;81:1361–7.Google Scholar
Figure 0

Figure 1: Diagnostic criteria for possible autoimmune encephalitis.

Figure 1

Figure 2: Diagnostic progression in autoimmune encephalitis (excluding ADEM, Bickerstaff’s). ADEM = Acute disseminated encephalomyelitis; CNS = central nervous system; CSF = cerebral spinal fluid; EEG = electroencephalography; GAD65 = glutamic acid decarboxylase-65; IgG = immunoglobulin G; MRI = magnetic resonance imaging (of the brain); OGBs = oligoclonal bands; PET = positron emission tomography (of the brain).

Figure 2

Table 1: Initial laboratory investigations of suspected AIE. Tests in bold are strongly recommended to establish the diagnosis and exclude common mimics. Optional tests are also listed if clinically indicated by presenting history

Figure 3

Table 2: Test methodologies employed for neural antibody detection in patients with suspected autoimmune encephalitis

Figure 4

Figure 3: Proposed treatment algorithm for AIE. PLEX = plasma exchange; IVIg = intravenous immunoglobulin; GAD = glutamic acid decarboxylase; GFAP = glial fibrillary acidic protein.

Figure 5

Table 3: Anti-neural antibodies associated with encephalitis

Figure 6

Figure 4: Neoplasm screening protocol. AIE: autoimmune encephalitis, CT: computed tomography, MRI: magnetic resonance imaging, PET: positron emission tomography. *Can be considered in a relapse, especially in patients with intermediate or high-risk antibodies. **For benign neoplasms oncology involvement may not be necessary as treatment is typically surgical.

Figure 7

Table 4: Antibody type and malignancy risk

Figure 8

Table 5: Recommended agents for neuropsychiatric symptoms

Figure 9

Table 6: Overview of screening response by pathogen

Figure 10

Table 7: Vaccination recommendations – adapted from CDC 2022 and Canadian guidelines (Rubin et al. 2014; Government of Canada 2021; CDC 2022)

Figure 11

Table 8: Recommendations for treatment related vaccine delays

Figure 12

Table 9: Steroid side effect mitigation226

Supplementary material: File

Hahn et al. supplementary material

Hahn et al. supplementary material
Download Hahn et al. supplementary material(File)
File 209.4 KB