key: cord-1016661-85iyfpg0 authors: Yamakawa, Mai; Hogan, Keenan O.; Leever, John; Jassam, Yasir N. title: Autopsy Case of Meningoencephalomyelitis Associated With Glial Fibrillary Acidic Protein Antibody date: 2021-10-12 journal: Neurol Neuroimmunol Neuroinflamm DOI: 10.1212/nxi.0000000000001081 sha: 702ea6896dd15a27d537fe8eb26d172c9b91bd7b doc_id: 1016661 cord_uid: 85iyfpg0 BACKGROUND AND OBJECTIVES: To describe the autopsy findings and neuropathologic evaluation of autoimmune meningoencephalomyelitis associated with glial fibrillary acidic protein (GFAP) antibody. METHODS: We reviewed the clinical course, imaging, laboratory, and autopsy findings of a patient with autoimmune meningoencephalomyelitis associated with GFAP antibody who had a refractory course to multiple immunosuppressive therapies. RESULTS: The patient was a 70-year-old man who was diagnosed as GFAP antibody-associated autoimmune meningoencephalomyelitis. MRI of the head showed linear perivascular enhancement in the midbrain and the basal ganglia. Despite treatment with high-dose corticosteroids, plasma exchange, IV immunoglobulins, and cyclophosphamide, he died with devastating neurologic complications. Autopsy revealed a coexistent neuroendocrine tumor in the small intestine and diffuse inflammation in the brain parenchyma, perivascular spaces, and leptomeninges, with predominant T-cells, macrophages, and activated microglia. B-cells and plasma cells were absent. There was no astrocyte involvement with change in GFAP immunostaining. DISCUSSION: This case illustrates autoimmune meningoencephalomyelitis associated with GFAP antibody in the CSF and coexistent neuroendocrine tumor. The autopsy findings were nonspecific and did not demonstrate astrocyte involvement. Further accumulation of cases is warranted to delineate the utility and pathogenic significance of the GFAP autoantibody. Initially, serotonin syndrome from concurrent bupropion and citalopram use was suspected, and cyproheptadine was started. However, he became lethargic and febrile (39.1°C) on day 2 of admission, requiring endotracheal intubation. CSF analysis on day 2 showed lymphocytic leukocytosis (nucleated cells 120/mm 3 ) and elevated protein (167 mg/dL). Abbreviated hospital course is shown in Figure 1 . Hyponatremia was corrected with fluid resuscitation and cessation of citalopram not requiring prolonged fluid restriction. MRI of the head showed linear symmetric perivascular enhancement in bilateral crus cerebri and basal ganglia that were not present 10 days before admission ( Figure 2 , A-C), as well as thin subdural fluid collections in the posterior convexity concerning for subdural empyema and meningitis ( Figure 2D ). Magnetic resonance angiogram of the head was normal. MRI of the thoracic spine on day 6 showed long segment thoracic cord signal abnormalities with a possible enhancement ( Figure 2F ). Extensive infectious and rheumatological assessments were negative. SARS-CoV-2 polymerase chain reaction was not performed because this presentation occurred before the pandemic. The patient was started on IV methylprednisolone 1 g/day for 5 days with a protracted taper. Findings on the MRI of the head and T-spine continued to improve (Figure 2 , D and E). Inflammation in CSF peaked on day 14 with 660/ mm 3 lymphocytes, with 5 oligoclonal bands, and IgG synthesis rate of 21.22. The patient underwent 3 sessions of plasma exchange. On day 24, the autoimmune encephalitis CSF panel came back positive for glial fibrillary acidic protein (GFAP) antibody, and a diagnosis of GFAP antibody-associated meningoencephalomyelitis was made. Anti-NMDA receptor antibody was negative. IV immunoglobulins were given, followed by cyclophosphamide 500 mg/m 2 . Whole-body CT, PET-CT, and scrotal ultrasound were negative for malignancy. Despite the improvement in CSF and MRI findings, the patient continued to have severe myoclonus, requiring continuous sedation and 3 antiseizure drugs; EEG developed bifrontal epileptic discharges while on these medications. The patient was palliatively extubated and died on day 47. His family agreed to proceed with an autopsy. The autopsy revealed acute pneumonia and undiagnosed well-differentiated neuroendocrine tumor (WDNET) in the small intestine; this was negative for GFAP immunohistochemistry. Gross examination of the brain demonstrated diffuse mild leptomeningeal fibrosis over the convexities with scattered arachnoid granulations. Sectioning revealed severe edema of the cerebral hemispheres with enlarged gyri, narrow sulci, and central herniation affecting the midbrain. A thorough microscopic examination revealed variable degrees of inflammation involving the entire brain except for the cerebellum. The inflammatory infiltrates were perivascular with extension into the parenchyma ( Figure 3A ). There was no evidence of demyelination or loss of GFAP stain, nor Glossary AQP-4 = aquaporin 4; CLIPPERS = chronic lymphocytic inflammation with pontine perivascular enhancement responsive to steroids; GFAP = glial fibrillary acidic protein; WDNET = well-differentiated neuroendocrine tumor. fragmented or macrophage-engulfed astrocytes; GFAP stain showed focal moderate cortical gliosis and some subpial gliosis. Aquaporin 4 (AQP-4) stain was not performed. There was no vasculitis or necrosis. The inflammatory cells were a mixture of CD4 + and CD8 + T lymphocytes and macrophages ( Figure 3 , B and C). No B lymphocytes or plasma cells were identified. CD68 immunostain showed prominent microglial activation and macrophages throughout the cortex and white matter, as well as highlighting perivascular and leptomeningeal infiltrates ( Figure 3D ). There were diffuse severe acute hypoxic-ischemic leukoencephalopathy and severe edema throughout the cortices. There were no Lewy bodies, inclusions, senile plaques, or neurofibrillary tangles as demonstrated by α-synuclein, β-amyloid, and Tau immunostains. The spinal cord was not examined. This was a case report and no IRB approval was needed. The consent for autopsy was obtained from the patient's wife. Abbreviations: CBA = cell-based assay; GFAP = glial fibrillary acidic protein; IFA = immunofluorescent assay; IVIG = IV immunoglobulin. All the data appear in the article. This article reports a case of autoimmune meningoencephalomyelitis with a positive GFAP antibody with an autopsy and complete neuropathologic evaluation of the whole brain. Autoimmune GFAP astrocytopathy defined by GFAP IgG positivity in the CSF is an emerging disease entity first described in 2016 as angiography-negative, corticosteroidresponsive subacute meningoencephalomyelitis with CSF lymphocytic pleocytosis. 1,2 Neuropathologic evaluation of the condition is limited to brain and meningeal biopsies of 5 patients to date, as summarized in Table 1 . [3] [4] [5] One case series from China showed astrocytopathy with a loss or decrease of GFAP and AQP-4 stain with GFAP antibody in CSF, notably with concurrent autoantibodies such as p-ANCA, antiendothelial cell, anti-MOG, antinuclear, anti-SSA, and anti-Ro-52 antibodies in 3 of 4 cases. 3 In our case, AQP-4 immunostaining or antemortem serum testing was not performed; however, there was no signs of astrocyte involvement with GFAP immunostaining, including loss or decrease of GFAP, fragmentation, or phagocytosis of the astrocytes. Our case goes against the causal pathogenicity to astrocyte decay of the GFAP antibody in CSF, in contrast to the well-documented pathogenicity of AQP-4 antibody in neuromyelitis optica spectrum disorders resulting in astrocytopathy. [6] [7] [8] [9] The neuropathologic finding of our case was relatively nonspecific. Differential diagnoses of the neuropathologic findings include chronic lymphocytic inflammation with pontine perivascular enhancement responsive to steroids (CLIPPERS) and anti-NMDA encephalitis. In CLIPPERS, the mainstay of inflammatory cells is CD4 T-cells, indicating possible major histocompatibility complex class II-restricted antigen presentation or allergic reaction, along with microglia, histiocytes, and B cells. Vascular damage with necrosis, fibrin deposition astrocytic fragmentation, neuronophagia, and focal demyelination have been seen. 10 Our case with inflammation in gray and white matter of whole brain contradicts CLIPPERS, which has the predilection to the white matter of the hindbrain. As for NMDA encephalitis, the hallmark of pathology is perivascular B-cell cuffing and scattered T cells in the parenchyma, which were not seen in our autopsy. Our case possibly was paraneoplastic autoimmune encephalitis with the GFAP-negative WDNET; this could infer that the GFAP antibody in the CSF was a byproduct of the ongoing inflammation, and rather not a causative cross-reacting autoantibody induced by the concurrent neoplasm. Periventricular perivascular enhancement is perceived as a classic finding of autoimmune astrocytopathy seen in half of cases; however, it is unknown if this directly indicates astrocyte involvement. 2, 3, 11 In our case, the perivascular enhancement in the midbrain and basal ganglia on MRI did not correlate with astrocyte involvement in the autopsy. Collectively, because positive GFAP antibody in CSF or periventricular perivascular enhancement on MRI does not confirm astrocytes involvement in the inflammation, the term "astrocytopathy" should be used carefully until the pathologic demonstration. Nevertheless, we believe that identifying associated autoantibodies would help to guide care of otherwise indistinguishable autoimmune encephalitides, possibly augmenting cancer screening in select cases. Further accumulation of cases is needed to better define this emerging disease entity. We have important limitations in our report. First, this was a postmortem study after a prolonged disease course, and the cardiopulmonary compromise shortly before death likely affected the pathology with hypoxic-ischemic changes. The extensive immunotherapies also likely altered the pathology findings, evidenced by antemortem oligoclonal bands in the CSF without postmortem B and plasma cells. Finally, we did not assess the spinal cord or stain for AQP-4. GFAP antibody is implicated in autoimmune meningoencephalitis; however, there is limited evidence that this is the causative antibody provoking downstream inflammation with astrocytes. Our case with CSF-positive GFAP antibody did not have astrocytic involvement in the autopsy, suggesting that GFAP antibody was a bystander autoantibody with the inflammation. The causality of GFAP antibody has to be investigated with more pathologic evaluations of similar cases. Autoimmune glial fibrillary acidic protein astrocytopathy: a novel meningoencephalomyelitis Glial fibrillary acidic protein immunoglobulin G as biomarker of autoimmune astrocytopathy: analysis of 102 patients Autoimmune glial fibrillary acidic protein astrocytopathy in Chinese patients: a retrospective study Brain immunohistopathology in a patient with autoimmune glial fibrillary acidic protein astrocytopathy Clinical and immunological characteristics of the spectrum of GFAP autoimmunity: a case series of 22 patients Astrocytic damage is far more severe than demyelination in NMO: a clinical CSF biomarker study Consequences of NMO-IgG binding to aquaporin-4 in neuromyelitis optica Loss of aquaporin 4 in lesions of neuromyelitis optica: distinction from multiple sclerosis Molecular outcomes of neuromyelitis optica (NMO)-IgG binding to aquaporin-4 in astrocytes CLIPPERS: chronic lymphocytic inflammation with pontine perivascular enhancement responsive to steroids. Review of an increasingly recognized entity within the spectrum of inflammatory central nervous system disorders Autoimmune glial fibrillary acidic protein astrocytopathy: a review of the literature The authors would like to thank the medical intensive care unit team and the neurology consult service, especially Dr. Mohammed Nashatizadeh, Dr. Brennen Bittel, and Dr. Brenton Massey for the compassionate continuity of care, and express gratitude for the family's dedication, through the contribution of their loved one, to the progress of science in neurology, neuroimmunology, and neuropathology. Dr. Yasir N. Jassam's current affiliation is Pickup Family Neuroscience Institute, Hoag Memorial Presbyterian Hospital, Newport Beach, CA, USA. The authors report no targeted funding.Disclosure Y.N. Jassam: Janssen Pharmaceuticals speakers bureau, unrelated to this article. The other authors report no disclosures relevant to the manuscript. Go to Neurology.org/NN for full disclosures. Drafting/revision of the manuscript for content, including medical writing for content, and analysis or interpretation of data