Demyelinating Disorders


OVERVIEW OF CHAPTER

Multiple sclerosis (MS) is the single most common demyelinating disorder and appears to be increasing in incidence and prevalence globally, even in traditionally low prevalence regions of the world. As such, when clinical and neuroimaging features suggest a demyelinating disorder in either adult or pediatric patients, the main differential diagnosis usually starts with confirming, or excluding, MS. This chapter details the features of MS in adult and pediatric patients and briefly describes other demyelinating disorders that might enter the differential diagnosis for each patient group.

The topics are discussed in the following order:

  • Clinical features of MS

  • Acute/tumefactive demyelinating lesions prompting biopsy: often but not exclusively MS

  • Acute disseminated encephalomyelitis (ADEM)

  • Acute hemorrhagic leukoencephalitis (AHLE)

  • Myelin oligodendrocyte glycoprotein (MOG) antibody disease (MOG-Ab, MOGAD)

  • Neuromyelitis optica (NMO)

  • Pathological features of MS at intermediate and late time periods/autopsy

  • Dysmyelinating disorders

MULTIPLE SCLEROSIS

Clinical Features

Multiple sclerosis is a chronic, immune-mediated disorder for which the exact cause is unknown; it is not the result of direct viral infection. The highest incidence is in females, and this sex skew appears to be increasing. Onset of multiple sclerosis usually occurs in young adulthood, between 20 and 40 years of age with a global median prevalence of 33 cases of MS per 100,000 people, with significant differences between different countries. Due to increasing longevity of persons with MS, the prevalence and overall age of the MS population are also increasing.

While some patients are mildly affected, MS remains one of the most frequent causes of acquired disability in young persons. The clinical disease course of MS usually evolves over decades. The three types of established disease are relapsing-remitting, secondary progressive, and primary progressive, with some also considering clinically isolated syndrome (CIS) as an initial type of MS. The vast majority of MS cases are either relapsing-remitting or primary progressive, with 85 % of patients having the former type. Relapsing-remitting disease includes episodes of neurological dysfunction lasting at least 24 hours in the absence of fever or infection; episodes are distributed over time (two or more sets of symptoms 30 + days apart) and space (two separate locations in the brain/spinal cord) without other identifiable cause. The disease may be the typical relapsing-remitting type on initial presentation, but the majority of patients eventually enter a secondary progressive stage with few relapses, but cumulative deficits over time. With advanced disease, brain atrophy often occurs.

Older statistics suggested a grim overall long-term prognosis for the disease, but today, many patients are 20 + years into their disease before the use of a cane is necessary and a larger percentage of benign MS is being diagnosed. Clinical parameters associated with more aggressive disease course, either singly or in combination, include male sex, older age at onset (> 35 or > 40 years), severe relapses, frequency of relapses in the first 5 years, short interval between relapses, early accrual of disability, and Expanded Disability Status Scale (EDSS) ≥3 in the first year after disease evolution. About 7 % of MS patients meet the criteria for aggressive disease, defined as patients who reached an EDSS of 6 within 5 years, those with EDSS ≥6 at the age of 40 years, and those who entered secondary progressive MS within 3 years after relapsing MS disease onset.

Zeydan and Kantarci noted that “transition from the relapsing-remitting phase to the progressive phase in MS often happens in the fifth decade. In MS, structural central nervous system reserve decreases with aging and MS-associated mechanisms. While clinical and subclinical disease activity decreases with aging, the post-relapse recovery potential decreases with aging as well. Moreover, the efficacy of disease-modifying treatments declines with older age. Aging emerges as the ultimate target for prevention of progressive disease course, which is the most important determinant of disability worsening in MS.”

Early in the disease course, isolated lesions may be encountered in a patient that require time to pass before the full spectrum of disease is recognized and before confident diagnosis of MS, a disease defined as distributed in time and space, can be rendered. CIS represents the first attack that is clinically recognized. Radiologically isolated syndrome (RIS) is found in individuals undergoing magnetic resonance imaging (MRI) for non-MS symptoms who show a typical MS lesion on neuroimaging. RIS is a possible diagnosis because very typical neuroimaging features occur in most cases of MS and they can be distinguished from other demyelinating disorders, especially in later disease stages; thus, descriptions of these neuroimaging features are provided below for both early and intermediate/later stages of MS.

Clinical signs and symptoms of MS are varied. A prodrome is increasingly being recognized, but this consists of a relatively nonspecific constellation of signs and symptoms. No single feature is specific for the prodrome of MS. However, most studies agree that there is an increased use of the medical/mental health systems in the 5 years prior to the first diagnosed demyelinating event in patients with MS.

Paresthesias (numbness and tingling), monocular loss of vision, gait problems, weakness, double vision, urinary urgency or frequency, and constipation are typical symptoms. Cognitive dysfunction and fatigue are not uncommon, even in the relapsing stages of disease. Seizures and hearing loss are rare. The EDSS developed by Krutzke in 1983 is the most widely utilized tool used to assess the extent of cumulative neurological compromise. Scores include 0 for normal, 2.0 for minimal disability, 4.0 for increased limitation in walking ability, 5.0 where disability affects daily routine, 6.0 for need for walking assistance, 7.0 for restriction to wheelchair, 9.0 when confined to bed, and 10.0 for demise.

Thus, the clinician making the diagnosis of MS relies on the constellation of clinical signs and symptoms described above, coupled with the neuroimaging features of MS, to make the diagnosis. No single laboratory test is definitive for MS, unlike the case with two other demyelinating disorders discussed in this chapter: positive serum serology for aquaporin 4 antibody (APQ4-Ab) in the case of NMO and positive serum myelin oligodendrocyte antibody in the case of MOG antibody disease (MOGAD). By definition, MS shows negative serum APQ4-Ab and negative serum MOG-Ab. Most MS patients will show the presence of oligoclonal bands (OCBs) in their cerebrospinal fluid (CSF) that represent immunoglobulins produced intrathecally; by definition, to be positive for OCBs, the patient should show two or more unique bands in their CSF that are not present in their serum. These unique immunoglobulins represent the unique production of proteins by inflammatory cells within the central nervous system (CNS). Test methods require collecting CSF by lumbar puncture and analyzing CSF by protein electrophoresis on agarose gel with Coomassie blue staining or an isoelectric focusing/silver staining procedure. Serum must be obtained for comparison.

Clinical diagnosis is challenging, and neurologists have devised a number of algorithms over the years, the history of which is reviewed by Poser and Brinar. In 2001, neuroimaging with MRI resulted in the McDonald criteria, which have been revised several times since, culminating in the 2017 revision. Even with multiple revisions of the McDonald criteria for MS, a significant number of patients continue to receive erroneous clinical diagnoses of MS. This is summarized best by Sand:

“The McDonald criteria for the diagnosis of multiple sclerosis (MS), first introduced in 2001 with revisions in 2005, 2010, and 2017, continue to evolve. The recently published 2017 revisions may facilitate earlier fulfillment of the diagnostic criteria for relapsing-remitting MS. However, confirming a diagnosis of MS is not always straightforward. Clinical heterogeneity along with a lengthy differential diagnosis results in the not infrequent incorrect assignment of a diagnosis of MS. While MS is a common disease, affecting around 900,000 persons in the United States, the astute clinician needs to be aware of alternative diagnoses, such as functional neurologic disorders, migraine, and vascular disease, along with uncommon inflammatory, infectious, and metabolic disorders that may mimic MS. Studies have indicated that, of all new referrals to MS subspecialty centers with a question of MS diagnosis, 30 %–67 % were ultimately determined not to have MS. Regrettably, misdiagnosis and initiation of MS disease-modifying therapy had already occurred in some of these patients. A multicenter case series consisting of patients who had been incorrectly diagnosed with MS11 revealed that over 50 % carried the misdiagnosis for at least 3 years, and more than 5 % were misdiagnosed for over 20 years. In this study, 31 % incurred unnecessary morbidity as a direct result of misdiagnosis. The primary reasons for MS misdiagnosis included inappropriate application of McDonald criteria in syndromes atypical for demyelination or lacking objective clinical findings consistent with MS, and the misinterpretation of or overreliance on MRI abnormalities in the setting of nonspecific neurologic symptoms.”

Thus, the pathologist, neurologist, and neurosurgeon should be aware that the diagnosis of MS might be unclear at various time points during the course of a patient's disease. Contrasting features of MS with other demyelinating disorders discussed in this chapter are provided in Tables 5.1 and 5.2 .

Table 5.1
Clinical comparison of MS, ADEM, NMO, and MOG
MS ADEM NMO MOG
Sex ratio (F:M) 2:1 Slight male predominance 9:1 2:1 to near equal
Median age (years) at onset 20–30 5–8 35–45 31–37
Coexisting autoimmune disease Rare No Frequent Rare
Diagnostic serum serology No specific test No specific test NMO-IgG (AQP4-Ab+) MOG-Ab+
Relapsing-remitting course Yes No No No
Progressive disease course Yes No No No
CSF oligoclonal bands Yes, 85% Rare (<2%) <30% <10%
CSF cell counts >50 µ/L <5% Nl in 42%–72%; counts >50 should prompt search for alternate diagnosis <30% 50%
CSF glial fibrillary acidic protein detectable No No Yes No
Optic nerve involvement Unilateral (<2% bilateral) Occasional Bilateral 8%–14% at onset of all ON
Preferentially chiasm
Bilateral 35%–41% at onset of all ON, often intraorbital nerves involved, sparing chiasm, enhancement of optic nerve sheath, involvement of orbital soft tissue
Pattern of spinal cord involvement Short segment of cord, lateral, enhancing Seen in up to 1/3 of patients, often large confluent lesions extending over multiple segments, sometimes associated with cord swelling LETM in 85%–94%, cord often swollen, centrally enhancing 2/3 LETM, 1/3 short, central, cord usually not swollen
Optic neuritis at onset 15%–20% May present with optic neuritis 45% 60%–74%
Myelitis at onset Rare Uncommon 47% 18%–23%
Brainstem encephalitis at onset Rare May present with dysarthria, oculomotor disturbances 3% 8%–14%
Supratentorial lesions at onset Very high Often 50% 35%
Brain lesions Periventricular, perpendicular to ventricles, abutting CSF spaces Reversible, ill-defined white matter lesions of brain, spinal cord, frequent involvement of thalami and basal ganglia; compared to MS are periventricular sparing, no periventricular ovoid lesions perpendicular to ventricular edge (Dawson fingers) as in MS; lesion margins indistinct Along areas enriched for aquaporin 4=leptomeningeal, ependymal enhancement, area postrema Brainstem, deep cerebellar peduncle especially characteristic
Adapted from Lucchinetti, ACTRIMS 2020, Teaching Course, Weber et al. Ther Adv Neurol Dis 2018;11:1-15, Pohl et al. Neurology. 2016 30;87(9 Suppl 2):S38-45.
ADEM , acute disseminated encephalomyelitis; CSF , cerebrospinal fluid; LETM , longitudinally extensive transverse myelitis; MOG , myelin oligodendrocyte glycoprotein; MS , multiple sclerosis; NMO , neuromyelitis optica; ON , optic neuritis.

Table 5.2
Histological comparison of MS, ADEM, NMO, and MOG
MS ADEM NMO MOG
Pattern of demyelination Confluent Perivascular, coalescence may occur Non-demyelinating lesions early with relative myelin and axon preservation compared to loss of AQP4 in early lesions
Later lesions cystic/cavitating with myelin loss
Perivenular, coalescent, confluent
Predominant sites of lesions Periventricular flame-shaped lesions perpendicular to ventricles, undersurface corpus callosum, brainstem and spinal cord lesions abutting CSF spaces and cerebral and cerebellar cortex Subcortical and central white matter and cortical gray-white matter junction, thalami, basal ganglia, cerebellum, and brainstem Sites with highest APQ4 concentration Deep gray matter, brainstem, cerebellar peduncles
Cortical demyelination Yes Yes No Yes
Type of cortical demyelination Subpial, leukocortical > intracortical Subpial, intracortical
May show cortical microglial activation unlike MS
None Intracortical > leukocortical
Lesion inflammation Predominantly
CD8 > CD4 T cells, highly variable B cells, granulocytes rare
T and B cells, occasional plasma cells, granulocytes Granulocytes, eosinophils typical
Few T cells
Predominantly T cells early, some granulocytes
CD4 >> CD8 T cells
Astrocytes Preserved, reactive, hypertrophic Preserved, reactive, hypertrophic Lost/lysed, dystrophic short blunted cell processes Preserved, reactive, hypertrophic
Aquaporin 4 IHC loss No No Yes No
Creutzfeldt-Peter astrocytes Present Present Absent Present
Axons Axonal swelling in acute lesions, loss of myelin > axons May show acute axonal injury, spheroid Better preservation in early lesions but may show severe loss (cystic change) in later lesions Axonal spheroids described
Smoldering plaques Present in secondary progressive disease Absent Absent Absent
Adapted from Luccinetti, ACTRIMS 2020, Teaching Course, Pohl et al. Neurology. 2016 30;87(9 Suppl 2):S38-45.
ADEM , acute disseminated encephalomyelitis; AQP , aquaporin; MOG , myelin oligodendrocyte glycoprotein; MS , multiple sclerosis; NMO , neuromyelitis optica.

MULTIPLE SCLEROSIS—FACT SHEET

Definition

  • n

    Acquired, usually progressive, chronic CNS inflammatory disorder causing disseminated demyelinating lesions in space and time, associated with myelin > axonal destruction

  • n

    Unknown cause: immune-genetic disorder, multifactorial genetic and environmental associations

Incidence and Location

  • n

    Highest incidence in Caucasians

  • n

    Incidence increases with distance from the equator

  • n

    High incidence areas: United Kingdom, Iceland, Canada, Australia, United States

  • n

    Immigrants adopt risk of new country if immigrating at age <15 years but retain risk for country of origin if immigrating at >15 years

  • n

    12,000 new cases/year in United States

  • n

    US prevalence >900,000

  • n

    Demyelination and axonal injury confined to CNS

Sex and Age Distribution

  • n

    60%–75% female

  • n

    75% present between 15 and 45 years of age

  • n

    5% have onset prior to age 18

  • n

    Prevalence highest in patients 55–64 years of age

  • n

    Genetic risk factors: mainly determined by major histocompatibility complex especially HLA DR2

  • n

    Significant role for environmental risk factors in pathogenesis

  • n

    Environmental risk factors: Epstein–Barr virus infection, cigarette smoking, low levels of vitamin D, increased body mass index during adolescence

  • n

    Increased risk in first-degree relative X 15–25 fold (2%–3%)

  • n

    Higher risk in monozygotic twins (33%) versus dizygotic (3%–5%)

MULTIPLE SCLEROSIS—FACT SHEET—cont'd

Clinical Features

  • n

    Relapsing-remitting type (RRMS): 85% of cases

  • n

    Primary progressive type (PPMS): 15% of cases; progressive disease from the onset, without true relapses

  • n

    Secondary progressive type (SPMS): conversion to progressive disease in majority of RRMS patients

  • n

    Many patients have CIS, such as optic neuritis or transverse myelitis at onset, i.e., at first attack

  • n

    CSF usually shows a mild elevation in protein and mononuclear cells, but glucose is always normal

  • n

    85% of cases show OCBs directed towards an unknown antigen: by definition >2 OCBs (clones of immunoglobulins in CSF, not seen in blood) and elevated immunoglobulin G (IgG) index, especially after two or more clinical attacks

  • n

    No specific serum serological test to diagnose MS, unlike MOGAD or NMO

  • n

    Diagnosis involves the use of the McDonald criteria, which incorporate distribution of disease in space and time, based on clinical and neuroimaging features

  • n

    Gray matter involvement increasingly appreciated as important substrate for functional clinical deficits

  • n

    Prodrome increasingly recognized, including increased physician/psychiatric visits in years prior to diagnosis, fibromyalgia, pain, headaches, urological visits, irritable bowel symptoms, and sleep disturbances

Radiologic Features

  • n

    Periventricular, infratentorial, juxtacortical, and cortical white matter T2-hyperintense lesions

  • n

    Central vein sign

  • n

    Supratentorial ovoid-shaped lesions often perpendicular to long axis of the lateral ventricle

  • n

    Progression of lesion burden and atrophy over time

  • n

    Brain atrophy in progressive phases of disease

  • n

    Optic chiasm lesions usually focal, often unilateral, in contrast to MOGAD, which is often bilateral and NMO, which is often chiasmal

  • n

    MS spinal cord lesions often shorter, more often peripherally enhancing, not central in contrast to MOGAD and NMO

  • n

    T1-weighted images often show “black holes” representing focal areas of more severe injury

  • n

    Band-like subpial cortical demyelination detectable on immunostaining of tissue is poorly detected on neuroimaging

  • n

    Gray matter involvement difficult to detect by conventional neuroimaging techniques

  • n

    RIS refers to individuals scanned for non-MS symptoms but typical lesions are found on MRI

Prognosis and Treatment

  • n

    Prognosis variable

  • n

    10% to 15% have benign MS with rare attacks and little accumulated disability

  • n

    5% of patients have rapidly progressive disease

  • n

    Symptomatic treatment for fatigue, spasticity, urinary and sexual dysfunction, pain, gait disturbance, depression, other symptoms

  • n

    Other therapies directed at reducing inflammation: Betaseron (interferon β-1b), Avonex (interferon β-1a), Copaxone (glatiramer acetate), and Rebif (interferon β-1a), Tysabri (natalizumab), and Novantrone (mitoxantrone)

Neurologists and primary physicians encounter patients with MS at multiple time points and all stages of the disease; however, pathologists are seldom involved with making/confirming the diagnosis of MS in patients until the time of demise, i.e., at autopsy. Most, but not all, patients who come to autopsy have met the McDonald criteria for a high likelihood of diagnosis of MS, but usually the diagnosis can be easily confirmed at autopsy due to the near-pathognomonic distribution of lesions and the sharp demarcation of many/most of the demyelinating lesions from surrounding white matter. These features are discussed below in detail since they usually allow for distinction from other rare demyelinating disorders at the time of autopsy.

During early, acute, active phases of the disease, most patients do not come to the attention of either pathologists or neurosurgeons, and biopsy plays no role in the routine diagnosis of MS patients. In other words, the pathologist and neurosurgeon are not involved in the diagnosis of the vast majority of MS patients.

Rarely, a patient will present with or develop acute onset of symptoms during the course of known MS that will prompt neuroimaging work-up. When neuroimaging studies show large enhancing lesions or otherwise unusual features, the clinician may be worried about alternative diagnoses to MS, or even a superimposed second diagnosis such as infection or tumor. Acute/tumefactive demyelinating disease is the label applied to such lesions. Although most examples of acute tumefactive/demyelinating disease prompting biopsy prove ultimately to be the first presentation of MS (i.e., CIS, after sufficient clinical follow-up), this is not true of all examples. Thus, the features of acute/tumefactive demyelinating lesions prompting biopsy are discussed below.

ACUTE/TUMEFACTIVE DEMYELINATING DISEASE PROMPTING BIOPSY

MS plaques usually appear on neuroimaging studies as multiple small, well-demarcated lesions without mass effect, and given their classic locations, multiplicity, and perpendicular orientation to the lateral ventricles or spinal cord lesional features, usually fulfill radiographic criteria for MS diagnosis. Thus, patients whose MRI results are typical do not usually undergo biopsy. However, occasionally patients with or without these typical MS neuroimaging features may present with symptoms that lead to neuroimaging studies showing a solitary large lesion greater than 2 cm in size or a lesion with extraordinary mass effect, edema, or ring enhancement. These unusual, large, mass-producing lesions, especially in isolation, can be mistaken on neuroimaging studies for a brain tumor (tumefactive/demyelinating disease) or cerebral abscess, and brain biopsy is required for diagnosis.

There has been strong emphasis on the open ring sign on MRI as being a useful feature for identifying acute/tumefactive demyelination ( Fig. 5.1 ). However, recent studies have suggested that most often a closed ring sign is actually seen. Poser et al. estimated that 1–2 per 1000 cases of MS will present with acute lesions.

Figure 5.1, Acute/tumefactive demyelinating lesions prompting biopsy often produce a lesion with an open ring sign on gadolinium enhanced magnetic resonance imaging studies (A–B), characterized by incomplete bright ring at the perimeter of the lesion. While this can serve to distinguish acute demyelinating lesions from cerebral abscess or a highly necrotic metastasis or glioblastoma, unfortunately, not all examples show an open ring.

Early series on acute/tumefactive demyelinating lesions suggested that acute/tumefactive lesions represented disorders intermediate between MS and a usually monophasic demyelinating condition most prevalent in children, ADEM . The largest series of 168 patients reported by Lucchinetti et al. found that most, but not all, patients presenting with an acute/tumefactive demyelinating lesion eventually fulfill criteria for MS diagnosis. In that series, the clinical course was variable prior to biopsy. An acute/tumefactive demyelinating lesion prompting biopsy was the first neurological event in 61 % of patients, but a large percentage, 29 %, had had a clinical history of RRMS disease, and 4 % had a history of progressive disease prior to the acute demyelinating lesion prompting biopsy. Prebiopsy MRI scan had revealed multiple lesions in 70 % of cases. At follow-up, 70 % developed definite MS and 14 % had an isolated demyelinating syndrome. Very large lesions of greater than 5 cm were associated with slightly more clinical dysfunction than smaller lesions. Most importantly, presentation with an acute demyelinating lesion prompting biopsy did not portend an aggressive subsequent downhill clinical course of MS. Median time to second attack was 4.8 years . This study confirmed that acute demyelinating disease prompting biopsy did not automatically predict a fulminant form of MS, sometimes denoted as aggressive or Marburg type MS, the latter a rare form of acute disease with downhill course and early demise, sometimes within 1 year of onset.

A more recent study by Altintas et al. came to much the same conclusions. Of 54 patients, 19 developed MS after a median follow-up of 38 months. Of the 54 study patients, 29 presented with a acute/tumefactive lesion, and 25 had the acute/tumefactive demyelinating lesion develop after diagnosis of MS; 10 had CIS. Most lesions had the closed ring sign (rather than the supposedly more common open ring neuroimaging sign), and OCBs were less frequent in patients with acute/tumefactive lesions.

Recent literature has emphasized that ADEM, AHLE, and NMO can also produce/present as acute/tumefactive demyelinating lesions. Jeong et al. described 31 patients from Korea presenting with acute/tumefactive demyelinating lesions with a median follow-up of 37.6 months. During this time, 11 patients were diagnosed with neuromyelitis optica spectrum disorder (NMOSD), 7 with MS, and 11 remained idiopathic; 6 did not experience any further clinical events (isolated demyelinating syndrome), and 5 patients experienced recurrent demyelinating events that were not consistent with either MS or NMOSD. Of the remaining two patients, one was diagnosed with hyperthyroidism-associated demyelination and one with tacrolimus-induced demyelination.

Spinal cord lesions presenting as acute/tumefactive demyelinating lesions can also occur. Zawelski et al. reviewed spinal cord MRIs for ring-enhancing lesions from 284 AQ4-IgG seropositive patients (i.e., NMO) at the Mayo Clinic from 1996 to 2014. They found that ring enhancement was detected in 50 of 156 (32 %) myelitis episodes in 41 patients (83 % single; 17 % multiple attacks). Thus, it must be emphasized that the clinician, neurosurgeon, and pathologist should eschew labeling all acute/tumefactive demyelinating lesions automatically as acute MS or acute tumefactive MS. In other words, other demyelinating conditions, especially NMO in spinal cord locations, can produce acute/tumefactive lesions that prompt biopsy.

Of the patients with a diagnosis of MS established by the current 2017 McDonald criteria, a few still develop large acute/tumefactive lesions that can prompt biopsy. The cooccurrence of glioma MS in the same patient is uncommon but a well-reported phenomenon. Most cases were high-grade astrocytic tumors that developed after the diagnosis of MS, leading authors to postulate that chronic gliosis in demyelinative plaques might be the underlying substrate for the secondary induction of a glial neoplasm. Rarely, a close anatomical relationship between the glioma and underlying plaque is demonstrated. However, no particular glioma type or genetic signature is associated with these tumors that develop in MS patients. The clinical issue that prompts biopsy is that often these lesions that ultimately prove to be truly neoplastic are presumed prebiopsy to be MS plaques and are followed for too long before biopsy/resection establishes diagnosis. Infections in MS patients may also lead to large lesions that require biopsy diagnosis.

Suspected acute/tumefactive demyelinating lesions are usually biopsied with stereotactic techniques, and tissue volume is often limited. Intraoperative frozen sections with a touch/squash preparation stained with hematoxylin and eosin (H&E) to provide optimal nuclear and cytoplasmic detail are key. The preparation will show hypercellularity and may include a number of blood vessels that lack microvascular proliferation ( Fig. 5.2A ). Higher magnification allows recognition of the monotonous round nuclei, bubbly cytoplasm with well-defined rounded borders, and low nuclear to cytoplasmic ratio characteristic of macrophages ( Fig. 5.2B ). Macrophages possess small delicate nucleoli and lack the large nucleoli seen in amoebae.

Figure 5.2, Acute/tumefactive demyelinating lesions prompting biopsy on touch/smear preparation will show hypercellularity (A) and macrophages with round nuclei, bubbly cytoplasm and well-defined rounded borders (B). On permanent formalin-fixed sections, the demyelination may show sharp demarcation between the demyelinating lesion and adjacent uninvolved brain may (C) or may not (D) be recognizable. Special stains for myelin better highlight the sharp demarcation between the demyelinating lesion and unaffected adjacent white matter (E). Moderate hypercellularity is due to admixed reactive astrocytes (D), and occasionally, mild vacuolization of white matter is apparent (F).

On permanent, formalin-fixed sections, the demyelination may be recognized as being perivenular, although coalescence of the demyelination can somewhat obscure this feature. A sharp demarcation between the demyelinating lesion and adjacent uninvolved brain may ( Fig. 5.2C ) or may not ( Fig. 5.2D ) be recognizable. Often special stains for myelin better highlight the sharp demarcation between the demyelinating lesion and unaffected adjacent white matter ( Fig. 5.2E ). Moderate hypercellularity may prompt consideration of glial tumor, especially diffuse astrocytoma, given the admixed reactive astrocytes ( Fig. 5.2D ). Occasionally, mild vacuolization of white matter is apparent ( Fig. 5.2F ).

Macrophages are usually found throughout acute demyelinating lesions and can simulate neoplastic oligodendrocytes, especially at the time of intraoperative frozen section consultation. Search for perivascular macrophages is quite helpful in diagnosis of an acute/tumefactive demyelinating lesion, since oligodendrogliomas and astrocytomas do not show macrophages in a perivascular arrangement ( Fig. 5.3A ). Indeed, untreated low-grade glial tumors lack conspicuous collections of macrophages anywhere in the tumor. While it is useful to search for perivascular lymphocytic cuffing, it rarely is brisk and is often absent on stereotactic biopsies; thus, these are not required for diagnosis ( Fig. 5.3B ). The pathologist should distinguish perivascular lymphocytic cuffing from true lymphocytic vasculitis that involves and distorts vessel wall and most often occurs in pediatric age groups. Lymphocytic vasculitis is not associated with fibrinoid vascular necrosis, and hence, the absence of this feature is not helpful in diagnosing this type of vasculitis. In instances where brisk perivascular lymphocytic cuffing is present and consideration is given to a clonal T cell lymphocytic process, and immunohistochemistry is performed for characterizing the lymphocytes, CD8 + T cells ( Fig. 5.3C ) are more frequent than CD4 + T cells ( Fig. 5.3D ). B cells are variable in number but may not be conspicuous.

Figure 5.3, Acute/tumefactive demyelinating lesions, but not tumors, can show macrophages in a perivascular arrangement (A). Perivascular lymphocytic cuffing is rarely brisk (B). CD8+ T cells (C) are more numerous than CD4+ T cells (D) in lesions that eventually prove to be first presentation of multiple sclerosis, whereas the reverse may be true in samples that eventually prove to be myelin oligodendrocyte glycoprotein antibody disease.

Work-up beyond H&E often includes confirmation of macrophages in the lesion by CD68 ( Fig. 5.4A ) and more specifically by CD163 ( Fig. 5.4B ). Rare granular cell astrocytoma/glioblastoma ( Fig. 5.4C ) will manifest immunoreactivity for CD68 due to increased lysosomes in cytoplasm, but this will not occur with CD163 IHC. If the pathologist considers a fungal infection and utilizes Gomori methenamine silver stains, it should be recognized that nonspecific staining of the myelin debris within cytoplasm can prompt diagnostic confusion ( Fig. 5.4D ).

Figure 5.4, Acute/tumefactive demyelinating lesions, but not untreated tumors, show macrophages with immunoreactivity for CD68 (A) and more specifically by CD163 (B). Rare granular cell astrocytoma/glioblastoma (C) will manifest immunoreactivity for CD68 due to increased lysosomes in cytoplasm, but this will not occur with CD163 immunohistochemistry. If the pathologist uses Gomori methenamine silver stain, it should be recognized that nonspecific staining of the myelin debris within cytoplasm can prompt diagnostic confusion with microorganisms (D). Acute (actively demyelinating) lesions contain macrophages filled with luxol fast blue–positive material (E), but lesions may be biopsied at the subacute stage and may contain periodic acid Schiff–positive material indicative of later stages of complex lipid/myelin breakdown (active post-demyelination), especially in perivascular locations (F).

All demyelinating lesions, by definition, show relatively greater loss of myelin than axons and histochemical or immunohistochemical stains for myelin > axon loss should be performed. Luxol fast blue/periodic acid Schiff (LFB-PAS) can be used instead of immunostaining for myelin basic protein (MBP) or proteolipid protein (PLP) in showing the full extent of acute demyelination. Acute (actively demyelinating) lesions contain macrophages filled with LFB-positive material ( Fig. 5.4E ), but lesions may be biopsied at the subacute stage and may contain PAS-positive material indicative of later stages of complex lipid/myelin breakdown (active post-demyelinating), especially in perivascular locations ( Fig. 5.4F ). The use of immunohistochemical stains for minor and major myelin proteins even more precisely aids in distinguishing actively demyelinating from post-demyelinating lesions, but they are seldom utilized at the time of biopsy. These IHC stains against minor myelin proteins include MOG and 2’3’-cyclic-nucleotide 3’-phosphodiesterase (CNPase).

Axonal preservation is considerably better than the degree of myelin loss and immunostaining for neurofilament protein ( Fig. 5.5A ) and should be conducted on the same section as that assessed for myelin loss to appreciate this discordance. Greater myelin than axonal loss leads to the correct interpretation of the lesion as demyelination, rather than infarctive, ischemic damage. However, this does not mean that axonal damage does not occur. Bielschowsky silver stains or anti-neurofilament immunostaining may show axonal swellings ( Fig. 5.5B ). Reactive astrocytosis is a key feature of MS lesions and diffuse, evenly distributed astrocytes show long tapering processes on immunostaining for glial fibrillary acidic protein (GFAP) and can be seen throughout the lesion ( Fig. 5.5C ). Highly reactive astrocytes may show slightly enlarged, vesicular nuclei and granular mitoses, Creutzfeldt-Peters cells with micronuclei ( Fig. 5.5D ), further simulate neoplasm. Although few specific glial tumor cell markers exist, in rare instances where glial neoplasm is still a consideration on biopsy, consideration should be given to performing immunohistochemistry for IDH-1 (R132H) or H3 K27M, both of which are never identified in reactive scenarios and/or the testing for whole chromosome arm loss of 1p + whole arm loss of 19q for identifying oligodendroglioma. Even more rarely, consideration might be given to mutation/fusion testing.

Figure 5.5, Axonal preservation is considerably better than the degree of myelin loss and immunostaining for neurofilament protein (A) but anti-neurofilament immunostaining may show axonal swellings (B). Reactive astrocytosis is a key feature of multiple sclerosis lesions (C). Highly reactive astrocytes may be present that show slightly enlarged vesicular nuclei and granular mitoses, Creutzfeldt-Peters cells with micronuclei (D). Creutzfeldt-Peters cells are not found in neuromyelitis optica lesions.

PML AND PRIMARY CNS LYMPHOMA AS DIAGNOSTIC CONSIDERATIONS FOR ACUTE/TUMEFACTIVE DEMYELINATING LESIONS PROMPTING BIOPSY

The diagnostic considerations of lymphocytic vasculitis, clonal T cell processes, and glial neoplasm are mentioned above in the context of work-up. In adults, two further conditions require further elimination: progressive multifocal leukoencephalopathy (PML) and demyelination secondary to CNS lymphoma, but pathologists are cautioned here about the fact that virally infected cells with PML will label with MIB-1 and p53 immunostains, further contributing to the false impression of glial tumor. The cooccurrence of gliomas and MS is detailed above.

In addition, too often PML is only considered in the differential diagnosis by either the clinician or pathologist when there are well-known risk factors in the patient, such as acquired immunodeficiency syndrome, hematological malignancy, transplant recipients, and collagen vascular or autoimmune disorders with or without treatment with immunosuppressive medication(s) such as rituximab. Less well-known risks for PML include hepatic cirrhosis, renal failure, low CD4 T cell counts (idiopathic CD4 lymphocytopenia, and age-related immunosuppression). Although beyond the scope of this chapter, MS patients receiving natalizumab treatment are at risk for development of PML, especially if seropositive for the JC virus of PML and especially if they have received the drug for an extended period of time. Thus, especially in adults, work-up to exclude PML by immunohistochemistry for SV40 may be prudent in acute/tumefactive demyelinating lesions prompting biopsy.

Demyelination can also be a harbinger of lymphoma since primary CNS lymphoma (PCNSL) can be preceded by sentinel demyelination. While demyelination with total, or near-total, eradication of the PCNSL cells can be spontaneous, this most often occurs in the setting of steroid administration to the patient prior to biopsy. The amount and duration of steroids received by the individual patient necessary to obliterate the lymphoma cells are highly varied and cannot be predicted.

Diagnostic red flags that provide possible hints that a lesion may be CNS lymphoma are older patient age, greater degree of perivascular inflammation, numerous apoptoses, and incomplete rather than confluent/complete demyelination in the lesion. Lesions tended to have less thorough myelin loss than in MS, a fuzzy lesion edge, and T cell infiltrates that exceeded those of MS/immune-mediated demyelination fivefold. LFB-PAS was superior to immunohistochemical myelin stains such as MBP and PLP in showing the extent of demyelination. Such biopsies are otherwise identical to immune-mediated demyelination such as MS in terms of manifesting sheets of macrophages, but may additionally contain enlarged, cytologically-atypical CD20 + /PAX5 +  B cells as well as some areas with greater axonal loss, although the latter “empty bed” is not detectable, and areas may manifest axonal preservation. In many cases, only clinical follow-up and reemergence of the PCNSL prove correct diagnosis. Rarely, other solid neoplasms can present with sentinel demyelinative lesion.

In pediatric patients, PML and PCNSL are uncommon considerations. In general, in either adult or pediatric patients, acute/tumefactive lesions prompting biopsy that prove to be truly demyelinating after extensive histological and immunohistochemical work-up and not infectious or demyelinating disorders, are believed to cause MS, ADEM, MOGAD, NMO, or rarely other conditions based on clinical, serological, and neuroimaging features and not on the basis of histological features.

In pediatric patients, two main differential considerations for acute/tumefactive demyelinating lesions are ADEM and AHLE (Weston-Hurst disease), which is thought to be a fulminant variation on ADEM, and can be encountered either at biopsy or autopsy. The relationship of AHLE to ADEM is discussed below.

MOGAD and NMO are also considerations, especially in pediatric patients. Contrasting clinical and histological features of MS, ADEM, NMO, and MOGAD are shown in Tables 5.1 and 5.2 .

Since all four of these disorders in children may be the substrate for acute/demyelinating lesions prompting biopsy, all four of these disorders are described below before returning to features of MS at intermediate/late stages, the latter of which are usually encountered by pathologists at autopsy.

ACUTE DISSEMINATED ENCEPHALOMYELITIS

Clinical Features

ADEM is typically a monophasic, acute, inflammatory demyelinating disorder that most commonly occurs in children. Older terms for ADEM included perivenous encephalomyelitis, postinfectious encephalomyelitis, and postvaccinal encephalomyelitis. It was originally described after smallpox vaccinations. The disease was later identified following measles, mumps, rubella, varicella, and vaccinia infections. Today, it usually occurs in children after a nonspecific upper respiratory illness. Onset is usually rapid, with a clinical course that evolves over hours to maximum deficits within days (mean, 4.5 days); there is a wide range in clinical severity of ADEM, but most patients make a full neurological recovery.

Today, ADEM needs first and foremost to be distinguished from MOG antibody disease, also referred to as MOGAD, as discussed below, especially since an ADEM-like presentation is classical for pediatric onset MOGAD (see below). It is estimated that 30 %–50 % of cases initially considered ADEM prove to be MOGAD after serological testing; thus, true ADEM cases, by definition, must be shown to be MOG-negative. Adding to the diagnostic confusion, pediatric ADEM can be the first manifestation of pediatric MS or pediatric NMO/NMOSD.

Clinical manifestations of ADEM are variable. Typically, there is an acute onset of encephalopathy (alteration in consciousness with stupor, lethargy) in association with polyfocal neurologic deficits, sometimes preceded by prodromal symptoms (fever, malaise, irritability, somnolence, headache, nausea, and vomiting). The clinical course of ADEM is usually rapidly progressive, with maximal deficits within 2 to 5 days. Only a minority of children with ADEM (15 %–25 %) require admission to an intensive care unit. Frequent neurologic manifestations include pyramidal signs, ataxia, acute hemiparesis, optic neuritis or other cranial nerve involvement, seizures, spinal cord syndrome, and impairment of speech.

Clinical consensus criteria have been established and revised several times. According to Krupp et al., patients should meet all these features: “A first polyfocal, clinical CNS event with presumed inflammatory demyelinating cause, encephalopathy that cannot be explained by fever, no new clinical and MRI findings emerge 3 months or more after the onset, brain MRI is abnormal during the acute (3 month) phase.” Typically on brain MRI there are “diffuse, poorly demarcated, large (> 1–2 cm) lesions involving predominantly the cerebral white matter, T1 hypointense lesions in the white matter are rare, and deep gray matter lesions (e.g., thalamus or basal ganglia) can be present.” Multiple ( Fig. 5.6A ) and bilateral deep gray matter lesions ( Fig. 5.6B ) are all of the same age and are usually characteristic enough to make the diagnosis without biopsy. Large lesions may be more clinically confusing, and thus, a subset of acute/tumefactive demyelinating lesions prompting biopsy prove to be true ADEM.

Figure 5.6, An acute/tumefactive demyelinating lesion that proves to be acute disseminated encephalomyelitis (ADEM) as it did in this child with negative serology for other demyelinating conditions often presents with multiple lesions on MRI (A), often involving deep gray matter such as basal ganglia (B). Histologically, ADEM is characterized by small zones of perivenular pallor with variable numbers of non-neoplastic lymphocytes (C), with myelin loss seen better on luxol fast blue–periodic acid Schiff (D) and immunostaining for proteolipid protein (E), sometimes with coalescence (F). ( Courtesy Dr. Ahmed Gilani )

While serology results distinguish pediatric ADEM from MOGAD or NMO/NMOSD, clinical and neuroimaging features are required to distinguish ADEM from pediatric MS since specific serological tests do not exist for either ADEM or MS. On neuroimaging studies, ADEM is more likely than MS to involve deep gray matter and show cortical involvement and to show diffuse bilateral lesions, poorly marginated lesions, and large globular lesions on neuroimaging studies. In contrast, a periventricular pattern of lesions, lesions perpendicular to the long axis of the corpus callosum, and black holes on T1 sequences indicative of tissue destruction all favor MS over ADEM. Other imaging features atypical for ADEM include diffuse symmetric brain lesions (which might indicate genetic/metabolic disorders, leukodystrophies or mitochondrial diseases), mesial temporal lesions (that might indicate autoimmune encephalitis), or ischemic lesions with restricted diffusion.

As summarized by Pohl et al., CSF leukocyte count can be normal in 42 %–72 % of children, pleocytosis is typically mild (mostly lymphocytes and monocytes), and CSF protein can be increased, but CSF OCBs are absent. Atypical features on CSF analysis that should prompt consideration of alternate diagnoses include cell count > 50/mm 3 , neutrophilic predominance, or protein > 100 mg/dL.

Due to its low frequency, the category of recurrent ADEM has been eliminated. The definition of multiphasic ADEM has also been revised and is now defined as “two episodes consistent with ADEM separated by 3 months but not followed by any further events. The second ADEM event can involve either new or a reemergence of prior neurologic symptoms, signs and MRI findings.” A percentage of children with relapsing ADEM have other diseases such as NMOSD or MOGAD.

High-dose corticosteroids are currently widely accepted as first-line therapy, and most children make a complete recovery, although children affected less than age 5 years may be left with long-term cognitive deficits. Since fatal examples are very rare, pathologists almost never encounter ADEM at autopsy.

Pathologic Features

The signature microscopic feature consists of narrow cuffs or sleeves of myelin loss around small veins. Veins may be congested. The perivenular areas often show fairly subtle demyelination on H&E ( Fig. 5.6C ) or LFB-PAS ( Fig. 5.6D ), although immunostains for myelin show the perivenular location well ( Fig. 5.6E ). Some coalescence can be found ( Fig. 5.6F ). The striking feature is often large numbers of cytologically normal lymphocytes surrounding blood vessels ( Fig.5.6C ). While this may raise consideration of lymphocytic vasculitis, there is no vessel wall destruction and the vessels involved are often in the white matter, rather than gray as is the usual case with lymphocytic vasculitis. A few lymphocytes are often present within the meninges, and sometimes microglial cells are noted in the cortex, possibly as a correlate to the encephalopathy, but numerous tight microglial clusters and neuronophagia (indicative of ongoing viral infection and tissue damage, i.e., viral encephalitis) are not present. Unlike typical MS, lesions are all of the same age. In the exceedingly rare instances where patients come to autopsy during the course of the disease, the brain is swollen and lesions can be widely distributed throughout brain and spinal cord.

Contrasting clinical and histological features of MS and ADEM, as well as other demyelinating disorders discussed below (NMO and MOGAD), are shown in Tables 5.1 and 5.2 .

ACUTE DISSEMINATED ENCEPHALOMYELITIS—FACT SHEET

Definition

  • n

    Monophasic, acute demyelinating disorder characterized by small perivascular sleeves of myelin loss widely disseminated throughout the brain and spinal cord, along with associated perivascular mononuclear cell infiltrates

  • n

    MOG antibody-negative, by definition

  • n

    NMO serology negative, by definition

Incidence and Location

  • n

    0.3–0.6/100,000 per year

  • n

    High incidence in past in older vaccines that are no longer in use that were contaminated with host animal CNS tissue in which they were propagated

  • n

    Later identified after measles, mumps, rubella, varicella, or vaccinia infections; also influenza, enterovirus, and bacterial infections

  • n

    Today, usually occurs after a nonspecific upper respiratory illness

  • n

    Risk of developing ADEM after vaccination considerably lower than after infection itself

  • n

    Usually affects cerebral white matter, may affect deep gray matter

  • n

    Demyelination confined to CNS

Sex and Age Distribution

  • n

    More frequent in children <10 years (mean age 5–8 years) (as opposed to pediatric MS which is more frequent in adolescents)

  • n

    No sex predilection (as opposed to pediatric MS which has a female predominance)

  • n

    Winter and spring seasonal peaks of presentation in some studies correlate with association with antecedent infection as trigger

Clinical Features

  • n

    Clinical onset of symptoms is usually acute, progressive, maximal deficits within 2–5 days

  • n

    Signs: unilateral or bilateral pyramidal signs, acute hemiplegia, ataxia, cranial nerve palsies, visual loss due to optic neuritis, seizures, spinal cord involvement

  • n

    Today, patients usually achieve full recovery and no residual neurologic deficit

  • n

    Usually monophasic course

Radiologic Features

  • n

    Neuroimaging features of ADEM versus MS: bilateral deep gray matter lesions in thalami, basal ganglia, poorly marginated lesions in ADEM versus corpus callosum long axis perpendicular lesions and periventricular lesions in MS, often with more demarcated margins

  • n

    Absence of tissue destruction due to prior demyelinating event (i.e., no T1 black holes)

Prognosis and Treatment

  • n

    Most cases now nonfatal; fatality rate historically was highest after measles infection and smallpox vaccination

  • n

    Today, patients usually achieve full recovery, although children <5 years of age may show cognitive deficits

  • n

    Treatment is high-dose corticosteroids

Differential Diagnosis

The main differential diagnosis for clinicians and pathologists is between ADEM and MOGAD or between ADEM and pediatric MS. Serological studies can distinguish the first two entities (see below), and time often needs to elapse before patients meet the McDonald criteria for MS. It can be particularly difficult to distinguish ADEM from MS clinically when ADEM occurs in adults. Often, follow-up clinical and MRI data are necessary to distinguish the monophasic versus disseminated in time and space nature of the two diseases.

ACUTE DISSEMINATED ENCEPHALOMYELITIS—PATHOLOGIC FEATURES

Gross Findings

  • n

    Patients dying in acute phases of the disease have a diffuse cerebral edema and often herniations (rarely seen today)

  • n

    Demyelination often difficult to identify grossly

Microscopic Features

  • n

    Characteristic narrow cuffs or sleeves of myelin loss around small veins, although often subtle on H&E

  • n

    Cellular infiltrates in demyelinated areas are composed of chiefly macrophages rather than lymphocytes

  • n

    Lesions predominantly involve white matter, cortical gray matter, and deep gray matter

  • n

    Vessel walls are intact and lack severe destruction, fibrinous exudate, and surrounding red blood cells and neutrophils that are seen in AHLE

  • n

    Lymphocytic perivenular cuffing may be intense; unlike lymphocytic vasculitis, no vessel wall destruction and involved vessels predominantly white matter

  • n

    Mild number of lymphocytes in the meninges is typical

  • n

    Microglial clusters and neuronophagia (indicative of ongoing viral infection and tissue damage) are not typically present

  • n

    Unlike typical MS, lesions are all of the same age

Ultrastructural Features

  • n

    No specific features

Immunohistochemical Features

  • n

    No specific features

Genetics

  • n

    No known genetic predisposition

Differential Diagnosis

  • n

    Acute MS

  • n

    AHLE

  • n

    Acute MOGAD, NMOSD

ACUTE HEMORRHAGIC LEUKOENCEPHALITIS (WESTON-HURST DISEASE)

Clinical Features

This disorder is often considered a rare, fulminant form of ADEM, with numerous perivascular hemorrhages as a result of more severe vascular injury, although some debate exists. Both children and adults can be affected. The disease is manifested by an abrupt onset of fever, neck stiffness, seizures, and focal signs. About half the patients have an antecedent upper respiratory tract illness or influenza that occurred 2 to 12 days before onset, similar to ADEM. Because of the fulminant onset, the disease can mimic a direct CNS infection or toxic ingestion. The disease in the past was usually fatal, but in recent years, an increasing number of survivors has been reported, often when treated with therapeutic plasma exchange or intravenous immunoglobulin. Occasional cases will prompt biopsy and, thus, the disease is considered in the differential of acute/tumefactive demyelinating lesions prompting biopsy, especially if hemorrhagic features are present.

ACUTE HEMORRHAGIC LEUKOENCEPHALITIS (HURST DISEASE)—FACT SHEET

Definition

  • n

    Hyperacute, fulminant form of acute disseminated encephalomyelitis with numerous perivascular hemorrhages because of more severe vascular injury

Incidence and Location

  • n

    Rare; estimated that only 2% of all ADEM is this hyperacute type

  • n

    Cerebral hemispheric white matter predominantly affected

  • n

    Involvement of the spinal cord uncommon, in contrast to acute disseminated encephalomyelitis

Sex and Age Distribution

  • n

    Childhood and young adult ages

Clinical Features

  • n

    Abrupt onset of fever, neck stiffness, seizures, focal signs

  • n

    In about half the cases, there is an antecedent upper respiratory tract illness 2 to 12 days before onset, similar to acute disseminated encephalomyelitis

  • n

    CSF shows increased protein, pleocytosis with chiefly neutrophils, and often red blood cells

  • n

    Peripheral neutrophilic leukocytosis may also be seen

Radiologic Features

  • n

    Cerebral swelling and variable petechial hemorrhages

  • n

    Confluent nonenhancing lesions predominantly affect cerebral white matter; lesions may be optimally identified on fluid attenuation inversion recovery (FLAIR) images

  • n

    Lesions may be asymmetric

  • n

    Lesions are larger, with more edema and mass effect, than those of acute disseminated encephalomyelitis

Prognosis and Treatment

  • n

    Usually considered fatal in the older literature, but recent studies have reported an improved prognosis

Pathologic Features

Grossly, the brain is swollen and edematous and herniations are frequent. The signature pathologic feature is the presence of large areas of white matter damage associated with multiple petechial to larger hemorrhages throughout the cerebral white matter. The extent of hemorrhage is highly variable. Microscopically, “ring and ball” hemorrhages surround necrotic venules, which show fibrinoid vascular necrosis, fibrin exudates, and neutrophilic debris ( Figs. 5.7A–C ). Perivenous demyelination is seen but may be overshadowed by the perivenous hemorrhages.

Figure 5.7, Acute hemorrhagic leukoencephalitis shows perivenular myelin loss on luxol fast blue–periodic acid Schiff (A), and unlike acute disseminated encephalomyelitis, vessels with necrosis and neutrophilic debris (B) as well as fibrinoid necrosis (C).

Differential Diagnosis

Both acute fat embolism to the brain and hypoxic-ischemic leukoencephalopathy can cause widespread white matter hemorrhages. Staining for fat globules (by oil red O or Sudan black B stains) within the small vessels will identify fat embolism. In endemic areas of the world, fulminant cerebral malaria could also be a consideration for the pathologist based on petechial hemorrhages.

AHLE should be distinguished from acute necrotizing encephalitis (ANE). ANE has been specifically reported in patients infected with COVID-19, following seasonal influenza, and following herpes virus infection. Necrotizing lesions primarily in the thalamus and brainstem, as well as in the temporal lobes and insula, should prompt consideration of an overlapping syndrome caused by mutations in the RANBP2 gene resulting in a familial autosomal dominant disease with adverse responses to influenza infection.

ACUTE HEMORRHAGIC LEUKOENCEPHALITIS (HURST DISEASE)—PATHOLOGIC FEATURES

Gross Findings

  • n

    Brain is swollen and edematous and herniations are frequent

  • n

    Multiple petechial to larger hemorrhages, sometimes with associated large areas of necrosis, seen throughout the cerebral white matter

  • n

    Basal ganglia may show necrosis

Microscopic Features

  • n

    Ring and ball hemorrhages surround necrotic venules

  • n

    Venules show fibrinoid vascular necrosis, fibrin exudates, and neutrophilic debris

  • n

    Neutrophilic inflammation contrasts with the lymphocytic inflammation of acute disseminated encephalomyelitis

  • n

    Perivenous demyelination is seen but may be overshadowed by the perivenous hemorrhages

  • n

    Demyelination is often more extensive and coalescent than that seen in ADEM

Ultrastructural Features

  • n

    No specific features

Immunohistochemical Features

  • n

    No specific features

Genetics

  • n

    ANE due to Ran binding protein 2 ( RANBP2 ) gene mutations

  • n

    RANBP2 is genetic polymorphism associated with recurrent episodes of necrotizing encephalitis with respiratory viral infections

Differential Diagnosis

  • n

    Cerebral fat embolism

  • n

    Acute disseminated encephalomyelitis

  • n

    Hypoxic-ischemic hemorrhagic leukoencephalopathy

MYELIN OLIGODENDROCYTE GLYCOPROTEIN ANTIBODY DISEASE

MOGAD is a rare autoimmune disorder with antibodies against MOG predominantly involving the optic nerve and spinal cord leading to vision loss and paralysis. Clinically the disease usually resembles NMOSD and up to 40 % of seronegative NMSOD patients (i.e., negative for AQ4-Ab may have MOGAD). MOG is expressed on the outer surface of myelin, not on astrocytes as is AQ4, and thus the disease histologically causes demyelination rather than the astrocyte damage seen in NMO, as discussed below. MOG is a minor protein component of myelin but is highly immunogenic. The diagnosis of MOGAD requires the detection of MOG-Ab on sensitive cell-based serum assay and the presence of one or more criteria: acute disseminated encephalitis-like clinical features, optic neuritis, transverse myelitis, brain/brainstem syndrome compatible with demyelination, and exclusion of other diagnoses. NMO-IgG, the serum antibody diagnostic for NMO, must be absent. Today it is recognized that what was formerly considered true ADEM in 30 %–50 % of cases is actually MOGAD.

MOGAD affects all age groups, with a median age of onset in the early to mid-30s and optic neuritis being the most frequent presenting phenotype. Some suggest a near equal sex skew, others a female to male 2:1 distribution. Disease course can be either monophasic or relapsing, with subsequent relapses most commonly involving the optic nerve. Transverse myelitis at onset is more common in adults than children and is the most significant predictor of long-term outcome. A brainstem syndrome with intractable nausea with hiccups and vomiting is characteristic.

In children, mean age of onset is 7.3 years, and age at presentation is strongly correlated with clinical features. Children less than age 11 years at presentation usually have an ADEM-like episode as their first clinical feature, as seen in 44 % of cases, and optic neuritis as their first clinical feature in 33 %. In children greater than 11 years of age, only 16 % will present with ADEM-like symptoms and 66 % with optic neuritis.

Similar to true ADEM, which is typically monophasic, most MOGAD children have a monophasic disease, although a subset will experience relapses. In one study by Waters et al., which involved serial serum analysis in children with MOGAD, 57 % of patients who were seropositive at onset became seronegative, with a median time to conversion of 1 year. Clinical relapses occurred in 38 % who remained persistently seropositive and in 13 % who became seronegative. The study concluded that “even when persistently positive, most anti-MOG antibody-positive children experience a monophasic disease.”

Unlike MS, MOGAD is not a relapsing-remitting disorder. MOGAD patients are also thought to be at lower risk of further relapses than AQP4-positive patients (NMO) and have better visual and motor outcomes. Therapy recommendations for MOGAD are evolving, but often high-dose steroids and plasma exchange are used for acute attacks and immunosuppressive therapies, such as steroids, oral immunosuppressants, and rituximab as maintenance treatment(s). The study of MOGAD children with serial serum analysis concluded that “The presence of anti-MOG antibodies at the time of incident demyelination should not immediately prompt the initiation of long-term immunomodulatory therapy.”

MOG antibody IgG is detected in serum, and the use of a cell-based assay (indirect fluorescence test or fluorescence-activated cell sorting) is mandatory to avoid false positives (as is employing full-length human MOG as the target antigen). Other CSF parameters are not distinctive in that pleocytosis occurs in 44 %–85 % of patients and is more common in children; positive OCBs are uncommon—unlike pediatric MS—and CSF protein is raised in around a third of cases. CSF in general is more similar to NMO than to MS in patients with MOGAD in that more than half of patients have white blood cell counts of 50 µ/L, while that degree of elevation is seen in < 5% of MS patients and OCBs are found in < 10% in comparison to MS where they are identified in greater than 85 % of patients. Unlike NMO, which is an astrocytopathy, the CSF does not show GFAP.

Similar to the situation with ADEM, occasional cases prompt biopsy since MRI can show multifocal lesions ( Figs. 5.8A–B ). Histologically, MOGAD at biopsy for acute/tumefactive demyelinating lesion is similar, if not identical, on routine light microscopic/H&E examination to ADEM or MS. There is perivenous pallor on H&E ( Fig. 5.8C ) and LFB-PAS ( Fig. 5.8D ) and variable numbers of perivascular non-neoplastic lymphocytes ( Fig. 5.8C ), macrophages ( Fig. 5.8E ) and reactive astrocytes Fig. 5.8F ). Unlike NMO, eosinophils, astrocytopathy with dystrophic blunted astrocytic processes, oligodendrocyte loss, perivascular complement deposition, and AQP4 loss are all absent. MS also lacks these features. While immunohistochemical assessment of complement deposition is seldom undertaken in routine practice, both MS and MOGAD disease can show complement in macrophages. The neuropathological features of MOGAD are felt to be comparable to one pattern described in MS, namely, the pattern II histopathology of MS, i.e., lesions have complement and IgG deposits at the sites of ongoing demyelination. The patterns of MS are discussed in more detail in the section on intermediate and later features of MS below.

Figure 5.8, Myelin oligodendrocyte glycoprotein antibody disease occasionally prompts biopsy similar to the situation with acute disseminated encephalomyelitis; MRI can show multifocal lesions (A–B).

While in active disease, MOGAD and MS may share similar histopathology, it should be emphasized that these are considered very different diseases in terms of clinical disease, pathogenesis, and response to therapy. Ultimately, serological, clinical, and neuroimaging features serve to make the correct diagnosis following biopsy for an acute/tumefactive demyelinating lesion.

Since the disease is seldom fatal, few autopsy descriptions have been published. However, in the study by Hoftberger et al., features of two autopsies and 22 biopsies were found to be similar. Cases showed coexistent perivenous and confluent demyelination, intracortical plaques, and complement deposition sometimes around blood vessels but not on the glial limitans (as in NMO). Although subpial demyelinative cortical lesions were found, there were far more frequent intracortical demyelinative lesions. Specifically, subpial plaques were found in about 15 % of MOGAD versus about 30 % in MS, leukocortical in about 5 % in MOGAD versus 50 % in MS, and intracortical in 80 % versus 20 %. MOGAD showed a predominance of CD4 T cells in contrast to MS, which shows a predominance of CD8 T cells, with CD4:CD8 ratios of 2.89 (range, 1.26–29) and also showed more granulocytes than does MS. Few B cells are present in MOGAD. MOGAD lesions did not show AQP loss, as would be found in NMO. No slowly radially expanding smoldering lesions were found in MOGAD, unlike MS during progressive disease. Interestingly, these authors found no preferential MOG loss on immunostaining. However, another study by Takai et al. did demonstrate preferential MOG loss on immunostaining.

Contrasting clinical and histological features of MS, ADEM, MOGAD, and NMO (see below), are shown in Tables 5.1 and 5.2 .

MYELIN OLIGODENDROCYTE GLYCOPROTEIN ANTIBODY DISEASE—FACT SHEET

Definition

  • n

    Acute demyelinating disorder, most monophasic

  • n

    MOG antibody positive, by definition

  • n

    NMO IgG serology (AQP4-Ab) negative, by definition

Incidence and Location

  • n

    30%–50% of what formerly was considered ADEM found to be MOGAD

  • n

    Higher risk of recurrence than with true ADEM

  • n

    More commonly involves deep gray matter than does MS

  • n

    Sites of lesions more easily distinguished from MS than from true NMO

  • n

    Demyelination confined to CNS

Sex and Age Distribution

  • n

    Far less strong female predilection than MS or NMO (i.e., as opposed to pediatric MS, which has a female predominance)

  • n

    Occasionally follows infection

Clinical Features

  • n

    Presentation may be acute disseminated encephalitis-like clinical features, optic neuritis, transverse myelitis, brain/brainstem syndrome compatible with demyelination, or any combination of these features

  • n

    Optic neuritis often more severe than in MS; may be similarly severe to NMO at nadir but patients show better recovery

  • n

    Optic nerve edema in majority, bilateral optic neuritis in 30%–40%

  • n

    Clues to optic neuritis in MOGAD: enhancement of optic nerve sheath, enhancement extending to orbital soft tissues, bilateral enhancement not involving chiasm

  • n

    Myelitis may be severe but generally better recovery than with NMO

  • n

    Recurrent longitudinally extensive transverse myelitis (LETM) far more common in NMO than MOGAD

  • n

    Usually monophasic course

Radiologic Features

  • n

    Neuroimaging more similar to NMO than to MS

  • n

    Absence of tissue destruction due to prior demyelinating event (i.e., no T1 black holes)

  • n

    Spinal cord if LETM found in MOGAD often show central location, less enhancement in contrast to NMO where cord is often swollen and shows enhancing lesions

  • n

    In contrast, MS spinal cord lesions often shorter, more often peripherally enhancing, not central

Prognosis and Treatment

  • n

    Mild accumulation of disability over time, in contrast to MS

  • n

    Course lacks the relapsing-remitting features of MS and lacks primary or secondary progressive clinical course of MS

  • n

    Prognosis significantly better than with NMO where there is major accumulation of disability over time with attacks

  • n

    Treatment is high-dose corticosteroids

MYELIN OLIGODENDROCYTE GLYCOPROTEIN DISEASE—PATHOLOGIC FEATURES

Gross Findings

  • n

    Usually nonfatal but few autopsy studies similar to biopsy studies

Microscopic Features

  • n

    CD4 T cells >> CD8 T cells unlike MS, where CD8 T cells dominate

  • n

    Cortical demyelination present

  • n

    More granulocytes than MS

  • n

    No AQP4 loss on immunostaining

Ultrastructural Features

  • n

    No specific features

Immunohistochemical Features

  • n

    No specific features

Genetics

  • n

    No known genetic predisposition

Differential Diagnosis

  • n

    Acute/active/early MS

  • n

    Acute disseminated encephalomyelitis

  • n

    NMO

NEUROMYELITIS OPTICA

Clinical Features

NMO, formerly called Devic disease, is an autoimmune demyelinating disorder of the CNS recognized to be a disease distinct and completely separate from MS, with a predilection for the optic nerves and spinal cord. It is far less common than MS. In the past, it was debated whether NMO was a variant of MS or a separate disorder, since involvement of these two sites can occur in both conditions. Complete distinction is now possible since NMO is now defined by clinical, neuroimaging, and laboratory criteria, specifically, the identification of a specific autoantibody response in serum against the astrocyte water channel AQP4 (NMO-IgG). AQP4-seronegative patients are considered to have NMOSD. There have been three published sets of NMOSD criteria (1999, 2006, 2015 International Panel for NMO Diagnosis). Criteria incorporate results of a pathognomonic serum test that detects NMO-IgG. This antibody targets the most abundant water channel protein in the CNS, AQP4-IgG, and on neuroimaging and at pathological examination, lesions are found in anatomic sites with the largest number of astrocytic foot processes. AQP4 is present in the pia and choroid plexus, and the ependymal and histological sections of these areas can show loss of AQP4 with immunostaining and complement deposition, accompanied by microglial activation, in these areas, sites not affected in MS.

The diagnosis of NMO with AQP4-IgG (NMO-AQP4 +) requires one of six core clinical characteristics and a positive serum test for AQP4-IgG. There is no role for CSF testing for the AQP4-Ab since it represents spillover from the serum.

The core clinical presentations are distinguished by their neuroanatomic locations: optic nerve, spinal cord, area postrema (dorsal medulla), diencephalon, brainstem, and cerebrum. Optic neuritis typically presents as acute vision or visual field loss in one or both eyes, whereas transverse myelitis (TM) may present with a variety of motor, sensory, or sphincter problems. TM is commonly (85 % of NMO cases) LETM, defined as involving the spinal cord and spanning the length of three or more vertebral body segments, and involves the central cord with contrast enhancement rather than the lateral cord as does MS. Patients may be wheelchair-bound at the peak of disease. Up to 30 % of patients with NMO TM can show ring enhancement, which, as noted above, serves as the substrate for biopsy of spinal cord acute/tumefactive demyelinating lesion. Optic neuritis is more often bilateral and more often involves the chiasm than in MS; it is often more severe, and complete remission is seen in only about one-third of patients. Visual symptoms usually precede spinal symptoms in NMO, but the reverse is not uncommon.

As outlined by Bennett, brainstem symptoms include ocular motor, motor, sensory, or cerebellar dysfunction associated with parenchymal or ependymal lesions. An area postrema syndrome (incidence: 16 %–43 %) is characterized by intractable hiccups or nausea/vomiting occurring for 7 consecutive days without, or 2 days with, an accompanying MRI lesion in the dorsal medulla. This is often reversible. The predilection for the area postrema is linked to the absence of the blood-brain barrier at this site, allowing access of large molecules such as NMO-IgG.

Diencephalic syndromes include hypersomnolence, narcolepsy, anorexia, hypothermia, hypo-natremia, and behavioral changes due to lesions in the thalamus, hypothalamus, or third ventricular region. Cerebral syndromes include hemiparesis, hemi-sensory loss, encephalopathy, postchiasmal visual field loss, and cortical vision loss due to large, confluent subcortical or deep white matter lesions. Unlike MS, a secondary progressive clinical course is quite rare.

CSF may show GFAP, reflecting the fact that NMO is an astrocytopathy. There is no role for measurement of AQP4-Ab in CSF rather than serum.

Reflecting the presence of lesions in areas with high concentration of APQ4, MRI may show gadolinium enhancement in leptomeningeal and ependymal areas. Contrasting clinical and histological features of MS, ADEM, NMO, and MOGAD are shown in Tables 5.1 and 5.2 .

Pathologic Features

NMO is an astrocytopathy rather than a pure demyelinative disorder, and CNS demyelination occurs only as a consequence of a primary destruction of astrocytes. Histologically, the lesions are more destructive, at least in late stages or severe examples, than those of MS. Early lesions may show relative myelin and axon preservation compared with the severe loss of AQP4, the latter as assessed by immunohistochemistry. NMO lesions often show granulocytes and eosinophils, cell types that are rare in MS lesions. In contrast, MS lesions usually contain lymphocytes.

In NMO lesions, there is a vasocentric deposition of IgG and IgM as well as complement (C9neo) in a rim and rosette pattern, while in at least one subtype of MS, complement is found instead in macrophages. The fact that complement was vasocentric in location lead originally to the suspicion that an antibody was targeting a nearby antigen present in perivascular spaces. It was subsequently shown that this antigen was AQP4, the most abundant water channel present on astrocytic end feet surrounding vessels. Astrocytic end feet are also abundant on the subpial surfaces, and thus NMO lesions are also found in APQ4-rich subpial regions.

In early lesions of NMO there may be prominent eosinophilic ( Fig.5.9A ) and neutrophilic infiltrates and vascular fibrosis, features that are rare in typical MS. In early NMO lesions, AQP loss as assessed by immunohistochemistry precedes astrocyte loss and some reactive astrocytosis can be found within a lesion. However, astrocytic loss precedes myelin loss, underscoring the fact that the astrocyte rather than myelin is the main target in NMO, in contrast to MS. In contrast, a key feature of early MS lesions is reactive astrocytosis with long tapering processes and plump eosinophilic cytoplasm are features that remain throughout early phases of injury. Prior to the demise of astrocytes in NMO, they may demonstrate multinucleation, mitoses, and (rarely) Rosenthal fibers. In NMO, the loss of astrocytes is evidenced by loss of GFAP immunostaining within the lesion, and GFAP + debris can be identified in perivascular macrophages that engulf the dying astrocytic debris. Astrocytes may show blunted processes, differing from the long tapering processes seen in reactive astrocytes that are typical of MS ( Fig. 5.9B ). Macrophages are present, and in subpial, choroid plexus, or ependymal areas with AQP loss, microglial activation may be evident along with loss of AQP immunostaining.

Figure 5.9, Neuromyelitis optica on biopsy may show perivenous eosinophils (A), dystrophic astrocytes with blunted processes on glial fibrillary acidic protein indicative of the disease being an astrocytoma (B) and other sites including the floor of the fourth ventricle (C) and at autopsy, cavitation of spinal cord (D).

At autopsy, the pathological features of NMO are more readily recognized as being distinctive from MS. In patients dying in early phases of the disease, the cord may be swollen, similar to a tumor, whereas in late stages, there may be considerable shrinkage of the cord and cavitation due to tissue destruction. NMO is histologically characterized by considerably greater loss of axons than is seen in typical MS, along with necrotizing demyelination, cavitation, and focally severe loss of axons in the spinal cord and optic nerves ( Fig. 5.9C ). The tissue involvement often extends over numerous spinal cord segments, paralleling the longitudinally extensive MRI findings. Cavitation in the floor of the third or fourth ventricle may occur ( Fig. 5.9D ). Granulocytes and eosinophils may be seen but can diminish in older lesions. Macrophages are present in older cavitated lesions, and in subpial, choroid plexus or ependymal areas with AQP loss, microglial activation may be evident along with loss of aquaporin immunostaining.

NEUROMYELITIS OPTICA—FACT SHEET

Definition

  • n

    Severe, idiopathic inflammatory demyelinating disorder of the spinal cord and optic nerves/chiasm, other sites, associated with AQP4-IgG

Incidence and Location

  • n

    Overrepresentation of East Asian and non-white populations worldwide, compared with multiple sclerosis

  • n

    Demyelination confined to the CNS

  • n

    Lesions occur in areas of strong APQ4 distribution (which is highly concentrated in astrocytic foot processes), including the optic nerve, spinal cord, periependymal sites in the cerebrum and cerebellum including the hypothalamus, third ventricle, corpus callosum, subependymal sites around lateral ventricles, and the floor of the fourth ventricle

You're Reading a Preview

Become a Clinical Tree membership for Full access and enjoy Unlimited articles

Become membership

If you are a member. Log in here