Skip to main content
Erschienen in: Journal of Neurology 5/2013

Open Access 01.05.2013 | Review

Paraneoplastic and non-paraneoplastic autoimmunity to neurons in the central nervous system

verfasst von: Nico Melzer, Sven G. Meuth, Heinz Wiendl

Erschienen in: Journal of Neurology | Ausgabe 5/2013

Abstract

Autoimmune central nervous system (CNS) inflammation occurs both in a paraneoplastic and non-paraneoplastic context. In a widening spectrum of clinical disorders, the underlying adaptive (auto) immune response targets neurons with a divergent role for cellular and humoral disease mechanisms: (1) in encephalitis associated with antibodies to intracellular neuronal antigens, neuronal antigen-specific CD8+ T cells seemingly account for irreversible progressive neuronal cell death and neurological decline with poor response to immunotherapy. However, a pathogenic effect of humoral immune mechanisms is also debated. (2) In encephalitis associated with antibodies to synaptic and extrasynaptic neuronal cell surface antigens, potentially reversible antibody-mediated disturbance of synaptic transmission and neuronal excitability occurs in the absence of excessive neuronal damage and accounts for a good response to immunotherapy. However, a pathogenic effect of cellular immune mechanisms is also debated. We provide an overview of entities, clinical hallmarks, imaging features, characteristic laboratory, electrophysiological, cerebrospinal fluid and neuropathological findings, cellular and molecular disease mechanisms as well as therapeutic options in these two broad categories of inflammatory CNS disorders.

Introduction

The human central nervous system (CNS) can be targeted by aberrant cellular and humoral immune responses, which can either be triggered by systemic infections, and vaccinations (“postinfectious/-vaccinal autoimmune encephalitis”) and a variety of cancers (“paraneoplastic autoimmune encephalitis”) or occur without an (yet) identifiable cause (“non-paraneoplastic autoimmune encephalitis”) [25]. The scope of neurological disorders, in which such misguided adaptive immune responses are directed towards the oligodendrocyte and myelin-sheath is well described [24]. However, in a variety of immune-mediated CNS disorders, neurons seem to be targeted by adaptive cellular and humoral (auto) immune responses of both paraneoplastic and non-paraneoplastic origin [35]. Here, we summarize important clinical phenotypes together with their typical paraclinical measures, putative disease mechanisms, and therapeutic options in this emerging class of inflammatory CNS disorders.

Prerequisites for neuron-directed autoimmunity in the CNS

Antigen-specific cellular and humoral immune responses directed towards CNS neurons are believed to develop as a multi-step process [73]. Soluble or cell-bound neuronal or neuronal-like antigens are engulfed and presented in the context of MHC II and co-stimulatory molecules to CD4+ T cells by professional antigen-presenting cell (APCs) within secondary lymphatic organs (e.g., cervical lymph nodes). This in turn permits CD4+ T cells via cytokine secretion and ligation of CD40 to license APCs to cross-present these antigens in the context of MHC I and co-stimulatory molecules to naive CD8+ T cells, which then become activated and may acquire cytotoxic effector functions (cellular effectors). A lack of such CD4+ T cell help usually results in anergy of CD8+ T cells. Depending on the local cytokine milieu mainly provided by CD4+ T cells, CD8+ T cells with different functional polarization may develop [86]. Tc1 cells are differentiated in the presence of IL-2 and IL-12, which induce the transcription factor T-bet. They produce IFN-γ and TNF-α and exert strong cytotoxicity. Tc2 cells are differentiated in the presence of IL-4, which induces the transcription factor GATA-3. They produce IL-4, IL-5, and IL-13 and exert a less robust cytotoxicity. Tc17 cells are differentiated in the presence of TGF-β and IL-6, which mainly induce the transcription factor RORγt. They produce IL-17 and have been reported to exert only weak cytotoxicity.
Naive B cells produce both IgM (and IgD) that are anchored in their plasma membrane and function as BCRs [48]. Naive B cells that encounter, ingest, and present their cognate so-called “thymus-dependent (TD) antigen” in the context of MHC II and co-stimulatory molecules to CD4+ T cells are in turn activated via cytokine secretion and ligation of CD40 and become antibody-secreting plasma cells (humoral effectors). Thereby, B cells can further diversify their Ig-genes by two DNA-modifying mechanisms [48]; somatic hypermutation and class switch recombination generate highly specific and adapted humoral responses. Somatic hypermutation introduces in a transcription-dependent manner non-templated point mutations in the variable (V) region of Ig genes, thereby enabling the selection of antibodies with increased affinity for the antigen. In contrast, class switch recombination modulates antibody effector function by replacing one constant (C) region with another, while retaining the binding specificity of the BCR. Depending on the cytokine milieu mainly provided by the CD4+ T cells, activated B cells undergo antibody class switching to produce IgG, IgA, or IgE antibodies [48]. Switch to IgG1 and IgG3 promoting complement activation and antibody-mediated cellular cytotoxicity by NK cells occurs in the presence of IFN-γ. In contrast, switch to IgG2, IgG4, and IgA promoting antigen-neutralizing effects occurs in the presence of IL-4 and IL-5. Although some pathogen-derived “thymus-independent (TI) antigens” may induce somatic hypermutation and class switch recombination in B cells independent from CD4+ T cell help, a lack of such help usually results in persistent secretion of complement-activating IgM (and IgD; [48]).
Following peripheral activation, both antibody-secreting plasma cells and cytotoxic CD8+ T cells (together with CD4+ T cells) may enter the CNS to attack neurons and cause functional and structural impairment [69, 97]. Moreover, under such inflammatory conditions, even antibodies produced in the periphery may permeate the blood–brain barrier (BBB) by various paracellular and transcellular mechanisms and thus contribute to neuron-directed immunity, whereas under physiological conditions, the BBB is usually impermeable for antibodies [23] (see Fig. 1).
In general, both effector arms of the adaptive immune response may be activated irrespective of the cellular localization of the neuronal antigen or its antigenic epitope (plasma membrane vs. interior cellular compartments). In terms of relevant effector mechanisms, plasma cell-derived antibodies usually recognize discontinuous conformational epitopes composed of segments of the respective neuronal plasma membrane protein antigen that are brought together in its three-dimensional structure and exposed on the neuronal plasma membrane. Antibodies may thus specifically impact the function and expression of theses antigens. Whether antibodies may also bind to and impact the function or expression of intracellular neuronal antigens, either by passive uptake into the neuron or by active binding to intracellular antigens that are transiently exposed to the plasma membrane is currently a matter of debate [30, 96]. Moreover, peptides derived from both intracellular and plasma membrane neuronal antigens might potentially be recognized by antibodies when exposed on the surface membrane in complex with MHC I molecules, although this is usually performed by CD8+ T cells.
Cytotoxic CD8+ T cells usually recognize continuous linear peptide epitopes consisting of 8–10 amino acids that are derived from intracellular neuronal proteins by extensive antigen processing and presented in the context of MHC I molecules on the cell surface membrane. Whether peptides derived from neuronal surface membrane antigens are also presented to cytotoxic CD8+ T cells in the context of MHC I molecules is unclear at present. In both cases, CD8+ T cells cannot directly impact the function or expression of their cognate antigens, but recognize their expression by the respective neuron. This enables them to contribute to neuronal dysfunction and cell death by the antigen-dependent release of effector molecules (perforin, granzymes) from cytotoxic granules. Indeed, we could show that two separate functional consequences result from a direct cell-to-cell contact between antigen-presenting neurons and antigen-specific CD8+ T cells. (1) An immediate impairment of electrical signaling in single neurons and neuronal networks occurs as a result of massive shunting of the membrane capacitance after insertion of channel-forming perforin (and probably activation of other transmembrane conductances), which is paralleled by an increase of intracellular Ca2+ levels. (2) Antigen-dependent neuronal apoptosis may occur independently of perforin and members of the granzyme B cluster, suggesting that extracellular effects can substitute for intracellular delivery of granzymes by perforin. Thus, electrical silencing is an immediate consequence of MHC I-restricted interaction of CD8+ T cells with neurons. Of course, these changes in neuronal excitability are not induced specifically in response to a certain antigen, but apply to all antigen-presenting neurons encountered by activated cytotoxic CD8+ T cells [69, 71].

Paraneoplastic autoimmune encephalitis is probably mediated by cytotoxic CD8+ T cells specific for intracellular neuronal antigens

An ever-growing number of paraneoplastic CNS disorders are defined by the presence of IgG antibodies in the serum and CSF directed against intracellular neuronal antigens aberrantly expressed also by tumor cells (“onco-neuronal antibodies”) [67]. These tumors often contain neuronally differentiated tissue (germ cell tumors), express certain neuroendokrine peptides (SCLC, neuroblastoma), or occur in organs with a role in immune regulation (thymoma). However, due to the intracellular localization of the antigens, the humoral immune response is considered a non-pathogenic “epiphenomenon” solely indicating neuron-directed immunity and defining its antigen. In contrast, a variety of findings suggest a pathogenic role of cytotoxic CD8+ T cells for neuronal damage in these disorders: (1) neuronal damage often correlates with the number of CD8+ T cells, (2) CD8+ T cells are found in the CNS parenchyma in close spatial proximity to neuronal target cells, (3) CD8+ T cells show an activated phenotype with substantial expression of the effector molecules (perforin and granzymes) in cytotoxic granules with a polar orientation towards the target cell membrane, (4) CD8+ T cells stain positive for CD107 indicating recent exocytosis of cytotoxic granules (i.e., degranulation), (5) neuronal target cells exhibit substantial cell surface expression of MHC I molecules allowing for cognate antigen-recognition by CD8+ T cells, (6) CD8+ T cells exhibit a restricted T cell receptor (TCR) repertoire (i.e., oligoclonal expansions) suggesting that they have expanded from a few precursors locally responding to a distinct neuronal antigen [6, 69]. These criteria, however, have not yet been demonstrated entirely for all entities.
In clinical terms, inflammatory CNS disorders associated with IgG antibodies against intracellular neuronal antigens are characterized by a multifocal presentation of CNS-related symptoms involving the neocortex, the limbic system, basal ganglia, brainstem, cerebellum, and spinal cord as well as PNS-related symptoms involving radices, plexus, and peripheral nerves in a variable extent (Table 1). The clinical presentation partially reflects the pattern of expression of the respective neuronal antigen: ANNA-1 targets nuclear ELAVL (“Hu”) proteins expressed in central and peripheral neurons, and the corresponding clinical syndrome typically includes CNS and PNS manifestations [58]. In contrast, ANNA-2 targets nuclear NOVA-1 and -2 (“Ri”) proteins expressed in central, but not peripheral neurons and the clinical syndrome is usually restricted to the CNS [75] (Table 1). MRI findings include T2/FLAIR hyperintense, occasionally contrast-enhancing lesions in the cortex, medial temporal lobes, basal ganglia, brainstem, cerebellum, and spinal cord. Inflammatory changes are usually found in CSF studies including lymphocytic pleocytosis, mildly elevated protein together with intrathecal IgG synthesis and oligoclonal bands, but normal glucose and lactate levels [19]. The disease entities usually exhibit a chronic progressive clinical course and poor response to immunotherapy, especially to antibody-depleting therapies. Even successful removal of the tumor considered to drive the pathogenic immune response is usually not associated with disease amelioration [19].
Table 1
Encephalitis associated with antibodies against intracellular neuronal antigens
Entity
Patients
Triggers
Clinical hallmarks
Imaging
Electrophysiology
Laboratory
 
ANNA-1 (Hu) encephalitis
Age 30–80 years (median 60 years), gender male 75 %, ANPR >500
Tumors: lung (SCLC in adults), neuroendocrine tissue (neuroblastoma in children), rarely thymus (thymoma)
Neocortical and limbic encephalitis, brainstem encephalitis, cerebellitis, myelitis, cranial neuropathy, radiculopathy, plexopathy, peripheral (sensory, motor, sensorimotor, autonomic) neuropathy
MRI: T2/FLAIR hyperintense signal, occasionally Gd-enhancement and atrophy in cortex, medial temporal lobes, brainstem, cerebellum or spinal cord FDG-PET: focal hypermetabolism at early disease-stages, focal hypometabolism at late disease-stages
EEG: 1. focal or widespread interictal and ictal epileptiform activity, 2. focal or generalized slowing Nerve conduction studies: predominantly axonal sensory, motor or sensorimotor neuropathy, plexopathy, radiculopathy
Anti-neuronal nuclear IgG1/3 antibody type 1 (ANNA-1; anti-Hu antibody) in serum and CSF targeting nuclear ELAVL (“Hu”) proteins expressed in central and peripheral neurons and tumor cells and implicated in neuronal post-transcriptional RNA regulation (“onco-neuronal” antibodies)
 
ANNA-2 (Ri) encephalitis
Age 50–80 years (median 65 years), gender female 80 %, ANPR 100
Tumors: lung (SCLC), breast
Neocortical and limbic encephalitis, brainstem encephalitis, cerebellitis, myelitis
MRI: T2/FLAIR hyperintense signal, occasionally Gd-enhancement and atrophy in cortex, medial temporal lobes, brainstem, cerebellum or spinal cord FDG-PET: focal hypermetabolism at early disease-stages, focal hypometabolism at late disease-stages
EEG: 1. focal or widespread interictal and ictal epileptiform activity, 2. focal or generalized slowing
Anti-neuronal nuclear IgG1/3 antibody type 2 (ANNA-2; anti-Ri antibody) in serum and CSF targeting nuclear NOVA-1 and –2 (“Ri”) proteins expressed in central but not peripheral neurons and tumor cells and implicated in regulation of alternative splicing of neuronal RNA encoding synaptic proteins (N-, P/Q-type Ca2+ channels; “onco-neuronal” antibodies)
 
ANNA-3 encephalitis
Age 10–85 years (median 60 years), gender female 50 %, ANPR 10
Tumors: lung (SCLC, adenocarcinoma), esophagus (adenocarcinoma)
Limbic encephalitis, brainstem encephalitis, cerebellitis, myelitis, peripheral (sensory, sensorimotor) neuropathy
MRI: T2/FLAIR hyperintense signal, occasionally Gd-enhancement and atrophy in medial temporal lobes, brainstem, cerebellum or spinal cord FDG-PET: focal hypermetabolism at early disease-stages, focal hypometabolism at late disease-stages
EEG: 1. focal or widespread interictal and ictal epileptiform activity, 2. focal or generalized slowing Nerve conduction studies: predominantly axonal sensory, motor or sensorimotor neuropathy
Anti-neuronal nuclear IgG1/3 antibody type 3 (ANNA-3) in serum and CSF targeting a nuclear 170 kDa protein of unknown molecular identity expressed in central (Purkinje neurons) and peripheral neurons and tumor cells (“onco-neuronal” antibodies)
 
AGNA (SOX-1) encephalitis
–,
Tumors: lung (SCLC)
Limbic encephalitis, brainstem encephalitis, cerebellitis, peripheral neuropathy Lambert-Eaton myasthenic syndrome (LEMS)
MRI: T2/FLAIR hyperintense signal, occasionally Gd-enhancement and atrophy in medial temporal lobes, brainstem or cerebellum FDG-PET: focal hypermetabolism at early disease-stages, focal hypometabolism at late disease-stages
EEG: 1. focal or widespread interictal and ictal epileptiform activity, 2. focal or generalized slowing Nerve conduction studies: predominantly axonal sensory, motor or sensorimotor neuropathy EMG: decrement of compound muscle action potential on 2–5/s repetitive nerve stimulation, increment of compound muscle action potential on 30–50/s repetitive nerve stimulation or “post-tetanic” stimulation
Anti-glial/neuronal nuclear IgG1/3 antibody (AGNA) in serum and CSF targeting nuclear SOX1 protein expressed predominantly in developing and adult cerebellar Bergmann glia cells, central and peripheral neurons and tumor cells and implicated in transcription regulation during neuronal development (“onco-neuronal” antibodies)
 
ANPR 100
Anti-Ma1/Ma2 encephalitis
Age 40–70 years (median 55 years), gender male 75 %, ANPR 75
Tumors: women with non-germ cell tumors of ovary, breast, colon, lung (combined anti-Ma1/Ma2-encephalitis), men with germ cell tumors of testis (pure anti-Ma2-encephalitis)
Anti-Ma1/Ma2 encephalitis: limbic encephalitis, diencephalitis, brainstem encephalitis and cerebellitis Anti-Ma2 encephalitis: limbic encephalitis, diencephalitis, brainstem encephalitis without cerebellitis
MRI: T2/FLAIR hyperintense signal, occasionally Gd-enhancement and atrophy in medial temporal lobes, diencephalon, brainstem or cerebellum FDG-PET: focal hypermetabolism at early disease-stages, focal hypometabolism at late disease-stages
EEG: 1. focal or widespread interictal and ictal epileptiform activity, 2. focal or generalized slowing
Anti-Ma1 (PNMA1)- and/or Anti-Ma2 (PNMA2) IgG1/3 antibody in serum and CSF targeting nucleolar/subnuclear Ma1 (PNMA1) and Ma2 (PNMA2) proteins expressed in central neurons and in tumor cells and implicated in transcription regulation (“onco-neuronal” antibodies)
 
Anti-CV2 (CRMP-3) encephalitis
Age 50–75 years (median 60 years), gender female 60 %, ANPR 30
Tumors: lung (SCLC), thymus (thymoma), kidney (carcinoma), thyroid gland (carcinoma)
Uveitis, retinitis, optic neuritis, limbic encephalitis, cerebellitis, myelitis peripheral (sensory, motor, sensorimotor) neuropathy neuromyelitis optica-like clinical phenotype (optic neuritis + myelitis)
MRI: T2/FLAIR hyperintense signal, occasionally Gd-enhancement and atrophy in optic nerve, medial temporal lobes, cerebellum or spinal cord FDG-PET: focal hypermetabolism at early disease-stages, focal hypometabolism at late disease-stages
EEG: 1. focal or widespread interictal and ictal epileptiform activity, 2. focal or generalized slowing Nerve conduction studies: axonal and demyelinating sensory, motor or sensorimotor neuropathy
Anti-collapsin response-mediated protein 3 IgG1/3 antibody (CRMP-3-IgG) in serum and CSF targeting cytoplasmic collapsin response-mediated protein 3 expressed in a subpopulation of oligodendrocytes and central neurons, Schwann cells and peripheral neurons and tumor cells and implicated in axon guidance, synaptic organization and other cellular responses (“onco-neuronal” antibodies)
 
Anti-CRMP-5 encephalitis
Age 50–75 years (median 60 years), gender female 60 %, ANPR 150
Tumors: lung (SCLC), thymus (thymoma), kidney (carcinoma), thyroid gland (carcinoma)
Optic neuritis with and without retinitis and other cranial neuropathies, neocortical and limbic encephalitis, “basal ganglionitis”, cerebellitis, myelitis, radiculopathy, plexopathy, peripheral (sensory, motor, sensorimotor, autonomic) neuropathy, myasthenia gravis (MG), Lambert-Eaton myasthenic syndrome (LEMS), neuromyotonia neuromyelitis optica-like clinical phenotype (optic neuritis + myelitis)
MRI: T2/FLAIR hyperintense signal, occasionally Gd-enhancement and atrophy in optic and other cranial nerves, neocortex, medial temporal lobes, basal ganglia, cerebellum or spinal cord FDG-PET: focal hypermetabolism at early disease-stages, focal hypometabolism at late disease-stages
EEG: 1. focal or widespread interictal and ictal epileptiform activity, 2. focal or generalized slowing Nerve conduction studies: axonal and demyelinating sensory, motor or sensorimotor neuropathy
Anti-collapsin response-mediated protein 5 IgG1/3 antibody (CRMP-5-IgG) in serum and CSF targeting cytoplasmic collapsin response-mediated protein 5 expressed in central and peripheral neurons including synapses and tumor cells and implicated in axon guidance, synaptic organization and other cellular responses (“onco-neuronal” antibodies)
 
Anti-PCA-1 (Yo) encephalitis
Age 60–70 years (median 65 years), gender female 90 %, ANPR 150
Tumors: breast, ovary, fallopian tube, endometrium
Cerebellitis, brainstem encephalitis, myelitis, peripheral (sensory, motor, sensorimotor, autonomic) neuropathy
MRI: T2/FLAIR hyperintense signal, occasionally Gd-enhancement and atrophy in cerebellum, brainstem and spinal cord FDG-PET: focal hypermetabolism at early disease-stages, focal hypometabolism at late disease-stages
EEG: Generalized slowing or normal Nerve conduction studies: predominantly axonal sensory, motor or sensorimotor neuropathy
Purkinje cell cytoplasmic IgG1/3 autoantibody type 1 (PCA-1; anti-Yo antibody) in serum and CSF targeting cytoplasmic CDR2 and CDR62 (“Yo”) proteins expressed in central and peripheral neurons especially cerebellar Purkinje neurons and tumors cells and implicated in downregulation of transcription via inhibition of c-Myc (“onco-neuronal” antibodies)
 
Anti-PCA-2 encephalitis
Age 45–85 years (median 60 years), gender female 70 %, ANPR 10
Tumors: lung (SCLC)
Limbic encephalitis, brainstem encephalitis, cerebellitis, peripheral neuropathy
MRI: T2/FLAIR hyperintense signal, occasionally Gd-enhancement and atrophy in medial temporal lobes, brainstem or cerebellum FDG-PET: focal hypermetabolism at early disease-stages, focal hypometabolism at late disease-stages
EEG: 1. focal or widespread interictal and ictal epileptiform activity, 2. focal or generalized slowing Nerve conduction studies: predominantly axonal sensory, motor or sensorimotor neuropathy
Purkinje cell cytoplasmic IgG1/3 autoantibody type 2 (PCA-2) in serum and CSF targeting a cytoplasmic 280 kDa protein of unknown molecular identity expressed in central and peripheral neurons especially cerebellar Purkinje neurons and tumors cells (“onco-neuronal” antibodies)
 
Anti-PCA-Tr encephalitis
Age 15–70 years (median 60 years), gender male 75 %, ANPR 120
Tumors: Hodgkin lymphoma, non-Hodgkin lymphoma, occasionally solid tumors
(Limbic encephalitis), cerebellitis
MRI: T2/FLAIR hyperintense signal, occasionally Gd-enhancement and atrophy in cerebellum FDG-PET: focal hypermetabolism at early disease-stages, focal hypometabolism at late disease-stages
EEG: generalized slowing or normal
Purkinje cell cytoplasmic IgG1/3 autoantibody type Tr (PCA-Tr) in serum and CSF targeting Delta/Notch-like epidermal growth factor-related receptor (DNER) expressed in central neurons especially in cerebellar Purkinje neurons and occasionally in tumor cells (Reed-Sternberg cells) and implicated in neuron–glia interactions through notch signaling (“onco-neuronal” antibodies)
 
Anti-amphiphysin encephalitis
Age 50–80 years (median 65 years), gender female 60 %, ANPR 100
Tumors: lung (SCLC, non-SCLC), breast, melanoma
Limbic encephalitis, cerebellitis, myelitis, stiff-person syndrome, radiculopathy, plexopathy, peripheral (sensory, motor, sensorimotor) neuropathy
MRI: usually normal, occasionally T2/FLAIR hyperintense signal, Gd-enhancement and atrophy in medial temporal lobes, cerebellum and spinal cord
EEG: Usually normal, occasionally focal or generalized epileptiform activity and slowing Nerve conduction studies: predominantly axonal sensory, motor or sensorimotor neuropathy EMG: excessive startle response, continuous involuntary motor activity in agonistic and antagonistic muscles
Anti-Amphiphysin IgG antibody in serum and CSF targeting cytoplasmic amphiphysin expressed in central and peripheral neurons (presynaptic terminals) and in tumor cells and implicated in retrieving vesicle membranes from the axon terminal’s plasma membrane after depolarization-induced exocytosis of neurotransmitter (“onco-neuronal” antibodies)
 
Anti-GAD 65 encephalitis
Age 15–80 years (median 60 years), gender female 80 %, ANPR 200
Tumors (occasionally): lung (SCLC, non-SCLC), thymus (thymoma), colon, pancreas, breast, thyroid, and renal cell carcinoma
Limbic encephalitis, epilepsy, basal ganglionitis, brainstem encephalitis, cerebellitis, myelitis, stiff-person syndrome, Progressive Encephalomyelitis with Rigidity and Myoclonus (PERM) stiffness/rigidity, excessive startle, brainstem dysfunction (anti-GAD 65 IgG antibody titer usually >2000 U/ml in RIA) Autoimmune diabetes mellitus (anti-GAD 65 IgG antibody titer usually <20 U/ml in RIA)
MRI: usually normal, occasionally T2/FLAIR hyperintense signal, Gd-enhancement and atrophy in medial temporal lobes, brainstem, cerebellum and spinal cord
EEG: Usually normal, occasionally focal or generalized epileptiform activity and slowing EMG: excessive startle response, continuous involuntary motor activity in agonistic and antagonistic muscles
Anti-GAD 65 IgG antibody in serum (>20,00 U/ml) and CSF, usually with intrathecal anti-GAD 65 IgG synthesis targeting the cytoplasmic 65 kDa isoform of glutamic acid decarboxylase expressed in central GABAergic neurons (presynaptic terminals), pancreatic islet cells and occasionally tumor cells and implicated in converting excitatory neurotransmitter glutamate to inhibitory neurotransmitter GABA (occasionally “onco-neuronal” antibodies)
 
Anti-GABAA-receptor-associated protein encephalitis
–,
Neocortical encephalitis, epilepsy, cerebellitis, stiff-person syndrome
EMG: continuous involuntary motor activity in agonistic and antagonistic muscles
Anti-GABARAP IgG antibody in serum and CSF targeting cytoplasmic and membrane GABARAP expressed in central neurons (postsynaptic density of GABAergic synapses) and implicated in clustering and anchoring GABAA-receptors in the postsynaptic membrane by facilitating binding to the cytoskeleton
 
ANPR 20
Anti-gephyrin encephalitis
–,
Tumor: undifferentiated mediastinal carcinoma
Stiff-person syndrome
MRI: normal
EMG: continuous involuntary motor activity in agonistic and antagonistic muscles
Anti-gephyrin IgG antibody in serum and CSF targeting cytoplasmic gephyrin protein expressed in central neurons (postsynaptic density of GABAergic and glycinergic synapses) and implicated in clustering and anchoring GABAA- and glycine receptors in the postsynaptic membrane by facilitating binding to the cytoskeleton
 
ANPR : 1
Entity
Patients
CSF
Neuropathology
Putative disease mechanisms
Therapy
Disease course and prognosis
References
ANNA-1 (Hu) encephalitis
Age 30–80 years (median 60 years), gender male 75 %, ANPR >500
Lymphocytic pleocytosis (median 3/μl, range 1–8/μl), normal glucose and lactate, mildly elevated protein (median 78 mg/dL, range 49–135 mg/dL), intrathecal IgG synthesis and OCB
Inflammatory infiltrates (perivascular CD4+ T cells and B cells, parenchymal CD8+ T cells), gliosis, microglia activation, neuronal loss and neuronophagia
Neuronal cell death mediated by neuronal-antigen specific cytotoxic CD8+ T cells
Immunotherapy: 1. GCS together with steroid-sparing agents, IVIG, PE/IA, 2. rituximab, cyclophosphamide; Tumor-therapy: (operation, radiation, chemotherapy)
Chronic disease course: usually poor response to immunotherapy; some improvement or stabilization may be achieved by prompt tumor-therapy
[1, 33, 37, 49, 58, 67, 79, 83, 85, 87]
ANNA-2 (Ri) encephalitis
Age 50–80 years (median 65 years), gender female 80 %, ANPR 100
Lymphocytic pleocytosis (median 5/μl, range 1–15/μl), normal glucose and lactate, mildly elevated protein (median 55 mg/dL, range 39–415 mg/dL), intrathecal IgG synthesis and OCB
Inflammatory infiltrates (perivascular CD4+ T cells and B cells, parenchymal CD8+ T cells and macrophages), gliosis, microglia activation, neuronal loss and neuronophagia
Neuronal cell death mediated by neuronal-antigen specific cytotoxic CD8+ T cells
Immunotherapy: 1. GCS together with steroid-sparing agents, IVIG, PE/IA, 2. rituximab, cyclophosphamide; Tumor-therapy: (operation, radiation, chemotherapy)
Chronic disease course: usually poor response to immunotherapy and tumor-therapy
[37, 59, 67, 75, 79, 83, 85]
ANNA-3 encephalitis
Age 10–85 years (median 60 years), gender female 50 %, ANPR 10
Lymphocytic pleocytosis, normal glucose and lactate, mildly elevated protein, intrathecal IgG synthesis and OCB
Presumably neuronal cell death mediated by neuronal-antigen specific cytotoxic CD8+ T cells
Immunotherapy: 1. GCS together with steroid-sparing agents, IVIG, PE/IA, 2. rituximab, cyclophosphamide; Tumor-therapy: (operation, radiation, chemotherapy)
Chronic disease course: usually poor response to immunotherapy and tumor-therapy
[12, 37, 67, 83, 85]
AGNA (SOX-1) encephalitis
–,
Lymphocytic pleocytosis, normal glucose and lactate, mildly elevated protein, intrathecal IgG synthesis and OCB
Presumably neuronal cell death mediated by neuronal-antigen specific cytotoxic CD8+ T cells
Immunotherapy: 1. GCS together with steroid-sparing agents, IVIG, PE/IA, 2. rituximab, cyclophosphamide; Tumor-therapy: (operation, radiation, chemotherapy)
Chronic disease course: usually poor response to immunotherapy and tumor-therapy
[36, 37, 67, 79, 83, 85, 92]
ANPR 100
Anti-Ma1/Ma2 encephalitis
Age 40–70 years (median 55 years), gender male 75 %, ANPR 75
Lymphocytic pleocytosis (median 3/μl, range 1–20/μl), normal glucose and lactate, mildly elevated protein (median 53 mg/dL, range 39–70 mg/dL), intrathecal IgG synthesis and OCB
Inflammatory infiltrates (perivascular CD4+ T cells and B cells, parenchymal CD8+ T cells and macrophages), gliosis, microglia activation, neuronal loss and neuronophagia
Neuronal cell death mediated by neuronal-antigen specific cytotoxic CD8+ T cells
Immunotherapy: 1. GCS together with steroid-sparing agents, IVIG, PE/IA, 2. rituximab, cyclophosphamide; Tumor-therapy: (operation, radiation, chemotherapy)
Chronic disease course: about 50 % may improve or stabilize, about 50 % deteriorate following combined tumor- and immunotherapy (outcome is better in pure anti-Ma2-encephalitis (men) than in combined anti-Ma1/Ma2-encephalitis (women))
[16, 17, 37, 39, 67, 79, 82, 83, 85, 98]
Anti-CV2 (CRMP-3) encephalitis
Age 50–75 years (median 60 years), gender female 60 %, ANPR 30
Lymphocytic pleocytosis (8–70/μl), normal glucose and lactate, mildly elevated protein (47–400 mg/dL), intrathecal IgG synthesis and OCB
Inflammatory infiltrates (perivascular CD4+ T cells and B cells, parenchymal CD8+ T cells), gliosis, microglia activation, neuronal loss and neuronophagia
Neuronal cell death mediated by neuronal-antigen specific cytotoxic CD8+ T cells
Immunotherapy: 1. GCS together with steroid-sparing agents, IVIG, PE/IA, 2. rituximab, cyclophosphamide; Tumor-therapy: (operation, radiation, chemotherapy)
Chronic disease course: usually poor response to immunotherapy and tumor-therapy
[2, 4143, 67, 79, 83]
Anti-CRMP-5 encephalitis
Age 50–75 years (median 60 years), gender female 60 %, ANPR 150
Lymphocytic pleocytosis (8–370/μl), normal glucose and lactate, mildly elevated protein (47–400 mg/dL), intrathecal IgG synthesis and OCB
Inflammatory infiltrates (perivascular CD4+ T cells and B cells, parenchymal CD8+ T cells), gliosis, microglia activation, neuronal loss and neuronophagia
Neuronal cell death mediated by neuronal-antigen specific cytotoxic CD8+ T cells
Immunotherapy: 1. GCS together with steroid-sparing agents, IVIG, PE/IA, 2. rituximab, cyclophosphamide; Tumor-therapy: (operation, radiation, chemotherapy)
Chronic disease course: usually poor response to immunotherapy and tumor-therapy
[14, 67, 79, 83, 99]
Anti-PCA-1 (Yo) encephalitis
Age 60–70 years (median 65 years), gender female 90 %, ANPR 150
Lymphocytic pleocytosis (median 4/μl, range 1–22/μl),normal glucose and lactate, mildly elevated protein (median 54 mg/dL, range 36–88 mg/dL), intrathecal IgG synthesis and OCB
Inflammatory infiltrates (perivascular CD4+ T cells and B cells, parenchymal CD8+ T cells and macrophages), gliosis, microglia activation, neuronal loss and neuronophagia
Neuronal cell death mediated by neuronal-antigen specific cytotoxic CD8+ T cells
Immunotherapy: 1. GCS together with steroid-sparing agents, IVIG, PE/IA, 2. rituximab, cyclophosphamide; Tumor-therapy: (operation, radiation, chemotherapy)
Chronic disease course: usually poor response to immunotherapy and tumor-therapy
[28, 67, 68, 79, 81, 83, 85]
Anti-PCA-2 encephalitis
Age 45–85 years (median 60 years), gender female 70 %, ANPR 10
Lymphocytic pleocytosis normal glucose and lactate, mildly elevated protein, intrathecal IgG synthesis and OCB
Neuronal cell death mediated by neuronal-antigen specific cytotoxic CD8+ T cells
Immunotherapy: 1. GCS together with steroid-sparing agents, IVIG, PE/IA, 2. rituximab, cyclophosphamide; Tumor-therapy: (operation, radiation, chemotherapy)
Chronic disease course: usually poor response to immunotherapy and tumor-therapy
[67, 79, 83, 85, 94]
Anti-PCA-Tr encephalitis
Age 15–70 years (median 60 years), gender male 75 %, ANPR 120
Lymphocytic pleocytosis (median 7/μl, range 3–10/μl) normal glucose and lactate, mildly elevated protein (median 41 mg/dL, range 25–72 mg/dL), usually no intrathecal IgG synthesis or OCB
Presumably neuronal cell death mediated by neuronal-antigen specific cytotoxic CD8+ T cells
Immunotherapy: 1. GCS together with steroid-sparing agents, IVIG, PE/IA, 2. rituximab, cyclophosphamide; Tumor-therapy: (operation, radiation, chemotherapy)
Usually chronic disease course with poor response to immunotherapy and tumor-therapy, a subset of patients may stabilize or improve with eradication of the PCA-Tr antibody following successful tumor-therapy
[5, 8, 9, 22, 37, 67, 79, 83, 85]
Anti-amphiphysin encephalitis
Age 50–80 years (median 65 years), gender female 60 %, ANPR 100
Lymphocytic pleocytosis (median 22/μl, range 2–42/μl), normal glucose and lactate, mildly elevated protein (median 104 mg/dL, range 57–151 mg/dL), intrathecal IgG synthesis and OCB
Inflammatory infiltrates (perivascular CD4+ T cells and B cells, parenchymal CD8+ T cells and macrophages), gliosis, microglia activation, neuronal loss and neuronophagia
Neuronal cell death and functional impairment mediated by neuronal-antigen specific cytotoxic CD8 + T cells alternatively: Binding to the amphiphysin protein and internalization of the IgG antibody into the presynaptic ending with disturbance of GABAergic > glutamatergic synaptic transmission
Immunotherapy: 1. GCS together with steroid-sparing agents, IVIG, PE/IA, 2. rituximab, cyclophosphamide; Tumor-therapy: (operation, radiation, chemotherapy)
Chronic disease course: usually poor response to immunotherapy and tumor-therapy
[21, 26, 30, 67, 76, 79, 83, 91]
Anti-GAD 65 encephalitis
Age 15–80 years (median 60 years), gender female 80 %, ANPR 200
Lymphocytic pleocytosis, normal glucose and lactate, mildly elevated protein intrathecal IgG synthesis and OCB
Inflammatory infiltrates (perivascular CD4+ T cells and B cells, parenchymal CD8+ T cells and macrophages), gliosis, microglia activation, neuronal loss and neuronophagia
Neuronal cell death and functional impairment mediated by neuronal-antigen specific cytotoxic CD8+ T cells
Immunotherapy: 1. GCS together with steroid-sparing agents, IVIG, PE/IA, 2. rituximab, cyclophosphamide; Tumor-therapy: occasionally
Chronic disease course: usually poor response to immunotherapy (and tumor-therapy)
[60, 67, 77, 79, 83, 84, 89, 90]
Anti-GABAA-receptor-associated protein encephalitis
–,
Neuronal cell death and functional impairment mediated by neuronal-antigen specific cytotoxic CD8+ T cells alternatively: binding to the GABARAP and internalization of the IgG antibody into the postsynaptic density, impairment of GABAA-receptor clustering and disturbance of GABAergic synaptic transmission
Immunotherapy: 1. GCS together with steroid-sparing agents, IVIG, PE/IA, 2. rituximab, cyclo-phosphamide
[80]
ANPR 20
Anti-gephyrin encephalitis
–,
Normal
Neuronal cell death and functional impairment mediated by neuronal-antigen specific cytotoxic CD8+ T cells alternatively: binding to the gephyrin protein and internalization of the IgG antibody into the postsynaptic density, impariment of GABAA- and glycine-receptor clustering and disturbance of GABAergic and glycinergic synaptic transmission
Immunotherapy: 1. GCS together with steroid-sparing agents, IVIG, PE/IA, 2. rituximab, cyclo-phosphamide; Tumor-therapy: (operation, radiation, chemotherapy)
Complete remission upon tumor removal
[10]
ANPR : 1
However, there exists a group of CNS disorders with antibodies against intracellular neuronal antigens located mainly at presynaptic (GAD65) or postsynaptic (GABARAP, Gephyrin) sites of inhibitory GABAergic and glycinergic synapses, which less frequently associate with tumors. In these entities, there is often no evidence for cellular or humoral neuronal cytotoxicity, although some patients show neuroaxonal swelling, chromatolysis and vacuolization of neurons, microglial proliferation, as well as infiltration and apposition of cytotoxic CD8+ T cells to neurons [6, 40]. Further, there are reports of potentially pathogenic humoral mechanisms probably targeting inhibitory CNS neuronal networks in anti-GAD encephalitis [29, 57, 61], but until now the specificity of possible pathogenic antibodies has not been elucidated. These findings together with the wide spectrum of clinical presentations suggest that anti-GAD encephalitis comprises of a quite heterogenous group of CNS disorders with regard to their etiologies and disease mechanisms.

Paraneoplastic and non-paraneoplastic autoimmune encephalitis is probably mediated by antibodies to neuronal surface membrane antigens

Autoimmune inflammatory CNS disorders associated with IgG antibodies in the serum and CSF directed against neuronal surface membrane antigens [54, 97] occur both in a paraneoplastic and non-paraneoplastic context. Tumors assumed to drive the pathogenic immune response usually contain neuronally differentiated tissue expressing the respective neuronal antigen or occur in organs with a role in immune regulation, such as the thymus.
Antibodies bind to synaptic and extra-synaptic ligand- and voltage-gated ion channels (Table 2) involved in excitatory (AMPA-, NMDA-, mGluR1-, mGluR5-, and nAch-receptors, VGCC, VGKC) and inhibitory (GABAB- and Glycine-receptors, VGKC) synaptic transmission and plasticity. Moreover, these antibodies also target neuronal membrane proteins implicated in clustering of voltage-gated potassium channels inside the synapse [leucine-rich glioma-inactivated 1 (LGI1)] or outside the synapse at the juxtaparanodal region of the node of Ranvier [contactin-2 and contactin-associated protein-like 2 (CASPR2)] thereby indirectly impacting neuronal excitability.
Table 2
Encephalitis associated with antibodies against neuronal surface membrane antigens (modified and extended from Melzer et al. [70])
Entity
Patients
Triggers
Clinical hallmarks
Imaging
Electrophysiology
Laboratory
 
Anti-NMDA-R encephalitis
Age 1–80 years (median 20 years), gender female 80, ANPR 500
Tumors (age-, gender-, race-dependent, about 50 %): ovary/testis (teratoma), breast, lung (SCLC), lymphoma
Multistage cortico-subcortical encephalopathy: 1. Prodromal phase (days) often with (viral) infections, 2. Psychiatric symptoms (1–2 weeks): psychosis, confusion, amnesia, dysphasia, 3. neurological symptoms (weeks): movement disorders (choreoathetoid, mute, catatonic), autonomic instability, respiratory failure, reduced consciousness, seizures, 4. recovery of symptoms in reverse of their appearance
MRI: no correlating signal abnormalities (50 %), transient T2/FLAIR hyperintense signal in cerebral cortex cerebellar cortex, basal ganglia, brainstem, spinal cord, Gd-enhancement in cortical meninges, basal ganglia, frontotemporal or mediotemporal cortical atrophy (50 %)
EEG: 1. focal or widespread interictal and ictal epileptiform activity, 2. generalized slowing
Anti-NMDA-R (NR1/NR2) IgG1/3-antibody in serum and CSF, intrathecal anti-NMDA-R IgG1/3 synthesis, titers correlate well with clinical disease course/therapy tumors often express NMDA-R
 
Anti-AMPA-R encephalitis
Age 40–80 years (median 60 years), gender female 90 %, ANPR 15
Tumors (about 70 %); thymus (thymoma), breast, lung (SCLC, non-SCLC)
Limbic encephalitis: 1. focal temporal lobe and secondary generalized seizures, 2. short-term memory loss/disorientation, 3. psychiatric symptoms (psychosis) evolving within days–weeks
MRI: T2/FLAIR hyperintense signal in one or both medial temporal lobes (often asymmetric), rarely Gd-enhancement (90 %)
EEG: 1. focal interictal and ictal epileptiform activity in one or both temporal lobes, 2. focal or generalized slowing
Anti-AMPA-R (GluR1/2) IgG antibody in serum and CSF, intrathecal anti-AMPA-R IgG synthesis, titers correlate with clinical disease course/therapy tumors often express AMPA-R
 
Anti-GABAB-R encephalitis
Age 25–75 years (median 60 years), gender female 50 %, ANPR 25
Tumors (about 60 %): lung (SCLC, non-SCLC), thymus (thymoma)
Limbic encephalitis with prominent seizures: 1. focal temporal lobe and secondary generalized seizures, 2. short-term memory loss/disorientation, 3. psychiatric symptoms (psychosis) evolving within days–weeks
MRI: T2/FLAIR hyperintense signal in one or both medial temporal lobes (often asymmetric), rarely Gd-enhancement (70 %)
EEG: 1. focal interictal and ictal epileptiform activity in one or both temporal lobes, 2. focal or generalized slowing
Anti-GABAB-R (GABAB1) IgG1 antibody in serum and CSF, intrathecal anti-GABAB-R IgG1 synthesis, correlation of titers with clinical disease course/therapy not yet determined expression of GABAB-R by tumors not yet determined
 
Anti-Glycine-R encephalitis
Age 30–60 years (median 50 years), gender male 80 %, ANPR 4
Tumors: typically none (thymoma)
Hyperekplexia, stiff-person syndrome, progressive encephalomyelitis with rigidity and myoclonus (PERM): stiffness/rigidity, excessive startle, brainstem dysfunction
MRI: typically normal
EMG: excessive startle response, continuous involuntary motor activity
Anti-Gly-R (GlyRα1) IgG1 antibody in serum and CSF, intrathecal anti-Gly-R IgG1 synthesis, titers seem to correlate with clinical disease course/therapy
 
Anti-VGKC complex encephalitis: LGI1
Age 30–80 years (median 60 years), gender male 65 %, ANPR 120
Tumors: (about 10 %): thymus (thymoma), lung (SCLC)
Limbic encephalitis: 1. focal temporal lobe and secondary generalized seizures, 2. short-term memory loss/disorientation, 3. psychiatric symptoms (psychosis) evolving within days–weeks
MRI: T2/FLAIR hyperintense signal in one or both medial temporal lobes (often asymmetric), rarely Gd-enhancement
EEG: 1. interictal focal epileptiform activity or slowing over one or both temporal lobes, 2. ictal focal or generalized epileptiform activity
Anti-LGI1 IgG4/1 antibody in serum and CSF, intrathecal anti-LGI1 IgG4/1 synthesis infrequent, correlation of titers with clinical disease course/therapy not yet determined expression of LGI1 by tumors not yet determined SIADH with hyponatremia (115–130 mmol/l)
 
Anti-VGKC complex encephalitis: CASPR2
Age 45–80 years (median 60 years), gender male 85 %, ANPR 40
Tumors (about 10 %): thymus (thymoma), lung (SCLC)
Morvan’s syndrome: 1. psychiatric disturbance 2. seizures, 3. sleep disturbance (insomnia), 4. dysautonomia, 5. neuromyotonia in various combinations cerebellitis
MRI: T2/FLAIR hyperintense signal in one or both medial temporal lobes (often asymmetric), rarely Gd-enhancement (about 40 %)
EEG: 1. focal or generalized interictal and ictal epileptiform activity, 2. focal or generalized slowing EMG: spontaneous doublet, triplet or multiplet single-unit discharges
Anti-CASPR2 IgG4/1 antibody in serum and CSF, intrathecal anti-CASPR2 IgG4/1 synthesis not determined, correlation of titers with clinical disease course/therapy not yet determined expression of CASPR2 by tumors not yet determined SIADH with hyponatremia (115–130 mmol/l)
 
Anti-mGlu-R1 encephalitis
Age 20–50 years, gender female 100 %, ANPR 3
Tumors: none or Hodgkin lymphoma (in remission)
Cerebellitis
MRI: normal or T2/FLAIR hyperintense signal and atrophy of the cerebellum
Anti-mGlu-R1 IgG antibody in serum and CSF, intrathecal anti-mGlu-R1 IgG synthesis, correlation of titers with clinical disease course/therapy not yet determined
 
Anti-mGlu-R5 encephalitis
Age 15–45 years, gender female 50 %, ANPR 2
Tumors: Hodgkin lymphoma
Limbic encephalitis (Ophelia syndrome): 1. focal temporal lobe and secondary generalized seizures, 2. short-term memory loss/disorientation, 3. psychiatric symptoms (psychosis) evolving within days–weeks
MRI: normal or T2/FLAIR hyperintense signal in one or both medial temporal lobes and other cortical and subcortical gray matter areas
EEG: 1. interictal focal epileptiform activity or slowing over one or both temporal lobes, 2. ictal focal or generalized epileptiform activity
Anti-mGlu-R5 IgG antibody in serum and CSF, intrathecal anti-mGlu-R5 IgG synthesis not determined, correlation of titers with clinical disease course/therapy not yet determined
 
Anti-P/Q type/N-type VGCC encephalitis
Age 30–80 years, gender male 80 %, ANPR 120
Tumors: (about 50 %) lung (SCLC), breast, ovary
Cerebellitis, Lambert–Eaton myasthenic syndrome (LEMS)
MRI: normal or T2/FLAIR hyperintense signal and atrophy of the cerebellum
EMG: decrement of compound muscle action potential on 2–5/s repetitive nerve stimulation, increment of compound muscle action potential on 30–50/s repetitive nerve stimulation or “post-tetanic” stimulation
Anti-P/Q type/N-type VGCC IgG antibody in serum and CSF, intrathecal anti-P/Q type/N-type VGCC IgG synthesis, correlation of titers with clinical disease course/therapy not yet determined expression of VGCC by tumors not yet determined
 
Anti-nAch-R encephalitis
Age 17–103 years (median 65 years), gender male (55 %), ANRP 150
Tumors (carcinoma in about 30 %): lung (non-SCLC, SCLC), breast, ovary, uterus, prostate, colon, thyroid, kidney, bladder, thymus (thymoma), melanoma
Cortical encephalitis, basal ganglionitis, dysautonomia peripheral (sensory, motor, sensorimotor, autonomic) neuropathy
MRI: usually normal, occasionally T2/FLAIR hyperintense signal in basal ganglia
EEG: generalized slowing Nerve conduction studies: predominantly axonal sensory, motor or sensorimotor neuropathy
Anti-nAch-R IgG antibody in serum and CSF, intrathecal anti- nAch-R IgG synthesis not yet determined, strong correlation of titers with clinical disease course/therapy expression of nAch-R by tumors not yet determined
 
Entity
Patients
CSF
Neuropathology
Putative disease mechanisms
Therapy
Disease course and prognosis
References
Anti-NMDA-R encephalitis
Age 1–80 years (median 20 years), gender female 80, ANPR 500
Lymphocytic pleocytosis (median 32/μl, range 5–480/μl), normal glucose and lactate, mildly elevated protein (median 67 mg/dl, range 49–213 mg/dl), intrathecal IgG synthesis and OCB
Microglia activation, perivascular B cell/plasma cell infiltrates, rare T cell infiltrates
Reversible IgG1/3-antibody-mediated NMDA-R crosslinking and internalization, alteration in glutamatergic synaptic transmission and plasticity, no neuronal cell death
Immunotherapy: 1. GCS together with steroid-sparing agents, IVIG, PE/IA, 2. rituximab, cyclophosphamide; tumor therapy: (operation, radiation, chemotherapy)
Monophasic or relapsing-remitting disease course: prolonged recovery over weeks–months (incomplete, spontaneous remission, accelerated and more complete remission under immunotherapy/tumor removal), frequent relapses (about 25 %) in patients without tumor or with insufficient immunotherapy
[15, 18, 20, 44, 47, 72, 100]
Anti-AMPA-R encephalitis
Age 40–80 years (median 60 years), gender female 90 %, ANPR 15
Lymphocytic pleocytosis (median 24/μl; range, 6–75/μl), normal glucose and lactate, normal–mildly elevated protein (median 51 mg/dl, range <46–420 mg/dl), intrathecal IgG synthesis and OCB
Reversible IgG-antibody-mediated AMPA-R crosslinking and internalization, alteration in glutamatergic synaptic transmission and plasticity, no neuronal cell death
Immunotherapy: 1. GCS together with steroid-sparing agents, IVIG, PE/IA, 2. rituximab, cyclophosphamide; Tumor therapy: (operation, radiation, chemotherapy)
Monophasic or relapsing-remitting disease course: good response to immunotherapy/tumor removal, frequent relapses (about 50 %) in patients without tumor or with insufficient immunotherapy
[32, 50, 72]
Anti-GABAB-R encephalitis
Age 25–75 years (median 60 years), gender female 50 %, ANPR 25
Lymphocytic pleocytosis (median 20/μl; range, 0–950/μl), normal glucose and lactate, normal–mildly elevated protein (median 35 mg/dl, range 22–109 mg/dl), intrathecal IgG synthesis and OCB
IgG1-antibody-mediated GABAB-R blockade without internalization, alteration in GABAergic synaptic transmission and plasticity, impairment of pre- and postsynaptic GABAergic inhibition, no neuronal cell death
Immunotherapy: 1. GCS together with steroid-sparing agents, IVIG, PE/IA, 2. rituximab, cyclophosphamide; Tumor therapy: (operation, radiation, chemotherapy)
Monophasic or chronic disease course: good response to immunotherapy/tumor removal, rare relapses
[7, 53, 72]
Anti-Glycine-R encephalitis
Age 30–60 years (median 50 years), gender male 80 %, ANPR 4
CSF normal or lymphocytic pleocytosis (median 33/μl, range 0–60/μl), normal glucose and lactate, normal protein, intrathecal IgG synthesis/OCB
IgG1-antibody-mediated Gly-R blockade, alteration in glycinergic synaptic transmission and plasticity
Immunotherapy: 1. GCS together with AZT/MMF, IVIG, PE/IA, 2. rituximab, cyclophosphamide, (tumor therapy)
Monophasic or relapsing disease course: good response to immunotherapy, occasional relapses
[45, 64, 72]
Anti-VGKC complex encephalitis: LGI1
Age 30–80 years (median 60 years), gender male 65 %, ANPR 120
CSF normal or lymphocytic pleocytosis, normal glucose and lactate, normal or mildly elevated protein (median 35 mg/dl range up to 44 mg/dl), intrathecal IgG synthesis/OCB
IgG4/1-antibody mediated disruption of the presynaptic VGKC complex and altered synaptic transmission
Immunotherapy: 1. GCS together with AZT/MMF, IVIG, PE/IA, 2. rituximab, cyclophosphamide, (tumor therapy)
Monophasic or relapsing disease course: good response to immunotherapy, occasional relapses
[46, 51, 72]
Anti-VGKC complex encephalitis: CASPR2
Age 45–80 years (median 60 years), gender male 85 %, ANPR 40
CSF normal or lymphocytic pleocytosis (median 3/μl, range 0–15/μl), normal glucose and lactate, normal or mildly elevated protein (median 35 mg/dl range up to 44 mg/dl), intrathecal IgG synthesis/OCB
IgG4/1-antibody mediated disruption of the para/juxtanodal VGKC complex and altered neuronal excitability
Immunotherapy: 1. GCS together with AZT/MMF, IVIG, PE/IA, 2. rituximab, cyclophosphamide; (tumor therapy)
Monophasic or relapsing disease course: spontaneous remission, good response to immunotherapy, relapses may occur
[4, 46, 52, 72]
Anti-mGlu-R1 encephalitis
Age 20–50 years, gender female 100 %, ANPR 3
CSF normal or lymphocytic pleocytosis (range 28–190/μl), normal glucose and lactate, normal or mildly elevated protein (range 28–72 mg/dl), intrathecal IgG synthesis/OCB
Purkinje cell loss with amputation of the dendritic tree, astrogliosis, no inflammatory cell infiltrates
IgG-antibody binding to mGlu-R1 at the perisynaptic site of Purkinje cell dendritic spines, impairment of cerebellar synaptic plasticity and motor learning, Purkinje cell death
Immunotherapy: 1. GCS together with AZT/MMF, IVIG, PE/IA, 2. rituximab, cyclophosphamide;
Monophasic or chronic disease course: good response to immunotherapy
[13, 62, 88]
Anti-mGlu-R5 encephalitis
Age 15–45 years, gender female 50 %, ANPR 2
Lymphocytic pleocytosis (range 23–114/μl), normal glucose and lactate, normal or mildly elevated, protein (range 40–55 mg/dl), intrathecal IgG synthesis/OCB
IgG-antibody binding to mGlu-R5 on hippocampal neurons, impairment of synaptic plasticity, learning and memory
Immunotherapy: GCS together with AZT/MMF, IVIG, PE/IA Tumor therapy
Monophasic or chronic disease course: good response to immunotherapy/tumor therapy
[11, 55]
Anti-P/Q type/N-type VGCC encephalitis
Age 30–80 years, gender male 80 %, ANPR 120
CSF normal or lymphocytic pleocytosis, normal glucose and lactate, normal or mildly elevated protein, intrathecal IgG synthesis/OCB
Purkinje cell loss, cerebellar cortical gliosis, rare or no inflammatory infiltrates in the cerebellum
IgG-antibody binding to P/Q type/N-type VGCC on central and peripheral neurons, impairment of neurotransmitter release, neuronal cell death
Immunotherapy: 1. GCS together with steroid-sparing agents, IVIG, PE/IA, 2. rituximab, cyclophosphamide; tumor therapy: (operation, radiation, chemotherapy)
Chronic disease course: limited response to immuno-therapy/tumor therapy
[27, 34, 56, 65]
Anti-nAch-R encephalitis
Age 17–103 years (median 65 years), gender male (55 %), ANRP 150
CSF normal or lymphocytic pleocytosis (range 10–100/μl), normal glucose and lactate, normal or mildly elevated protein (median, 63 mg/dl; range, 48–181 mg/dl), intrathecal IgG synthesis/OCB
IgG-antibody binding to nAch-R on central and peripheral neurons, impairment of synaptic transmission
Immunotherapy: 1. GCS together with steroid-sparing agents, IVIG, PE/IA, 2. rituximab, cyclophosphamide, tumor therapy: (operation, radiation, chemotherapy)
Monophasic or chronic disease course: good response to immuno-therapy/tumor therapy
[3, 31, 66, 95]
AMPA 2-amino-3-(5-methyl-3-oxo-1,2-oxazol-4-yl)propanoic acid, ANPR approximate number of patients reported, AZA azathioprine, CSF cerebrospinal fluid, EEG electroencephalography, EMG electromyography, FLAIR fluid attenuated inversion recovery, GABA γ-aminobutyric acid, GABARAP γ-aminobutyric acid receptor associated protein, GCS glucocorticosteroids, IA immunoadsorption, IVIG intravenous immunoglobulins, mGluR metabotropic glutamate receptor, MMF mycophenolate mofetil, n-Ach nicotinic acetylcholine, NMDA N-methyl-D-aspartate, OCB oligoclonal bands, PE plasma exchange, SIADH syndrome of inappropriate antidiuretic hormone secretion, SCLC small cell lung cancer, VGCC voltage-gated calcium channels, VGKC voltage-gated potassium channels
In principle, depending on the IgG subtype, antibodies may (1) specifically activate or block the function of their target molecules (GABAB-, Glycine- nAch-receptors, VGCC, VGKC), (2) crosslink and internalize the receptors (AMPA- and NMDA-receptors), (3) activate the complement cascade with subsequent formation of the terminal membrane attack complex and target cell lysis (probably mGluR1/5 receptors, VGCC, VGKC) and (4) activate Fc-receptors with subsequent antibody-dependent cell-mediated cytotoxicity (ADCC) mainly by NK cells [23]. However, the effector mechanisms involved in the pathogenic effect of the autoantibodies within the CNS are not yet fully understood. In fact, autoantibodies in anti-NMDA-R and -AMPA-R, GABAB-R encephalitis are of the IgG1 and IgG3 type and are thus capable of activating complement in the presence of patient plasma (containing high concentrations of complement factors). However, in none of the reported autopsy or biopsy studies complement depositions could be detected on neurons suggesting that in the presence of patient CSF (containing low concentrations of complement factors) these IgG 1 and 3 autoantibodies do not lead to relevant complement activation [6, 32, 47, 50, 63, 93]. In contrast, autoantibodies in VGKC-complex encephalitis are predominantly of the IgG4 (and IgG1) type and thus are unable to activate complement in the presence of patient plasma. However, in the only biopsy study reported thus far, complement depositions could be detected on neurons in VGKC-complex encephalitis suggesting that in the presence of patient CSF these IgG4 (or the IgG1) autoantibodies are capable of activating complement [6]. These conflicting results suggest that effector mechanisms of distinct autoantibody classes and subtypes may differ between the CNS and peripheral organs.
Moreover, with the growing clinical awareness of the autoantibodies and improved diagnostic possibilities due to cell-based detection systems more patients are currently identified with persisting IgM antibodies without subsequent class switch to IgG and rarely with IgA antibodies against neuronal plasma membrane antigens [78]. This class-diversity of the neuron-directed adaptive humoral immune response in CNS may indeed reflect different milieus (e.g., absence or presence of different tumors), in which B cell activation takes place. Moreover, lack of class switch might indicate insufficient CD4+ T cell-mediated help in B cell activation. However, these issues need further experimental investigation. Further, it remains to be determined, whether cytotoxicity by CD8+ T cells contributes to functional and structural neuronal impairment.
Inflammatory CNS disorders associated with IgG antibodies against neuronal surface membrane antigens are characterized by a presentation of CNS- and sometimes PNS-related clinical symptoms representing the expression distribution and function of the respective target antigen [54, 97]. On cerebral MRI cortical and subcortical gray matter regions may display mild and often transient T2/FLAIR hyperintense signals. CSF findings may initially be normal, but often include inflammatory changes (lymphocytic pleocytosis, mildly elevated protein, intrathecal IgG synthesis with oligoclonal bands, but normal glucose and lactate levels). The clinical course is usually either monophasic or relapsing-remitting, but rarely progressive and clinical symptoms and paraclinical measures usually display a good response to immunotherapy, especially to antibody-depleting therapies. Upon detection of a tumor, sufficient tumor therapy is crucial to halt the pathogenic immune response [54, 97].
It should be noted that antibody-mediated autoimmunity to non-neuronal ion channels on the cell surface membrane [aquaporin 4 [74] on astrocytes (and neurons), Na(x) channel [38] on ependymal cells, astrocytes, and pituicytes (circumventricular organs)] may occur also in a paraneoplastic context (neuromyelitis optica: breast cancer, Hürthle cell thyroid carcinoma, carcinoid, pituitary somatotropinoma, B cell lymphoma, monoclonal gammopathy [74]; essential hypernatremia [38]: ganglioneuroma).

Conclusions

A growing number of immune-mediated CNS disorders of paraneoplastic and non-paraneoplastic autoimmune origin have recently emerged, in which neurons are the target of both adaptive cellular and humoral immune responses. In autoimmune encephalitis associated with antibodies to neuronal surface membrane antigens, potentially reversible mechanisms of antibody-mediated impairment of synaptic transmission and neuronal excitability prevail. Hence, these disorders offer unique insight and provoke further investigation into the consequences of immune-mediated disruption of distinct neuronal signaling pathways within the living CNS.
In contrast, paraneoplastic autoimmune encephalitis associated with antibodies to intracellular neuronal antigens seems to be mediated by cytotoxic CD8+ T cells that cause functional and structural neuronal impairment in a way not specific for the respective antigen.

Acknowledgments

Scientific and clinical work of the authors is supported by the German research foundation (DFG), the BMBF, the IZKF Münster, the IMF Münster, and industry.

Conflicts of interest

All authors declare no relevant conflicts of interest. N.M. has received honoraria for lecturing and travel expenses for attending meetings from Biogen Idec, GlaxoSmithKline, and Fresenius Medical Care. S.G.M. has received honoraria for lecturing and travel expenses for attending meetings and has received financial research support from Bayer, Biogen Idec, Sanofi-Aventis, Bayer Schering, Novo Nordisk, Merck Serono Novartis, and Teva. H.W. has received funding for travel and speaker honoraria from Bayer Schering Pharma, Biogen Idec/Elan Corporation, Sanofi-Aventis, Merck Serono, and Teva Pharmaceutical Industries Ltd.; has served/serves as a consultant for Merck Serono, Medac, Inc., Sanofi-Aventis/Teva Pharmaceutical Industries Ltd., Biogen Idec, Bayer Schering Pharma, Novartis, and Novo Nordisk, and receives research support from Bayer Schering Pharma, Biogen Idec/Elan Corporation, Sanofi-Aventis, Merck Serono, and Novo Nordisk.

Open Access

This article is distributed under the terms of the Creative Commons Attribution License which permits any use, distribution, and reproduction in any medium, provided the original author(s) and the source are credited.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 2.0 International License (https://​creativecommons.​org/​licenses/​by/​2.​0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

Neuer Inhalt

e.Med Neurologie & Psychiatrie

Kombi-Abonnement

Mit e.Med Neurologie & Psychiatrie erhalten Sie Zugang zu CME-Fortbildungen der Fachgebiete, den Premium-Inhalten der dazugehörigen Fachzeitschriften, inklusive einer gedruckten Zeitschrift Ihrer Wahl.

Weitere Produktempfehlungen anzeigen
Literatur
1.
Zurück zum Zitat Alamowitch S, Graus F, Uchuya M, Rene R, Bescansa E, Delattre JY (1997) Limbic encephalitis and small cell lung cancer. Clinical and immunological features. Brain 120(Pt 6):923–928PubMedCrossRef Alamowitch S, Graus F, Uchuya M, Rene R, Bescansa E, Delattre JY (1997) Limbic encephalitis and small cell lung cancer. Clinical and immunological features. Brain 120(Pt 6):923–928PubMedCrossRef
2.
Zurück zum Zitat Antoine JC, Honnorat J, Camdessanche JP, Magistris M, Absi L, Mosnier JF, Petiot P, Kopp N, Michel D (2001) Paraneoplastic anti-CV2 antibodies react with peripheral nerve and are associated with a mixed axonal and demyelinating peripheral neuropathy. Ann Neurol 49:214–221PubMedCrossRef Antoine JC, Honnorat J, Camdessanche JP, Magistris M, Absi L, Mosnier JF, Petiot P, Kopp N, Michel D (2001) Paraneoplastic anti-CV2 antibodies react with peripheral nerve and are associated with a mixed axonal and demyelinating peripheral neuropathy. Ann Neurol 49:214–221PubMedCrossRef
3.
Zurück zum Zitat Baker SK, Morillo C, Vernino S (2009) Autoimmune autonomic ganglionopathy with late-onset encephalopathy. Auton Neurosci 146:29–32PubMedCrossRef Baker SK, Morillo C, Vernino S (2009) Autoimmune autonomic ganglionopathy with late-onset encephalopathy. Auton Neurosci 146:29–32PubMedCrossRef
4.
Zurück zum Zitat Becker EB, Zuliani L, Pettingill R, Lang B, Waters P, Dulneva A, Sobott F, Wardle M, Graus F, Bataller L, Robertson NP, Vincent A (2012) Contactin-associated protein-2 antibodies in non-paraneoplastic cerebellar ataxia. J Neurol Neurosurg Psychiatry 83:437–440PubMedCrossRef Becker EB, Zuliani L, Pettingill R, Lang B, Waters P, Dulneva A, Sobott F, Wardle M, Graus F, Bataller L, Robertson NP, Vincent A (2012) Contactin-associated protein-2 antibodies in non-paraneoplastic cerebellar ataxia. J Neurol Neurosurg Psychiatry 83:437–440PubMedCrossRef
5.
Zurück zum Zitat Bernal F, Shams’ili S, Rojas I, Sanchez-Valle R, Saiz A, Dalmau J, Honnorat J, Sillevis Smitt P, Graus F (2003) Anti-Tr antibodies as markers of paraneoplastic cerebellar degeneration and Hodgkin’s disease. Neurology 60:230–234PubMedCrossRef Bernal F, Shams’ili S, Rojas I, Sanchez-Valle R, Saiz A, Dalmau J, Honnorat J, Sillevis Smitt P, Graus F (2003) Anti-Tr antibodies as markers of paraneoplastic cerebellar degeneration and Hodgkin’s disease. Neurology 60:230–234PubMedCrossRef
6.
Zurück zum Zitat Bien CG, Vincent A, Barnett MH, Becker AJ, Blumcke I, Graus F, Jellinger KA, Reuss DE, Ribalta T, Schlegel J, Sutton I, Lassmann H, Bauer J (2012) Immunopathology of autoantibody-associated encephalitides: clues for pathogenesis. Brain 135:1622–1638PubMedCrossRef Bien CG, Vincent A, Barnett MH, Becker AJ, Blumcke I, Graus F, Jellinger KA, Reuss DE, Ribalta T, Schlegel J, Sutton I, Lassmann H, Bauer J (2012) Immunopathology of autoantibody-associated encephalitides: clues for pathogenesis. Brain 135:1622–1638PubMedCrossRef
7.
Zurück zum Zitat Boronat A, Sabater L, Saiz A, Dalmau J, Graus F (2011) GABA(B) receptor antibodies in limbic encephalitis and anti-GAD-associated neurologic disorders. Neurology 76:795–800PubMedCrossRef Boronat A, Sabater L, Saiz A, Dalmau J, Graus F (2011) GABA(B) receptor antibodies in limbic encephalitis and anti-GAD-associated neurologic disorders. Neurology 76:795–800PubMedCrossRef
8.
Zurück zum Zitat Briani C, Vitaliani R, Grisold W, Honnorat J, Graus F, Antoine JC, Bertolini G, Giometto B (2011) Spectrum of paraneoplastic disease associated with lymphoma. Neurology 76:705–710PubMedCrossRef Briani C, Vitaliani R, Grisold W, Honnorat J, Graus F, Antoine JC, Bertolini G, Giometto B (2011) Spectrum of paraneoplastic disease associated with lymphoma. Neurology 76:705–710PubMedCrossRef
9.
Zurück zum Zitat Bruylant K, Crols R, Humbel RL, Appel B, De Deyn PP (2006) Probably anti-Tr associated paraneoplastic cerebellar degeneration as initial presentation of a squamous cell carcinoma of the lung. Clin Neurol Neurosurg 108:415–417PubMedCrossRef Bruylant K, Crols R, Humbel RL, Appel B, De Deyn PP (2006) Probably anti-Tr associated paraneoplastic cerebellar degeneration as initial presentation of a squamous cell carcinoma of the lung. Clin Neurol Neurosurg 108:415–417PubMedCrossRef
10.
Zurück zum Zitat Butler MH, Hayashi A, Ohkoshi N, Villmann C, Becker CM, Feng G, De Camilli P, Solimena M (2000) Autoimmunity to gephyrin in stiff-man syndrome. Neuron 26:307–312PubMedCrossRef Butler MH, Hayashi A, Ohkoshi N, Villmann C, Becker CM, Feng G, De Camilli P, Solimena M (2000) Autoimmunity to gephyrin in stiff-man syndrome. Neuron 26:307–312PubMedCrossRef
11.
12.
Zurück zum Zitat Chan KH, Vernino S, Lennon VA (2001) ANNA-3 anti-neuronal nuclear antibody: marker of lung cancer-related autoimmunity. Ann Neurol 50:301–311PubMedCrossRef Chan KH, Vernino S, Lennon VA (2001) ANNA-3 anti-neuronal nuclear antibody: marker of lung cancer-related autoimmunity. Ann Neurol 50:301–311PubMedCrossRef
13.
Zurück zum Zitat Coesmans M, Smitt PA, Linden DJ, Shigemoto R, Hirano T, Yamakawa Y, van Alphen AM, Luo C, van der Geest JN, Kros JM, Gaillard CA, Frens MA, de Zeeuw CI (2003) Mechanisms underlying cerebellar motor deficits due to mGluR1-autoantibodies. Ann Neurol 53:325–336PubMedCrossRef Coesmans M, Smitt PA, Linden DJ, Shigemoto R, Hirano T, Yamakawa Y, van Alphen AM, Luo C, van der Geest JN, Kros JM, Gaillard CA, Frens MA, de Zeeuw CI (2003) Mechanisms underlying cerebellar motor deficits due to mGluR1-autoantibodies. Ann Neurol 53:325–336PubMedCrossRef
14.
Zurück zum Zitat Cross SA, Salomao DR, Parisi JE, Kryzer TJ, Bradley EA, Mines JA, Lam BL, Lennon VA (2003) Paraneoplastic autoimmune optic neuritis with retinitis defined by CRMP-5-IgG. Ann Neurol 54:38–50PubMedCrossRef Cross SA, Salomao DR, Parisi JE, Kryzer TJ, Bradley EA, Mines JA, Lam BL, Lennon VA (2003) Paraneoplastic autoimmune optic neuritis with retinitis defined by CRMP-5-IgG. Ann Neurol 54:38–50PubMedCrossRef
15.
Zurück zum Zitat Dalmau J, Gleichman AJ, Hughes EG, Rossi JE, Peng X, Lai M, Dessain SK, Rosenfeld MR, Balice-Gordon R, Lynch DR (2008) Anti-NMDA-receptor encephalitis: case series and analysis of the effects of antibodies. Lancet Neurol 7:1091–1098PubMedCrossRef Dalmau J, Gleichman AJ, Hughes EG, Rossi JE, Peng X, Lai M, Dessain SK, Rosenfeld MR, Balice-Gordon R, Lynch DR (2008) Anti-NMDA-receptor encephalitis: case series and analysis of the effects of antibodies. Lancet Neurol 7:1091–1098PubMedCrossRef
16.
Zurück zum Zitat Dalmau J, Graus F, Villarejo A, Posner JB, Blumenthal D, Thiessen B, Saiz A, Meneses P, Rosenfeld MR (2004) Clinical analysis of anti-Ma2-associated encephalitis. Brain 127:1831–1844PubMedCrossRef Dalmau J, Graus F, Villarejo A, Posner JB, Blumenthal D, Thiessen B, Saiz A, Meneses P, Rosenfeld MR (2004) Clinical analysis of anti-Ma2-associated encephalitis. Brain 127:1831–1844PubMedCrossRef
17.
Zurück zum Zitat Dalmau J, Gultekin SH, Voltz R, Hoard R, DesChamps T, Balmaceda C, Batchelor T, Gerstner E, Eichen J, Frennier J, Posner JB, Rosenfeld MR (1999) Ma1, a novel neuron- and testis-specific protein, is recognized by the serum of patients with paraneoplastic neurological disorders. Brain 122(Pt 1):27–39PubMedCrossRef Dalmau J, Gultekin SH, Voltz R, Hoard R, DesChamps T, Balmaceda C, Batchelor T, Gerstner E, Eichen J, Frennier J, Posner JB, Rosenfeld MR (1999) Ma1, a novel neuron- and testis-specific protein, is recognized by the serum of patients with paraneoplastic neurological disorders. Brain 122(Pt 1):27–39PubMedCrossRef
18.
Zurück zum Zitat Dalmau J, Lancaster E, Martinez-Hernandez E, Rosenfeld MR, Balice-Gordon R (2011) Clinical experience and laboratory investigations in patients with anti-NMDAR encephalitis. Lancet Neurol 10:63–74PubMedCrossRef Dalmau J, Lancaster E, Martinez-Hernandez E, Rosenfeld MR, Balice-Gordon R (2011) Clinical experience and laboratory investigations in patients with anti-NMDAR encephalitis. Lancet Neurol 10:63–74PubMedCrossRef
19.
20.
Zurück zum Zitat Dalmau J, Tuzun E, Wu HY, Masjuan J, Rossi JE, Voloschin A, Baehring JM, Shimazaki H, Koide R, King D, Mason W, Sansing LH, Dichter MA, Rosenfeld MR, Lynch DR (2007) Paraneoplastic anti-N-methyl-D-aspartate receptor encephalitis associated with ovarian teratoma. Ann Neurol 61:25–36PubMedCrossRef Dalmau J, Tuzun E, Wu HY, Masjuan J, Rossi JE, Voloschin A, Baehring JM, Shimazaki H, Koide R, King D, Mason W, Sansing LH, Dichter MA, Rosenfeld MR, Lynch DR (2007) Paraneoplastic anti-N-methyl-D-aspartate receptor encephalitis associated with ovarian teratoma. Ann Neurol 61:25–36PubMedCrossRef
21.
Zurück zum Zitat De Camilli P, Thomas A, Cofiell R, Folli F, Lichte B, Piccolo G, Meinck HM, Austoni M, Fassetta G, Bottazzo G et al (1993) The synaptic vesicle-associated protein amphiphysin is the 128-kD autoantigen of stiff-man syndrome with breast cancer. J Exp Med 178:2219–2223PubMedCrossRef De Camilli P, Thomas A, Cofiell R, Folli F, Lichte B, Piccolo G, Meinck HM, Austoni M, Fassetta G, Bottazzo G et al (1993) The synaptic vesicle-associated protein amphiphysin is the 128-kD autoantigen of stiff-man syndrome with breast cancer. J Exp Med 178:2219–2223PubMedCrossRef
22.
Zurück zum Zitat de Graaff E, Maat P, Hulsenboom E, van den Berg R, van den Bent M, Demmers J, Lugtenburg PJ, Hoogenraad CC, Sillevis Smitt P (2012) Identification of delta/notch-like epidermal growth factor-related receptor as the Tr antigen in paraneoplastic cerebellar degeneration. Ann Neurol 71(6):815–824 de Graaff E, Maat P, Hulsenboom E, van den Berg R, van den Bent M, Demmers J, Lugtenburg PJ, Hoogenraad CC, Sillevis Smitt P (2012) Identification of delta/notch-like epidermal growth factor-related receptor as the Tr antigen in paraneoplastic cerebellar degeneration. Ann Neurol 71(6):815–824
23.
Zurück zum Zitat Diamond B, Huerta PT, Mina-Osorio P, Kowal C, Volpe BT (2009) Losing your nerves? Maybe it’s the antibodies. Nat Rev Immunol 9:449–456PubMedCrossRef Diamond B, Huerta PT, Mina-Osorio P, Kowal C, Volpe BT (2009) Losing your nerves? Maybe it’s the antibodies. Nat Rev Immunol 9:449–456PubMedCrossRef
24.
Zurück zum Zitat Eckstein C, Saidha S, Levy M (2011) A differential diagnosis of central nervous system demyelination: beyond multiple sclerosis. J Neurol 259(5):801–816 Eckstein C, Saidha S, Levy M (2011) A differential diagnosis of central nervous system demyelination: beyond multiple sclerosis. J Neurol 259(5):801–816
25.
26.
Zurück zum Zitat Folli F, Solimena M, Cofiell R, Austoni M, Tallini G, Fassetta G, Bates D, Cartlidge N, Bottazzo GF, Piccolo G et al (1993) Autoantibodies to a 128-kD synaptic protein in three women with the stiff-man syndrome and breast cancer. N Engl J Med 328:546–551PubMedCrossRef Folli F, Solimena M, Cofiell R, Austoni M, Tallini G, Fassetta G, Bates D, Cartlidge N, Bottazzo GF, Piccolo G et al (1993) Autoantibodies to a 128-kD synaptic protein in three women with the stiff-man syndrome and breast cancer. N Engl J Med 328:546–551PubMedCrossRef
27.
Zurück zum Zitat Fukuda T, Motomura M, Nakao Y, Shiraishi H, Yoshimura T, Iwanaga K, Tsujihata M, Eguchi K (2003) Reduction of P/Q-type calcium channels in the postmortem cerebellum of paraneoplastic cerebellar degeneration with Lambert–Eaton myasthenic syndrome. Ann Neurol 53:21–28PubMedCrossRef Fukuda T, Motomura M, Nakao Y, Shiraishi H, Yoshimura T, Iwanaga K, Tsujihata M, Eguchi K (2003) Reduction of P/Q-type calcium channels in the postmortem cerebellum of paraneoplastic cerebellar degeneration with Lambert–Eaton myasthenic syndrome. Ann Neurol 53:21–28PubMedCrossRef
28.
Zurück zum Zitat Furneaux HM, Rosenblum MK, Dalmau J, Wong E, Woodruff P, Graus F, Posner JB (1990) Selective expression of Purkinje-cell antigens in tumor tissue from patients with paraneoplastic cerebellar degeneration. N Engl J Med 322:1844–1851PubMedCrossRef Furneaux HM, Rosenblum MK, Dalmau J, Wong E, Woodruff P, Graus F, Posner JB (1990) Selective expression of Purkinje-cell antigens in tumor tissue from patients with paraneoplastic cerebellar degeneration. N Engl J Med 322:1844–1851PubMedCrossRef
29.
Zurück zum Zitat Geis C, Weishaupt A, Grunewald B, Wultsch T, Reif A, Gerlach M, Dirkx R, Solimena M, Perani D, Heckmann M, Toyka KV, Folli F, Sommer C (2011) Human stiff-person syndrome IgG induces anxious behavior in rats. PLoS ONE 6:e16775PubMedCrossRef Geis C, Weishaupt A, Grunewald B, Wultsch T, Reif A, Gerlach M, Dirkx R, Solimena M, Perani D, Heckmann M, Toyka KV, Folli F, Sommer C (2011) Human stiff-person syndrome IgG induces anxious behavior in rats. PLoS ONE 6:e16775PubMedCrossRef
30.
Zurück zum Zitat Geis C, Weishaupt A, Hallermann S, Grunewald B, Wessig C, Wultsch T, Reif A, Byts N, Beck M, Jablonka S, Boettger MK, Uceyler N, Fouquet W, Gerlach M, Meinck HM, Siren AL, Sigrist SJ, Toyka KV, Heckmann M, Sommer C (2010) Stiff person syndrome-associated autoantibodies to amphiphysin mediate reduced GABAergic inhibition. Brain 133:3166–3180PubMedCrossRef Geis C, Weishaupt A, Hallermann S, Grunewald B, Wessig C, Wultsch T, Reif A, Byts N, Beck M, Jablonka S, Boettger MK, Uceyler N, Fouquet W, Gerlach M, Meinck HM, Siren AL, Sigrist SJ, Toyka KV, Heckmann M, Sommer C (2010) Stiff person syndrome-associated autoantibodies to amphiphysin mediate reduced GABAergic inhibition. Brain 133:3166–3180PubMedCrossRef
31.
Zurück zum Zitat Gibbons CH, Centi J, Vernino S, Freeman R (2012) Autoimmune autonomic ganglionopathy with reversible cognitive impairment. Arch Neurol 69:461–466PubMedCrossRef Gibbons CH, Centi J, Vernino S, Freeman R (2012) Autoimmune autonomic ganglionopathy with reversible cognitive impairment. Arch Neurol 69:461–466PubMedCrossRef
32.
Zurück zum Zitat Graus F, Boronat A, Xifro X, Boix M, Svigelj V, Garcia A, Palomino A, Sabater L, Alberch J, Saiz A (2010) The expanding clinical profile of anti-AMPA receptor encephalitis. Neurology 74:857–859PubMedCrossRef Graus F, Boronat A, Xifro X, Boix M, Svigelj V, Garcia A, Palomino A, Sabater L, Alberch J, Saiz A (2010) The expanding clinical profile of anti-AMPA receptor encephalitis. Neurology 74:857–859PubMedCrossRef
33.
Zurück zum Zitat Graus F, Keime-Guibert F, Rene R, Benyahia B, Ribalta T, Ascaso C, Escaramis G, Delattre JY (2001) Anti-Hu-associated paraneoplastic encephalomyelitis: analysis of 200 patients. Brain 124:1138–1148PubMedCrossRef Graus F, Keime-Guibert F, Rene R, Benyahia B, Ribalta T, Ascaso C, Escaramis G, Delattre JY (2001) Anti-Hu-associated paraneoplastic encephalomyelitis: analysis of 200 patients. Brain 124:1138–1148PubMedCrossRef
34.
Zurück zum Zitat Graus F, Lang B, Pozo-Rosich P, Saiz A, Casamitjana R, Vincent A (2002) P/Q type calcium-channel antibodies in paraneoplastic cerebellar degeneration with lung cancer. Neurology 59:764–766PubMedCrossRef Graus F, Lang B, Pozo-Rosich P, Saiz A, Casamitjana R, Vincent A (2002) P/Q type calcium-channel antibodies in paraneoplastic cerebellar degeneration with lung cancer. Neurology 59:764–766PubMedCrossRef
35.
Zurück zum Zitat Graus F, Saiz A, Dalmau J (2010) Antibodies and neuronal autoimmune disorders of the CNS. J Neurol 257:509–517PubMedCrossRef Graus F, Saiz A, Dalmau J (2010) Antibodies and neuronal autoimmune disorders of the CNS. J Neurol 257:509–517PubMedCrossRef
36.
Zurück zum Zitat Graus F, Vincent A, Pozo-Rosich P, Sabater L, Saiz A, Lang B, Dalmau J (2005) Anti-glial nuclear antibody: marker of lung cancer-related paraneoplastic neurological syndromes. J Neuroimmunol 165:166–171PubMedCrossRef Graus F, Vincent A, Pozo-Rosich P, Sabater L, Saiz A, Lang B, Dalmau J (2005) Anti-glial nuclear antibody: marker of lung cancer-related paraneoplastic neurological syndromes. J Neuroimmunol 165:166–171PubMedCrossRef
37.
Zurück zum Zitat Gultekin SH, Rosenfeld MR, Voltz R, Eichen J, Posner JB, Dalmau J (2000) Paraneoplastic limbic encephalitis: neurological symptoms, immunological findings and tumour association in 50 patients. Brain 123(Pt 7):1481–1494PubMedCrossRef Gultekin SH, Rosenfeld MR, Voltz R, Eichen J, Posner JB, Dalmau J (2000) Paraneoplastic limbic encephalitis: neurological symptoms, immunological findings and tumour association in 50 patients. Brain 123(Pt 7):1481–1494PubMedCrossRef
38.
Zurück zum Zitat Hiyama TY, Matsuda S, Fujikawa A, Matsumoto M, Watanabe E, Kajiwara H, Niimura F, Noda M (2010) Autoimmunity to the sodium-level sensor in the brain causes essential hypernatremia. Neuron 66:508–522PubMedCrossRef Hiyama TY, Matsuda S, Fujikawa A, Matsumoto M, Watanabe E, Kajiwara H, Niimura F, Noda M (2010) Autoimmunity to the sodium-level sensor in the brain causes essential hypernatremia. Neuron 66:508–522PubMedCrossRef
39.
Zurück zum Zitat Hoffmann LA, Jarius S, Pellkofer HL, Schueller M, Krumbholz M, Koenig F, Johannis W, la Fougere C, Newman T, Vincent A, Voltz R (2008) Anti-Ma and anti-Ta associated paraneoplastic neurological syndromes: 22 newly diagnosed patients and review of previous cases. J Neurol Neurosurg Psychiatry 79:767–773PubMedCrossRef Hoffmann LA, Jarius S, Pellkofer HL, Schueller M, Krumbholz M, Koenig F, Johannis W, la Fougere C, Newman T, Vincent A, Voltz R (2008) Anti-Ma and anti-Ta associated paraneoplastic neurological syndromes: 22 newly diagnosed patients and review of previous cases. J Neurol Neurosurg Psychiatry 79:767–773PubMedCrossRef
40.
Zurück zum Zitat Holmoy T, Skorstad G, Roste LS, Scheie D, Alvik K (2009) Stiff-person syndrome associated with lower motor neuron disease and infiltration of cytotoxic T cells in the spinal cord. Clin Neurol Neurosurg 111:708–712PubMedCrossRef Holmoy T, Skorstad G, Roste LS, Scheie D, Alvik K (2009) Stiff-person syndrome associated with lower motor neuron disease and infiltration of cytotoxic T cells in the spinal cord. Clin Neurol Neurosurg 111:708–712PubMedCrossRef
41.
Zurück zum Zitat Honnorat J, Aguera M, Zalc B, Goujet C, Quach T, Antoine JC, Belin MF (1998) POP66, a paraneoplastic encephalomyelitis-related antigen, is a marker of adult oligodendrocytes. J Neuropathol Exp Neurol 57:311–322PubMedCrossRef Honnorat J, Aguera M, Zalc B, Goujet C, Quach T, Antoine JC, Belin MF (1998) POP66, a paraneoplastic encephalomyelitis-related antigen, is a marker of adult oligodendrocytes. J Neuropathol Exp Neurol 57:311–322PubMedCrossRef
42.
Zurück zum Zitat Honnorat J, Antoine JC, Derrington E, Aguera M, Belin MF (1996) Antibodies to a subpopulation of glial cells and a 66-kDa developmental protein in patients with paraneoplastic neurological syndromes. J Neurol Neurosurg Psychiatry 61:270–278PubMedCrossRef Honnorat J, Antoine JC, Derrington E, Aguera M, Belin MF (1996) Antibodies to a subpopulation of glial cells and a 66-kDa developmental protein in patients with paraneoplastic neurological syndromes. J Neurol Neurosurg Psychiatry 61:270–278PubMedCrossRef
43.
Zurück zum Zitat Honnorat J, Byk T, Kusters I, Aguera M, Ricard D, Rogemond V, Quach T, Aunis D, Sobel A, Mattei MG, Kolattukudy P, Belin MF, Antoine JC (1999) Ulip/CRMP proteins are recognized by autoantibodies in paraneoplastic neurological syndromes. Eur J Neurosci 11:4226–4232PubMedCrossRef Honnorat J, Byk T, Kusters I, Aguera M, Ricard D, Rogemond V, Quach T, Aunis D, Sobel A, Mattei MG, Kolattukudy P, Belin MF, Antoine JC (1999) Ulip/CRMP proteins are recognized by autoantibodies in paraneoplastic neurological syndromes. Eur J Neurosci 11:4226–4232PubMedCrossRef
44.
Zurück zum Zitat Hughes EG, Peng X, Gleichman AJ, Lai M, Zhou L, Tsou R, Parsons TD, Lynch DR, Dalmau J, Balice-Gordon RJ (2010) Cellular and synaptic mechanisms of anti-NMDA receptor encephalitis. J Neurosci 30:5866–5875PubMedCrossRef Hughes EG, Peng X, Gleichman AJ, Lai M, Zhou L, Tsou R, Parsons TD, Lynch DR, Dalmau J, Balice-Gordon RJ (2010) Cellular and synaptic mechanisms of anti-NMDA receptor encephalitis. J Neurosci 30:5866–5875PubMedCrossRef
45.
Zurück zum Zitat Hutchinson M, Waters P, McHugh J, Gorman G, O’Riordan S, Connolly S, Hager H, Yu P, Becker CM, Vincent A (2008) Progressive encephalomyelitis, rigidity, and myoclonus: a novel glycine receptor antibody. Neurology 71:1291–1292PubMedCrossRef Hutchinson M, Waters P, McHugh J, Gorman G, O’Riordan S, Connolly S, Hager H, Yu P, Becker CM, Vincent A (2008) Progressive encephalomyelitis, rigidity, and myoclonus: a novel glycine receptor antibody. Neurology 71:1291–1292PubMedCrossRef
46.
Zurück zum Zitat Irani SR, Alexander S, Waters P, Kleopa KA, Pettingill P, Zuliani L, Peles E, Buckley C, Lang B, Vincent A (2010) 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 133:2734–2748PubMedCrossRef Irani SR, Alexander S, Waters P, Kleopa KA, Pettingill P, Zuliani L, Peles E, Buckley C, Lang B, Vincent A (2010) 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 133:2734–2748PubMedCrossRef
47.
Zurück zum Zitat Irani SR, Bera K, Waters P, Zuliani L, Maxwell S, Zandi MS, Friese MA, Galea I, Kullmann DM, Beeson D, Lang B, Bien CG, Vincent A (2010) N-methyl-D-aspartate antibody encephalitis: temporal progression of clinical and paraclinical observations in a predominantly non-paraneoplastic disorder of both sexes. Brain 133:1655–1667PubMedCrossRef Irani SR, Bera K, Waters P, Zuliani L, Maxwell S, Zandi MS, Friese MA, Galea I, Kullmann DM, Beeson D, Lang B, Bien CG, Vincent A (2010) N-methyl-D-aspartate antibody encephalitis: temporal progression of clinical and paraclinical observations in a predominantly non-paraneoplastic disorder of both sexes. Brain 133:1655–1667PubMedCrossRef
48.
Zurück zum Zitat Janeway CA, Travers P, Walport M, Shlomchik MJ (2001) Immunobiology: the immune system in health and disease. vol 5. Garland Science, New York Janeway CA, Travers P, Walport M, Shlomchik MJ (2001) Immunobiology: the immune system in health and disease. vol 5. Garland Science, New York
49.
Zurück zum Zitat Keime-Guibert F, Graus F, Broet P, Rene R, Molinuevo JL, Ascaso C, Delattre JY (1999) Clinical outcome of patients with anti-Hu-associated encephalomyelitis after treatment of the tumor. Neurology 53:1719–1723PubMedCrossRef Keime-Guibert F, Graus F, Broet P, Rene R, Molinuevo JL, Ascaso C, Delattre JY (1999) Clinical outcome of patients with anti-Hu-associated encephalomyelitis after treatment of the tumor. Neurology 53:1719–1723PubMedCrossRef
50.
Zurück zum Zitat Lai M, Hughes EG, Peng X, Zhou L, Gleichman AJ, Shu H, Mata S, Kremens D, Vitaliani R, Geschwind MD, Bataller L, Kalb RG, Davis R, Graus F, Lynch DR, Balice-Gordon R, Dalmau J (2009) AMPA receptor antibodies in limbic encephalitis alter synaptic receptor location. Ann Neurol 65:424–434PubMedCrossRef Lai M, Hughes EG, Peng X, Zhou L, Gleichman AJ, Shu H, Mata S, Kremens D, Vitaliani R, Geschwind MD, Bataller L, Kalb RG, Davis R, Graus F, Lynch DR, Balice-Gordon R, Dalmau J (2009) AMPA receptor antibodies in limbic encephalitis alter synaptic receptor location. Ann Neurol 65:424–434PubMedCrossRef
51.
Zurück zum Zitat Lai M, Huijbers MG, Lancaster E, Graus F, Bataller L, Balice-Gordon R, Cowell JK, Dalmau J (2010) Investigation of LGI1 as the antigen in limbic encephalitis previously attributed to potassium channels: a case series. Lancet Neurol 9:776–785PubMedCrossRef Lai M, Huijbers MG, Lancaster E, Graus F, Bataller L, Balice-Gordon R, Cowell JK, Dalmau J (2010) Investigation of LGI1 as the antigen in limbic encephalitis previously attributed to potassium channels: a case series. Lancet Neurol 9:776–785PubMedCrossRef
52.
Zurück zum Zitat Lancaster E, Huijbers MG, Bar V, Boronat A, Wong A, Martinez-Hernandez E, Wilson C, Jacobs D, Lai M, Walker RW, Graus F, Bataller L, Illa I, Markx S, Strauss KA, Peles E, Scherer SS, Dalmau J (2011) Investigations of caspr2, an autoantigen of encephalitis and neuromyotonia. Ann Neurol 69:303–311PubMedCrossRef Lancaster E, Huijbers MG, Bar V, Boronat A, Wong A, Martinez-Hernandez E, Wilson C, Jacobs D, Lai M, Walker RW, Graus F, Bataller L, Illa I, Markx S, Strauss KA, Peles E, Scherer SS, Dalmau J (2011) Investigations of caspr2, an autoantigen of encephalitis and neuromyotonia. Ann Neurol 69:303–311PubMedCrossRef
53.
Zurück zum Zitat Lancaster E, Lai M, Peng X, Hughes E, Constantinescu R, Raizer J, Friedman D, Skeen MB, Grisold W, Kimura A, Ohta K, Iizuka T, Guzman M, Graus F, Moss SJ, Balice-Gordon R, Dalmau J (2010) Antibodies to the GABA(B) receptor in limbic encephalitis with seizures: case series and characterisation of the antigen. Lancet Neurol 9:67–76PubMedCrossRef Lancaster E, Lai M, Peng X, Hughes E, Constantinescu R, Raizer J, Friedman D, Skeen MB, Grisold W, Kimura A, Ohta K, Iizuka T, Guzman M, Graus F, Moss SJ, Balice-Gordon R, Dalmau J (2010) Antibodies to the GABA(B) receptor in limbic encephalitis with seizures: case series and characterisation of the antigen. Lancet Neurol 9:67–76PubMedCrossRef
54.
Zurück zum Zitat Lancaster E, Martinez-Hernandez E, Dalmau J (2011) Encephalitis and antibodies to synaptic and neuronal cell surface proteins. Neurology 77:179–189PubMedCrossRef Lancaster E, Martinez-Hernandez E, Dalmau J (2011) Encephalitis and antibodies to synaptic and neuronal cell surface proteins. Neurology 77:179–189PubMedCrossRef
55.
Zurück zum Zitat Lancaster E, Martinez-Hernandez E, Titulaer MJ, Boulos M, Weaver S, Antoine JC, Liebers E, Kornblum C, Bien CG, Honnorat J, Wong S, Xu J, Contractor A, Balice-Gordon R, Dalmau J (2011) Antibodies to metabotropic glutamate receptor 5 in the Ophelia syndrome. Neurology 77:1698–1701PubMedCrossRef Lancaster E, Martinez-Hernandez E, Titulaer MJ, Boulos M, Weaver S, Antoine JC, Liebers E, Kornblum C, Bien CG, Honnorat J, Wong S, Xu J, Contractor A, Balice-Gordon R, Dalmau J (2011) Antibodies to metabotropic glutamate receptor 5 in the Ophelia syndrome. Neurology 77:1698–1701PubMedCrossRef
56.
Zurück zum Zitat Lennon VA, Kryzer TJ, Griesmann GE, O’Suilleabhain PE, Windebank AJ, Woppmann A, Miljanich GP, Lambert EH (1995) Calcium-channel antibodies in the Lambert–Eaton syndrome and other paraneoplastic syndromes. N Engl J Med 332:1467–1474PubMedCrossRef Lennon VA, Kryzer TJ, Griesmann GE, O’Suilleabhain PE, Windebank AJ, Woppmann A, Miljanich GP, Lambert EH (1995) Calcium-channel antibodies in the Lambert–Eaton syndrome and other paraneoplastic syndromes. N Engl J Med 332:1467–1474PubMedCrossRef
57.
Zurück zum Zitat Levy LM, Levy-Reis I, Fujii M, Dalakas MC (2005) Brain gamma-aminobutyric acid changes in stiff-person syndrome. Arch Neurol 62:970–974PubMedCrossRef Levy LM, Levy-Reis I, Fujii M, Dalakas MC (2005) Brain gamma-aminobutyric acid changes in stiff-person syndrome. Arch Neurol 62:970–974PubMedCrossRef
58.
Zurück zum Zitat Lucchinetti CF, Kimmel DW, Lennon VA (1998) Paraneoplastic and oncologic profiles of patients seropositive for type 1 antineuronal nuclear autoantibodies. Neurology 50:652–657PubMedCrossRef Lucchinetti CF, Kimmel DW, Lennon VA (1998) Paraneoplastic and oncologic profiles of patients seropositive for type 1 antineuronal nuclear autoantibodies. Neurology 50:652–657PubMedCrossRef
59.
Zurück zum Zitat Luque FA, Furneaux HM, Ferziger R, Rosenblum MK, Wray SH, Schold SC Jr, Glantz MJ, Jaeckle KA, Biran H, Lesser M et al (1991) Anti-Ri: an antibody associated with paraneoplastic opsoclonus and breast cancer. Ann Neurol 29:241–251PubMedCrossRef Luque FA, Furneaux HM, Ferziger R, Rosenblum MK, Wray SH, Schold SC Jr, Glantz MJ, Jaeckle KA, Biran H, Lesser M et al (1991) Anti-Ri: an antibody associated with paraneoplastic opsoclonus and breast cancer. Ann Neurol 29:241–251PubMedCrossRef
60.
Zurück zum Zitat Malter MP, Helmstaedter C, Urbach H, Vincent A, Bien CG (2010) Antibodies to glutamic acid decarboxylase define a form of limbic encephalitis. Ann Neurol 67:470–478PubMedCrossRef Malter MP, Helmstaedter C, Urbach H, Vincent A, Bien CG (2010) Antibodies to glutamic acid decarboxylase define a form of limbic encephalitis. Ann Neurol 67:470–478PubMedCrossRef
61.
Zurück zum Zitat Manto MU, Laute MA, Aguera M, Rogemond V, Pandolfo M, Honnorat J (2007) Effects of anti-glutamic acid decarboxylase antibodies associated with neurological diseases. Ann Neurol 61:544–551PubMedCrossRef Manto MU, Laute MA, Aguera M, Rogemond V, Pandolfo M, Honnorat J (2007) Effects of anti-glutamic acid decarboxylase antibodies associated with neurological diseases. Ann Neurol 61:544–551PubMedCrossRef
62.
Zurück zum Zitat Marignier R, Chenevier F, Rogemond V, Sillevis Smitt P, Renoux C, Cavillon G, Androdias G, Vukusic S, Graus F, Honnorat J, Confavreux C (2010) Metabotropic glutamate receptor type 1 autoantibody-associated cerebellitis: a primary autoimmune disease? Arch Neurol 67:627–630PubMedCrossRef Marignier R, Chenevier F, Rogemond V, Sillevis Smitt P, Renoux C, Cavillon G, Androdias G, Vukusic S, Graus F, Honnorat J, Confavreux C (2010) Metabotropic glutamate receptor type 1 autoantibody-associated cerebellitis: a primary autoimmune disease? Arch Neurol 67:627–630PubMedCrossRef
63.
Zurück zum Zitat Martinez-Hernandez E, Horvath J, Shiloh-Malawsky Y, Sangha N, Martinez-Lage M, Dalmau J (2011) Analysis of complement and plasma cells in the brain of patients with anti-NMDAR encephalitis. Neurology 77:589–593PubMedCrossRef Martinez-Hernandez E, Horvath J, Shiloh-Malawsky Y, Sangha N, Martinez-Lage M, Dalmau J (2011) Analysis of complement and plasma cells in the brain of patients with anti-NMDAR encephalitis. Neurology 77:589–593PubMedCrossRef
64.
Zurück zum Zitat Mas N, Saiz A, Leite MI, Waters P, Baron M, Castano D, Sabater L, Vincent A, Graus F (2011) Antiglycine-receptor encephalomyelitis with rigidity. J Neurol Neurosurg Psychiatry 82:1399–1401PubMedCrossRef Mas N, Saiz A, Leite MI, Waters P, Baron M, Castano D, Sabater L, Vincent A, Graus F (2011) Antiglycine-receptor encephalomyelitis with rigidity. J Neurol Neurosurg Psychiatry 82:1399–1401PubMedCrossRef
65.
Zurück zum Zitat Mason WP, Graus F, Lang B, Honnorat J, Delattre JY, Valldeoriola F, Antoine JC, Rosenblum MK, Rosenfeld MR, Newsom-Davis J, Posner JB, Dalmau J (1997) Small-cell lung cancer, paraneoplastic cerebellar degeneration and the Lambert–Eaton myasthenic syndrome. Brain 120(Pt 8):1279–1300PubMedCrossRef Mason WP, Graus F, Lang B, Honnorat J, Delattre JY, Valldeoriola F, Antoine JC, Rosenblum MK, Rosenfeld MR, Newsom-Davis J, Posner JB, Dalmau J (1997) Small-cell lung cancer, paraneoplastic cerebellar degeneration and the Lambert–Eaton myasthenic syndrome. Brain 120(Pt 8):1279–1300PubMedCrossRef
66.
Zurück zum Zitat McKeon A, Lennon VA, Lachance DH, Fealey RD, Pittock SJ (2009) Ganglionic acetylcholine receptor autoantibody: oncological, neurological, and serological accompaniments. Arch Neurol 66:735–741PubMedCrossRef McKeon A, Lennon VA, Lachance DH, Fealey RD, Pittock SJ (2009) Ganglionic acetylcholine receptor autoantibody: oncological, neurological, and serological accompaniments. Arch Neurol 66:735–741PubMedCrossRef
67.
Zurück zum Zitat McKeon A, Pittock SJ (2011) Paraneoplastic encephalomyelopathies: pathology and mechanisms. Acta Neuropathol 122:381–400PubMedCrossRef McKeon A, Pittock SJ (2011) Paraneoplastic encephalomyelopathies: pathology and mechanisms. Acta Neuropathol 122:381–400PubMedCrossRef
68.
Zurück zum Zitat McKeon A, Tracy JA, Pittock SJ, Parisi JE, Klein CJ, Lennon VA (2011) Purkinje cell cytoplasmic autoantibody type 1 accompaniments: the cerebellum and beyond. Arch Neurol 68:1282–1289PubMedCrossRef McKeon A, Tracy JA, Pittock SJ, Parisi JE, Klein CJ, Lennon VA (2011) Purkinje cell cytoplasmic autoantibody type 1 accompaniments: the cerebellum and beyond. Arch Neurol 68:1282–1289PubMedCrossRef
69.
Zurück zum Zitat Melzer N, Meuth SG, Wiendl H (2009) CD8 + T cells and neuronal damage: direct and collateral mechanisms of cytotoxicity and impaired electrical excitability. FASEB J 23:3659–3673PubMedCrossRef Melzer N, Meuth SG, Wiendl H (2009) CD8 + T cells and neuronal damage: direct and collateral mechanisms of cytotoxicity and impaired electrical excitability. FASEB J 23:3659–3673PubMedCrossRef
70.
Zurück zum Zitat Melzer N, Meuth SG, Wiendl H (2012) Neuron-directed autoimmunity in the central nervous system: entities, mechanisms, diagnostic clues, and therapeutic options. Curr Opin Neurol 25:341–348PubMedCrossRef Melzer N, Meuth SG, Wiendl H (2012) Neuron-directed autoimmunity in the central nervous system: entities, mechanisms, diagnostic clues, and therapeutic options. Curr Opin Neurol 25:341–348PubMedCrossRef
71.
Zurück zum Zitat Meuth SG, Herrmann AM, Simon OJ, Siffrin V, Melzer N, Bittner S, Meuth P, Langer HF, Hallermann S, Boldakowa N, Herz J, Munsch T, Landgraf P, Aktas O, Heckmann M, Lessmann V, Budde T, Kieseier BC, Zipp F, Wiendl H (2009) Cytotoxic CD8 + T cell-neuron interactions: perforin-dependent electrical silencing precedes but is not causally linked to neuronal cell death. J Neurosci 29:15397–15409PubMedCrossRef Meuth SG, Herrmann AM, Simon OJ, Siffrin V, Melzer N, Bittner S, Meuth P, Langer HF, Hallermann S, Boldakowa N, Herz J, Munsch T, Landgraf P, Aktas O, Heckmann M, Lessmann V, Budde T, Kieseier BC, Zipp F, Wiendl H (2009) Cytotoxic CD8 + T cell-neuron interactions: perforin-dependent electrical silencing precedes but is not causally linked to neuronal cell death. J Neurosci 29:15397–15409PubMedCrossRef
72.
Zurück zum Zitat Moscato EH, Jain A, Peng X, Hughes EG, Dalmau J, Balice-Gordon RJ (2010) Mechanisms underlying autoimmune synaptic encephalitis leading to disorders of memory, behavior and cognition: insights from molecular, cellular and synaptic studies. Eur J Neurosci 32:298–309PubMedCrossRef Moscato EH, Jain A, Peng X, Hughes EG, Dalmau J, Balice-Gordon RJ (2010) Mechanisms underlying autoimmune synaptic encephalitis leading to disorders of memory, behavior and cognition: insights from molecular, cellular and synaptic studies. Eur J Neurosci 32:298–309PubMedCrossRef
73.
Zurück zum Zitat Pedemonte E, Mancardi G, Giunti D, Corcione A, Benvenuto F, Pistoia V, Uccelli A (2006) Mechanisms of the adaptive immune response inside the central nervous system during inflammatory and autoimmune diseases. Pharmacol Ther 111:555–566PubMedCrossRef Pedemonte E, Mancardi G, Giunti D, Corcione A, Benvenuto F, Pistoia V, Uccelli A (2006) Mechanisms of the adaptive immune response inside the central nervous system during inflammatory and autoimmune diseases. Pharmacol Ther 111:555–566PubMedCrossRef
74.
Zurück zum Zitat Pittock SJ, Lennon VA (2008) Aquaporin-4 autoantibodies in a paraneoplastic context. Arch Neurol 65:629–632PubMedCrossRef Pittock SJ, Lennon VA (2008) Aquaporin-4 autoantibodies in a paraneoplastic context. Arch Neurol 65:629–632PubMedCrossRef
75.
Zurück zum Zitat Pittock SJ, Lucchinetti CF, Lennon VA (2003) Anti-neuronal nuclear autoantibody type 2: paraneoplastic accompaniments. Ann Neurol 53:580–587PubMedCrossRef Pittock SJ, Lucchinetti CF, Lennon VA (2003) Anti-neuronal nuclear autoantibody type 2: paraneoplastic accompaniments. Ann Neurol 53:580–587PubMedCrossRef
76.
Zurück zum Zitat Pittock SJ, Lucchinetti CF, Parisi JE, Benarroch EE, Mokri B, Stephan CL, Kim KK, Kilimann MW, Lennon VA (2005) Amphiphysin autoimmunity: paraneoplastic accompaniments. Ann Neurol 58:96–107PubMedCrossRef Pittock SJ, Lucchinetti CF, Parisi JE, Benarroch EE, Mokri B, Stephan CL, Kim KK, Kilimann MW, Lennon VA (2005) Amphiphysin autoimmunity: paraneoplastic accompaniments. Ann Neurol 58:96–107PubMedCrossRef
77.
Zurück zum Zitat Pittock SJ, Yoshikawa H, Ahlskog JE, Tisch SH, Benarroch EE, Kryzer TJ, Lennon VA (2006) Glutamic acid decarboxylase autoimmunity with brainstem, extrapyramidal, and spinal cord dysfunction. Mayo Clin Proc 81:1207–1214PubMedCrossRef Pittock SJ, Yoshikawa H, Ahlskog JE, Tisch SH, Benarroch EE, Kryzer TJ, Lennon VA (2006) Glutamic acid decarboxylase autoimmunity with brainstem, extrapyramidal, and spinal cord dysfunction. Mayo Clin Proc 81:1207–1214PubMedCrossRef
78.
Zurück zum Zitat Pruss H, Holtje M, Maier N, Gomez A, Buchert R, Harms L, Ahnert-Hilger G, Schmitz D, Terborg C, Kopp U, Klingbeil C, Probst C, Kohler S, Schwab JM, Stoecker W, Dalmau J, Wandinger KP (2012) IgA NMDA receptor antibodies are markers of synaptic immunity in slow cognitive impairment. Neurology 78:1743–1753PubMedCrossRef Pruss H, Holtje M, Maier N, Gomez A, Buchert R, Harms L, Ahnert-Hilger G, Schmitz D, Terborg C, Kopp U, Klingbeil C, Probst C, Kohler S, Schwab JM, Stoecker W, Dalmau J, Wandinger KP (2012) IgA NMDA receptor antibodies are markers of synaptic immunity in slow cognitive impairment. Neurology 78:1743–1753PubMedCrossRef
79.
Zurück zum Zitat Psimaras D, Carpentier AF, Rossi C (2010) Cerebrospinal fluid study in paraneoplastic syndromes. J Neurol Neurosurg Psychiatry 81:42–45PubMedCrossRef Psimaras D, Carpentier AF, Rossi C (2010) Cerebrospinal fluid study in paraneoplastic syndromes. J Neurol Neurosurg Psychiatry 81:42–45PubMedCrossRef
80.
Zurück zum Zitat Raju R, Rakocevic G, Chen Z, Hoehn G, Semino-Mora C, Shi W, Olsen R, Dalakas MC (2006) Autoimmunity to GABAA-receptor-associated protein in stiff-person syndrome. Brain 129:3270–3276PubMedCrossRef Raju R, Rakocevic G, Chen Z, Hoehn G, Semino-Mora C, Shi W, Olsen R, Dalakas MC (2006) Autoimmunity to GABAA-receptor-associated protein in stiff-person syndrome. Brain 129:3270–3276PubMedCrossRef
81.
Zurück zum Zitat Rojas I, Graus F, Keime-Guibert F, Rene R, Delattre JY, Ramon JM, Dalmau J, Posner JB (2000) Long-term clinical outcome of paraneoplastic cerebellar degeneration and anti-Yo antibodies. Neurology 55:713–715PubMedCrossRef Rojas I, Graus F, Keime-Guibert F, Rene R, Delattre JY, Ramon JM, Dalmau J, Posner JB (2000) Long-term clinical outcome of paraneoplastic cerebellar degeneration and anti-Yo antibodies. Neurology 55:713–715PubMedCrossRef
82.
Zurück zum Zitat Rosenfeld MR, Eichen JG, Wade DF, Posner JB, Dalmau J (2001) Molecular and clinical diversity in paraneoplastic immunity to Ma proteins. Ann Neurol 50:339–348PubMedCrossRef Rosenfeld MR, Eichen JG, Wade DF, Posner JB, Dalmau J (2001) Molecular and clinical diversity in paraneoplastic immunity to Ma proteins. Ann Neurol 50:339–348PubMedCrossRef
83.
Zurück zum Zitat Sadeghian H, Vernino S (2010) Progress in the management of paraneoplastic neurological disorders. Ther Adv Neurol Disord 3:43–52PubMedCrossRef Sadeghian H, Vernino S (2010) Progress in the management of paraneoplastic neurological disorders. Ther Adv Neurol Disord 3:43–52PubMedCrossRef
84.
Zurück zum Zitat Saiz A, Blanco Y, Sabater L, Gonzalez F, Bataller L, Casamitjana R, Ramio-Torrenta L, Graus F (2008) Spectrum of neurological syndromes associated with glutamic acid decarboxylase antibodies: diagnostic clues for this association. Brain 131:2553–2563PubMedCrossRef Saiz A, Blanco Y, Sabater L, Gonzalez F, Bataller L, Casamitjana R, Ramio-Torrenta L, Graus F (2008) Spectrum of neurological syndromes associated with glutamic acid decarboxylase antibodies: diagnostic clues for this association. Brain 131:2553–2563PubMedCrossRef
85.
Zurück zum Zitat Shams’ili S, Grefkens J, de Leeuw B, van den Bent M, Hooijkaas H, van der Holt B, Vecht C, Sillevis Smitt P (2003) Paraneoplastic cerebellar degeneration associated with antineuronal antibodies: analysis of 50 patients. Brain 126:1409–1418PubMedCrossRef Shams’ili S, Grefkens J, de Leeuw B, van den Bent M, Hooijkaas H, van der Holt B, Vecht C, Sillevis Smitt P (2003) Paraneoplastic cerebellar degeneration associated with antineuronal antibodies: analysis of 50 patients. Brain 126:1409–1418PubMedCrossRef
86.
Zurück zum Zitat Shrikant PA, Rao R, Li Q, Kesterson J, Eppolito C, Mischo A, Singhal P (2010) Regulating functional cell fates in CD8 T cells. Immunol Res 46:12–22PubMedCrossRef Shrikant PA, Rao R, Li Q, Kesterson J, Eppolito C, Mischo A, Singhal P (2010) Regulating functional cell fates in CD8 T cells. Immunol Res 46:12–22PubMedCrossRef
87.
Zurück zum Zitat Sillevis Smitt P, Grefkens J, de Leeuw B, van den Bent M, van Putten W, Hooijkaas H, Vecht C (2002) Survival and outcome in 73 anti-Hu-positive patients with paraneoplastic encephalomyelitis/sensory neuronopathy. J Neurol 249:745–753PubMedCrossRef Sillevis Smitt P, Grefkens J, de Leeuw B, van den Bent M, van Putten W, Hooijkaas H, Vecht C (2002) Survival and outcome in 73 anti-Hu-positive patients with paraneoplastic encephalomyelitis/sensory neuronopathy. J Neurol 249:745–753PubMedCrossRef
88.
Zurück zum Zitat Sillevis Smitt P, Kinoshita A, De Leeuw B, Moll W, Coesmans M, Jaarsma D, Henzen-Logmans S, Vecht C, De Zeeuw C, Sekiyama N, Nakanishi S, Shigemoto R (2000) Paraneoplastic cerebellar ataxia due to autoantibodies against a glutamate receptor. N Engl J Med 342:21–27PubMedCrossRef Sillevis Smitt P, Kinoshita A, De Leeuw B, Moll W, Coesmans M, Jaarsma D, Henzen-Logmans S, Vecht C, De Zeeuw C, Sekiyama N, Nakanishi S, Shigemoto R (2000) Paraneoplastic cerebellar ataxia due to autoantibodies against a glutamate receptor. N Engl J Med 342:21–27PubMedCrossRef
89.
Zurück zum Zitat Solimena M, Folli F, Aparisi R, Pozza G, De Camilli P (1990) Autoantibodies to GABA-ergic neurons and pancreatic beta cells in stiff-man syndrome. N Engl J Med 322:1555–1560PubMedCrossRef Solimena M, Folli F, Aparisi R, Pozza G, De Camilli P (1990) Autoantibodies to GABA-ergic neurons and pancreatic beta cells in stiff-man syndrome. N Engl J Med 322:1555–1560PubMedCrossRef
90.
Zurück zum Zitat Solimena M, Folli F, Denis-Donini S, Comi GC, Pozza G, De Camilli P, Vicari AM (1988) Autoantibodies to glutamic acid decarboxylase in a patient with stiff-man syndrome, epilepsy, and type I diabetes mellitus. N Engl J Med 318:1012–1020PubMedCrossRef Solimena M, Folli F, Denis-Donini S, Comi GC, Pozza G, De Camilli P, Vicari AM (1988) Autoantibodies to glutamic acid decarboxylase in a patient with stiff-man syndrome, epilepsy, and type I diabetes mellitus. N Engl J Med 318:1012–1020PubMedCrossRef
91.
Zurück zum Zitat Sommer C, Weishaupt A, Brinkhoff J, Biko L, Wessig C, Gold R, Toyka KV (2005) Paraneoplastic stiff-person syndrome: passive transfer to rats by means of IgG antibodies to amphiphysin. Lancet 365:1406–1411PubMedCrossRef Sommer C, Weishaupt A, Brinkhoff J, Biko L, Wessig C, Gold R, Toyka KV (2005) Paraneoplastic stiff-person syndrome: passive transfer to rats by means of IgG antibodies to amphiphysin. Lancet 365:1406–1411PubMedCrossRef
92.
Zurück zum Zitat Tschernatsch M, Gross O, Kneifel N, Kaps M, Blaes F (2009) SOX-1 autoantibodies in patients with paraneoplastic neurological syndromes. Autoimmun Rev 8:549–551PubMedCrossRef Tschernatsch M, Gross O, Kneifel N, Kaps M, Blaes F (2009) SOX-1 autoantibodies in patients with paraneoplastic neurological syndromes. Autoimmun Rev 8:549–551PubMedCrossRef
93.
Zurück zum Zitat Tuzun E, Zhou L, Baehring JM, Bannykh S, Rosenfeld MR, Dalmau J (2009) Evidence for antibody-mediated pathogenesis in anti-NMDAR encephalitis associated with ovarian teratoma. Acta Neuropathol 118:737–743PubMedCrossRef Tuzun E, Zhou L, Baehring JM, Bannykh S, Rosenfeld MR, Dalmau J (2009) Evidence for antibody-mediated pathogenesis in anti-NMDAR encephalitis associated with ovarian teratoma. Acta Neuropathol 118:737–743PubMedCrossRef
94.
Zurück zum Zitat Vernino S, Lennon VA (2000) New Purkinje cell antibody (PCA-2): marker of lung cancer-related neurological autoimmunity. Ann Neurol 47:297–305PubMedCrossRef Vernino S, Lennon VA (2000) New Purkinje cell antibody (PCA-2): marker of lung cancer-related neurological autoimmunity. Ann Neurol 47:297–305PubMedCrossRef
95.
Zurück zum Zitat Vernino S, Low PA, Fealey RD, Stewart JD, Farrugia G, Lennon VA (2000) Autoantibodies to ganglionic acetylcholine receptors in autoimmune autonomic neuropathies. N Engl J Med 343:847–855PubMedCrossRef Vernino S, Low PA, Fealey RD, Stewart JD, Farrugia G, Lennon VA (2000) Autoantibodies to ganglionic acetylcholine receptors in autoimmune autonomic neuropathies. N Engl J Med 343:847–855PubMedCrossRef
96.
Zurück zum Zitat Vincent A (2010) Successful ‘passive transfer’ of paraneoplastic stiff person syndrome with antibodies to an intracellular antigen. Brain 133:3164–3165PubMedCrossRef Vincent A (2010) Successful ‘passive transfer’ of paraneoplastic stiff person syndrome with antibodies to an intracellular antigen. Brain 133:3164–3165PubMedCrossRef
97.
Zurück zum Zitat Vincent A, Bien CG, Irani SR, Waters P (2011) Autoantibodies associated with diseases of the CNS: new developments and future challenges. Lancet Neurol 10:759–772PubMedCrossRef Vincent A, Bien CG, Irani SR, Waters P (2011) Autoantibodies associated with diseases of the CNS: new developments and future challenges. Lancet Neurol 10:759–772PubMedCrossRef
98.
Zurück zum Zitat Voltz R, Gultekin SH, Rosenfeld MR, Gerstner E, Eichen J, Posner JB, Dalmau J (1999) A serologic marker of paraneoplastic limbic and brain-stem encephalitis in patients with testicular cancer. N Engl J Med 340:1788–1795PubMedCrossRef Voltz R, Gultekin SH, Rosenfeld MR, Gerstner E, Eichen J, Posner JB, Dalmau J (1999) A serologic marker of paraneoplastic limbic and brain-stem encephalitis in patients with testicular cancer. N Engl J Med 340:1788–1795PubMedCrossRef
99.
Zurück zum Zitat Yu Z, Kryzer TJ, Griesmann GE, Kim K, Benarroch EE, Lennon VA (2001) CRMP-5 neuronal autoantibody: marker of lung cancer and thymoma-related autoimmunity. Ann Neurol 49:146–154PubMedCrossRef Yu Z, Kryzer TJ, Griesmann GE, Kim K, Benarroch EE, Lennon VA (2001) CRMP-5 neuronal autoantibody: marker of lung cancer and thymoma-related autoimmunity. Ann Neurol 49:146–154PubMedCrossRef
100.
Zurück zum Zitat Zhang Q, Tanaka K, Sun P, Nakata M, Yamamoto R, Sakimura K, Matsui M, Kato N (2012) Suppression of synaptic plasticity by cerebrospinal fluid from anti-NMDA receptor encephalitis patients. Neurobiol Dis 45:610–615PubMedCrossRef Zhang Q, Tanaka K, Sun P, Nakata M, Yamamoto R, Sakimura K, Matsui M, Kato N (2012) Suppression of synaptic plasticity by cerebrospinal fluid from anti-NMDA receptor encephalitis patients. Neurobiol Dis 45:610–615PubMedCrossRef
Metadaten
Titel
Paraneoplastic and non-paraneoplastic autoimmunity to neurons in the central nervous system
verfasst von
Nico Melzer
Sven G. Meuth
Heinz Wiendl
Publikationsdatum
01.05.2013
Verlag
Springer-Verlag
Erschienen in
Journal of Neurology / Ausgabe 5/2013
Print ISSN: 0340-5354
Elektronische ISSN: 1432-1459
DOI
https://doi.org/10.1007/s00415-012-6657-5

Weitere Artikel der Ausgabe 5/2013

Journal of Neurology 5/2013 Zur Ausgabe

Leitlinien kompakt für die Neurologie

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Stuhltransfusion könnte Fortschreiten von Parkinson-Symptomen bremsen

03.05.2024 Parkinson-Krankheit Nachrichten

Kann eine frühzeitige Stuhltransplantation das Fortschreiten von Parkinson-Symptomen verlangsamen? Die Ergebnisse einer randomisierten Phase-2-Studie scheinen dafür zu sprechen.

Frühe Tranexamsäure-Therapie nützt wenig bei Hirnblutungen

02.05.2024 Hirnblutung Nachrichten

Erhalten Personen mit einer spontanen Hirnblutung innerhalb von zwei Stunden nach Symptombeginn eine Tranexamsäure-Therapie, kann dies weder die Hämatomexpansion eindämmen noch die Mortalität senken.

Sind Frauen die fähigeren Ärzte?

30.04.2024 Gendermedizin Nachrichten

Patienten, die von Ärztinnen behandelt werden, dürfen offenbar auf bessere Therapieergebnisse hoffen als Patienten von Ärzten. Besonders scheint das auf weibliche Kranke zuzutreffen, wie eine Studie zeigt.

Akuter Schwindel: Wann lohnt sich eine MRT?

28.04.2024 Schwindel Nachrichten

Akuter Schwindel stellt oft eine diagnostische Herausforderung dar. Wie nützlich dabei eine MRT ist, hat eine Studie aus Finnland untersucht. Immerhin einer von sechs Patienten wurde mit akutem ischämischem Schlaganfall diagnostiziert.

Update Neurologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.