3474 PART 13 Neurologic Disorders
Autologous hematopoietic stem cell transplantation appears to be
highly effective in reducing the occurrence of relapses and may
improve disability in relapsing MS. It appears to be ineffective for
patients with progressive MS. Stem cell transplantation also carries
significant risk, and randomized trials with appropriate comparators
are needed in order to position this procedure with respect to available
pharmacologic interventions.
■ PROMISING EXPERIMENTAL THERAPIES
Numerous clinical trials of promising experimental therapies are currently underway. These include studies of molecules to promote remyelination; autologous hematopoietic stem cell transplantation; higher
doses of ocrelizumab; and selective kinase inhibitors including Bruton’s
tyrosine kinase (BTK).
OTHER THERAPEUTIC CLAIMS
Many purported treatments for MS have never been subjected to scientific scrutiny. These include dietary therapies (e.g., the Swank diet,
the Paleo diet, the Wahls diet), megadose vitamins, calcium orotate,
bee stings, cow colostrum, hyperbaric oxygen, procarin (a combination of histamine and caffeine), chelation, acupuncture, acupressure,
various Chinese herbal remedies, and removal of mercury-amalgam
tooth fillings, among many others. Patients should avoid costly or
potentially hazardous unproven treatments. Many such treatments lack
biologic plausibility. No reliable case of mercury poisoning resembling
typical MS has ever been described, therefore challenging the notion
that removal of mercury-amalgam tooth fillings would be beneficial.
Although potential roles for EBV, human herpesvirus (HHV) 6, or
chlamydia have been suggested for MS, treatment with antiviral agents
or antibiotics is not recommended. A chronic cerebrospinal insufficiency (CCSVI) was proposed as a cause of MS with vascular-surgical
intervention recommended. However, multiple independent studies
have failed to even approximate the initial claims, and patients should
be strongly advised to avoid diagnostic procedures and potentially dangerous surgery for this condition. A double-blind trial of high-dose biotin to improve disability in progressive forms of MS found no benefit.
SYMPTOMATIC THERAPY
For all patients, it is important to encourage attention to a healthy lifestyle, including maintaining an optimistic outlook, a healthy diet, and
regular exercise as tolerated (swimming is often well-tolerated because
of the cooling effect of cold water). It is reasonable also to correct
vitamin D deficiency with oral vitamin D.
Ataxia/tremor is often intractable. Clonazepam, 1.5–20 mg/d;
primidone, 50–250 mg/d; propranolol, 40–200 mg/d; or ondansetron,
8–16 mg/d, may help. Wrist weights occasionally reduce tremor in the
arm or hand. Thalamotomy and deep-brain stimulation have been
tried with mixed success.
Spasticity and spasms may improve with physical therapy, regular
exercise, and stretching. Avoidance of triggers (e.g., infections, fecal
impactions, bed sores) is extremely important. Effective medications
include baclofen (20–120 mg/d), diazepam (2–40 mg/d), tizanidine
(8–32 mg/d), dantrolene (25–400 mg/d), and cyclobenzaprine hydrochloride (10–60 mg/d). For severe spasticity, a baclofen pump (delivering medication directly into the CSF) can provide substantial relief.
Weakness can sometimes be improved with the use of potassium channel blockers such as 4-aminopyridine (20 mg/d) and
3,4-di-aminopyridine (40–80 mg/d), particularly in the setting where
lower-extremity weakness interferes with the patient’s ability to ambulate. The FDA approved extended-release 4-aminopyridine (at 10 mg
twice daily), and this can be obtained either as dalfampridine (Ampyra)
or through a compounding pharmacy. The principal concern with the
use of these agents is the possibility of inducing seizures at high doses.
Pain is treated with anticonvulsants (carbamazepine, 100–1000 mg/d;
phenytoin, 300–600 mg/d; gabapentin, 300–3600 mg/d; or pregabalin,
50–300 mg/d), antidepressants (amitriptyline, 25–150 mg/d; nortriptyline, 25–150 mg/d; desipramine, 100–300 mg/d; or venlafaxine,
75–225 mg/d), or antiarrhythmics (mexiletine, 300–900 mg/d). If
these approaches fail, patients should be referred to a comprehensive
pain-management program.
Bladder dysfunction management is best guided by urodynamic
testing. Evening fluid restriction or frequent voluntary voiding may
help detrusor hyperreflexia. If these methods fail, propantheline
bromide (10–15 mg/d), oxybutynin (5–15 mg/d), hyoscyamine sulfate (0.5–0.75 mg/d), tolterodine tartrate (2–4 mg/d), or solifenacin
(5–10 mg/d) may help. Coadministration of pseudoephedrine
(30–60 mg) is sometimes beneficial.
Detrusor/sphincter dyssynergia may respond to phenoxybenzamine
(10–20 mg/d) or terazosin hydrochloride (1–20 mg/d). Loss of reflex
bladder wall contraction may respond to bethanechol (30–150 mg/d).
However, both conditions often require catheterization.
Urinary tract infections should be treated promptly. Patients with
postvoid residual urine volumes >200 mL are predisposed to infections. Prevention by urine acidification (with cranberry juice or
vitamin C) inhibits some bacteria. Prophylactic administration of antibiotics is sometimes necessary but may lead to colonization by resistant
organisms. Intermittent catheterization may help to prevent recurrent
infections and reduce overflow incontinence.
Treatment of constipation includes high-fiber diets and fluids. Natural or other laxatives may help. Fecal incontinence may respond to a
reduction in dietary fiber.
Depression should be treated. Useful drugs include the selective
serotonin reuptake inhibitors (fluoxetine, 20–80 mg/d, or sertraline,
50–200 mg/d), the tricyclic antidepressants (amitriptyline, 25–150 mg/d;
nortriptyline, 25–150 mg/d; or desipramine, 100–300 mg/d), and the
nontricyclic antidepressants (venlafaxine, 75–225 mg/d).
Fatigue may improve with assistive devices, help in the home, or
successful management of spasticity. Patients with frequent nocturia
may benefit from anticholinergic medication at bedtime. Excessive
daytime somnolence caused by MS may respond to amantadine
(200 mg/d), methylphenidate (5–25 mg/d), modafinil (100–400 mg/d),
or armodafinil (150–250 mg/d).
Cognitive problems may respond marginally to lisdexamfetamine
(40 mg/d).
Paroxysmal symptoms respond dramatically to low-dose anticonvulsants (acetazolamide, 200–600 mg/d; carbamazepine, 50–400 mg/d;
phenytoin, 50–300 mg/d; or gabapentin, 600–1800 mg/d).
Heat sensitivity may respond to heat avoidance, air-conditioning, or
cooling garments.
Sexual dysfunction may be helped by lubricants to aid in genital
stimulation and sexual arousal. Management of pain, spasticity, fatigue,
and bladder/bowel dysfunction may also help. Sildenafil (50–100 mg),
tadalafil (5–20 mg), or vardenafil (5–20 mg), taken 1–2 h before sex,
are standard treatments for erectile dysfunction.
CLINICAL VARIANTS OF MS
Acute MS (Marburg’s variant) is a fulminant demyelinating process
that in some cases progresses inexorably to death within 1–2 years.
Typically, there are no remissions. Marburg’s variant does not seem to
follow infection or vaccination, and it is unclear whether this syndrome
represents an extreme form of MS or another disease altogether. When
an acute demyelinating syndrome presents as a solitary expansile
lesion, a brain tumor is often suspected (Fig. 444-4). Such cases are
designated tumefactive MS, and a brain biopsy may be required to
establish the diagnosis.
Balo’s concentric sclerosis is another fulminant demyelinating syndrome characterized by concentric brain or spinal cord lesions with
alternating spheres of demyelination and remyelination (Fig. 444-4).
For these fulminant demyelinating states, no controlled trials of therapy exist; high-dose glucocorticoids, plasma exchange, and cyclophosphamide have been tried, with uncertain benefit.
ACUTE DISSEMINATED
ENCEPHALOMYELITIS (ADEM)
ADEM has a monophasic course and is most frequently associated
with an antecedent infection (postinfectious encephalomyelitis); ~5%
of ADEM cases follow immunization (postvaccinal encephalomyelitis).
ADEM is far more common in children than adults, and many adult
cases initially thought to represent ADEM subsequently experience
3475 Multiple Sclerosis CHAPTER 444
A B
C D
FIGURE 444-4 Magnetic resonance imaging findings in variants of MS. A and B. Acute tumefactive MS. In A, a sagittal T2-weighted fluid-attenuated inversion recovery
(FLAIR) image of a large solitary right parieto-occipital white matter lesion is shown, with effacement of overlying cortical sulci consistent with mass effect. In B,
T1-weighted image obtained after the intravenous administration of gadolinium DTPA reveals a large serpiginous area of blood-brain barrier disruption consistent with
acute inflammation. C and D. Balo’s concentric sclerosis. In C, an axial T2-weighted sequence shows multiple areas of abnormal ovoid bright signal in the supratentorial
white matter bilaterally; some lesions reveal concentric layers, typical of Balo’s concentric sclerosis. In D, T1-weighted MR images postgadolinium demonstrate abnormal
enhancement of all lesions with some lesions demonstrating concentric ring enhancement.
late relapses qualifying as either MS or another chronic inflammatory
disorder such as vasculitis, sarcoidosis, or lymphoma. The hallmark of
ADEM is the presence of widely scattered foci of perivenular inflammation and demyelination that can involve both white matter and grey
matter structures, in contrast to larger confluent white matter lesions
typical of MS. In the most explosive form of ADEM, acute hemorrhagic
leukoencephalitis, the lesions are vasculitic and hemorrhagic, and the
clinical course is devastating.
Postinfectious encephalomyelitis is most frequently associated with
the viral exanthems of childhood. Infection with measles virus is
the most common antecedent (1 in 1000 cases). Worldwide, measles
encephalomyelitis is still common, although use of the live measles
3476 PART 13 Neurologic Disorders
vaccine has dramatically reduced its incidence in developed countries.
An ADEM-like illness rarely follows vaccination with live measles
vaccine (1–2 in 106
immunizations). ADEM is now most frequently
associated with varicella (chickenpox) infections (1 in 4000–10,000
cases). It may also follow infection with rubella, mumps, influenza,
parainfluenza, EBV, HHV-6, HIV, dengue, Zika, other viruses, and
Mycoplasma pneumoniae. Recently, cases have been described in
association with COVID-19 infection. Some patients may have a nonspecific upper respiratory infection or no known antecedent illness. In
addition to measles, postvaccinal encephalomyelitis may also follow
the administration of vaccines for smallpox (5 cases per million), the
Semple rabies, and Japanese encephalitis. Modern vaccines that do not
require viral culture in CNS tissue have reduced the ADEM risk.
All forms of ADEM presumably result from a cross-reactive immune
response to the infectious agent or vaccine that then triggers an inflammatory demyelinating response. Autoantibodies to MBP and to other
myelin antigens have been detected in the CSF from some patients
with ADEM, and approximately half of children with ADEM have
circulating and CSF antibodies against MOG (Chap. 445). Attempts to
demonstrate direct viral invasion of the CNS have been unsuccessful.
CLINICAL MANIFESTATIONS
In severe cases, onset is abrupt and progression rapid (hours to days). In
postinfectious ADEM, the neurologic syndrome generally begins late
in the course of the viral illness as the exanthem is fading. Fever reappears, and headache, meningismus, and lethargy progressing to coma
may develop. Seizures are common. Signs of disseminated neurologic
disease are consistently present (e.g., hemiparesis or quadriparesis,
extensor plantar responses, lost or hyperactive tendon reflexes, sensory loss, and brainstem involvement). In ADEM due to chickenpox,
cerebellar involvement is often conspicuous. CSF protein is modestly
elevated (0.5–1.5 g/L [50–150 mg/dL]). Lymphocytic pleocytosis,
generally ≥200 cells/μL, occurs in 80% of patients. Occasional patients
have higher counts or a mixed polymorphonuclear-lymphocytic pattern during the initial days of the illness. Transient CSF oligoclonal
banding has been reported. MRI usually reveals extensive changes in
the brain and spinal cord, consisting of white matter hyperintensities
on T2 and fluid-attenuated inversion recovery (FLAIR) sequences with
Gd enhancement on T1-weighted sequences.
DIAGNOSIS
The diagnosis is most reliably established when there is a history
of recent vaccination or viral exanthematous illness. In severe cases
with predominantly cerebral involvement, acute encephalitis due to
infection with herpes simplex or other viruses including HIV may
be difficult to exclude; other considerations include hypercoagulable
states including the antiphospholipid antibody syndrome, autoimmune
(paraneoplastic) limbic encephalitis, vasculitis, neurosarcoid, primary
CNS lymphoma, or metastatic cancer. An explosive presentation of
MS can mimic ADEM, and, especially in adults, it may not be possible
to distinguish these conditions at onset. The simultaneous onset of
disseminated symptoms and signs is common in ADEM and rare in
MS. Similarly, meningismus, drowsiness, coma, and seizures suggest
ADEM rather than MS. Unlike MS, in ADEM, optic nerve involvement
is generally bilateral and transverse myelopathy complete. MRI findings that favor ADEM include extensive and relatively symmetric white
matter abnormalities, basal ganglia or cortical gray matter lesions,
and Gd enhancement of all abnormal areas. By contrast, OCBs in the
CSF are more common in MS. In one study of adult patients initially
thought to have ADEM, 30% experienced additional relapses over a
follow-up period of 3 years, and they were reclassified as having MS.
Other patients initially classified as ADEM are subsequently found to
have neuromyelitis optica spectrum disorder (Chap. 445). Occasional
patients with “recurrent ADEM” have also been reported, especially
children; however, it is not possible to distinguish this entity from atypical MS. Because of the clinical overlap at presentation between ADEM
and MS, it is crucial that routine surveillance imaging be performed
following recovery from ADEM so that subclinical disease activity due
to MS can be recognized and treatment for MS initiated.
TREATMENT
■ ACUTE DISSEMINATED ENCEPHALOMYELITIS
Initial treatment is with high-dose glucocorticoids; depending on the
response, treatment may need to be continued for 8 weeks. Patients
who fail to respond within a few days may benefit from a course of
plasma exchange or intravenous immunoglobulin. The prognosis
reflects the severity of the underlying acute illness. In recent case series
of presumptive ADEM in adults, mortality rates of 5–20% are reported,
and many survivors have permanent neurologic sequelae.
GLIAL FIBRILLARY ACIDIC PROTEIN
(GFAP) AUTOIMMUNITY
Autoimmunity against the astrocyte protein GFAP presents with a
range of symptoms referable to meningismus, encephalitis, myelitis,
and optic neuritis. MRI shows characteristic patterns of gadolinium
enhancement localized to GFAP-enriched CNS regions including
venous structures in a periventricular radial orientation, the leptomeninges, the peri-ependymal spinal cord, and a striking serpiginous
pattern involving brain parenchyma. These enhancement patterns
share some similarities with patterns that can be observed in neurosarcoidosis. The presence of these patterns should prompt consideration
for either condition. A lymphocytic pleocytosis is commonly present
in the CSF. Antibodies against GFAP can be measured in the CSF or
serum. GFAP autoimmunity is found as a paraneoplastic syndrome
in 25% of cases, most commonly associated with ovarian teratoma,
and can coexist with anti-N-methyl-d-aspartate receptor (NMDAR)
encephalitis or neuromyelitis optica spectrum disorder (NMOSD).
T cells are implicated in pathophysiology based on histopathology
and association with checkpoint inhibitor treatment for cancer or in
the setting of HIV. GFAP autoimmunity is generally glucocorticoid
responsive. Early recognition with prompt intervention is associated
with more favorable outcomes. Relapses occur in approximately 20%
of patients and require use of immune suppression therapy.
■ FURTHER READING
Absinta M et al: Mechanisms underlying progression in multiple sclerosis. Curr Opin Neurol 33:277, 2020.
Bevan RJ et al: Meningeal inflammation and cortical demyelination in
acute multiple sclerosis. Ann Neurol 84:829, 2018.
Brown JWL et al: Association of initial disease-modifying therapy
with later conversion to secondary progressive multiple sclerosis.
JAMA 321:175, 2019.
Cree BAC et al: Silent progression in disease activity-free relapsing
multiple sclerosis. Ann Neurol 85:653, 2019.
Giovannoni G et al: Alemtuzumab improves preexisting disability
in active relapsing-remitting MS patients. Neurology 87:1985, 2016.
Hauser SL et al: Ocrelizumab versus interferon beta-1a in relapsing
multiple sclerosis. N Engl J Med 376:221, 2017.
Hauser SL et al: Ofatumumab versus teriflunomide in multiple sclerosis. N Engl J Med 383:546, 2020.
Luna G et al: Infection risks among patients with multiple sclerosis
treated with fingolimod, natalizumab, rituximab, and injectable therapies. JAMA Neurol 77:184, 2020.
Malpas CB et al: Early clinical markers of aggressive multiple sclerosis.
Brain 143:1400, 2020.
Naegelin Y et al: Association of rituximab treatment with disability
progression among patients with secondary progressive multiple
sclerosis. JAMA Neurol 76:274, 2019.
Montalban X et al: Ocrelizumab versus placebo in primary progressive multiple sclerosis. N Engl J Med 376:209, 2017.
Pohl D et al: Acute disseminated encephalomyelitis: Updates on an
inflammatory CNS syndrome. Neurology 87(9 Suppl 2):S38, 2016.
Shan F et al: Autoimmune glial fibrillary acidic protein astrocytopathy:
A review of the literature. Front Immunol 9:2802, 2018.
Thompson AJ et al: Diagnosis of multiple sclerosis: 2017 revisions of
the McDonald criteria. Lancet Neurol 17:162, 2018.
Zamvil SS, Hauser SL: Antigen presentation by B cells in multiple
sclerosis. [Review] N Engl J Med 384:378, 2021.
3477 Neuromyelitis Optica CHAPTER 445
INTRODUCTION
Neuromyelitis optica (NMO; Devic’s disease) is an aggressive inflammatory disorder characterized by recurrent attacks of optic neuritis
(ON) and myelitis; the more inclusive term NMO spectrum disorder
(NMOSD) was proposed to include individuals with partial forms, and
also those with involvement of additional regions in the central nervous system (Table 445-1). NMO is more frequent in women than men
(9:1), and typically begins in adulthood, with a mean age of onset of
40 years, but can arise at any age. An important consideration, especially early in its presentation, is distinguishing NMO from multiple
sclerosis (MS; Chap. 444). In patients with NMO, attacks of ON can be
bilateral and produce severe visual loss (uncommon in MS); myelitis
can be severe and transverse (rare in MS) and is typically longitudinally
extensive (Fig. 445-1), involving three or more contiguous vertebral
segments. In contrast to MS, progressive symptoms typically do not
occur in NMO. The brain MRI was earlier thought to be normal in
NMO, but it is now recognized that in many cases brain lesions are
present, including areas of nonspecific signal change as well as lesions
445
associated with specific syndromes such as the hypothalamus causing
an endocrinopathy; the area postrema in the lower medulla presenting
as intractable hiccups or vomiting; or the cerebral hemispheres producing focal symptoms, encephalopathy, or seizures. Large MRI lesions
in the cerebral hemispheres can be asymptomatic, sometimes have a
“cloudlike” appearance and, unlike MS lesions, are often not destructive
and can resolve completely. Spinal cord MRI lesions typically consist of
focal enhancing areas of swelling and tissue destruction, extending
over three or more spinal cord segments, and on axial sequences, these
are centered on the grey matter of the cord. Cerebrospinal fluid (CSF)
findings include pleocytosis greater than that observed in MS, with
neutrophils and eosinophils present in many acute cases; oligoclonal
bands (OCBs) are uncommon, occurring in <20% of NMO patients.
The pathology of NMO is a distinctive astrocytopathy with inflammation, loss of astrocytes, and an absence of staining of the water channel
protein AQP4 by immunohistochemistry, plus thickened blood vessel
walls, demyelination, and deposition of antibody and complement.
IMMUNOLOGY
NMO is an autoimmune disease associated with a highly specific autoantibody directed against aquaporin-4 (AQP-4) that is present in the
sera of ~90% of patients with a clinical diagnosis of NMO. AQP-4 is
localized to the foot processes of astrocytes in close apposition to endothelial surfaces, as well as at paranodal regions near nodes of Ranvier.
It is likely that AQP-4 antibodies are pathogenic because their passive
transfer into laboratory animals can reproduce histologic features
of the disease. Antibody-mediated complement fixation is thought
to represent the primary mechanism of astrocyte injury in NMO.
During acute attacks of myelitis, CSF levels of interleukin-6 (IL-6; a
proinflammatory cytokine) and astrocyte-specific glial fibrillary acidic
protein (GFAP) levels are markedly elevated, consistent with active
inflammation and astrocyte injury. Proinflammatory T-lymphocytes
of the Th17 type recognize an immunodominant epitope of AQP4 and
may also contribute to pathogenesis. Because of the high specificity of
the antibody, its presence is considered to be diagnostic when found
in conjunction with a typical clinical presentation. Anti-AQP4 seropositive patients have a high risk for future relapses; more than half of
untreated patients will relapse within 1 year.
CLINICAL COURSE
NMO is typically a recurrent disease; the course is monophasic in <10%
of patients. Individuals who test negative for AQP4 antibodies are somewhat more likely to have a monophasic course. Untreated NMO is usually
quite disabling over time; in one series, respiratory failure from cervical
myelitis was present in one-third of patients, and 8 years after onset,
60% of patients were blind and more than half had permanent paralysis
of one or more limbs. The long-term course of NMO appears to have
been substantially improved with the development of therapies to treat
acute attacks and prevent relapses. Estimates of the 5-year survival rate
increased from 68–75% in 1999 to 91–98% in 2017, a change likely due
to improved diagnosis and widespread use of immunosuppressant drugs.
GLOBAL CONSIDERATIONS
The incidence and prevalence of NMO show considerable variation
between populations and geographic regions, with prevalence estimates that range from <1 to > 4 per 100,000. Although NMO can occur
in people of any ethnic background, individuals of Asian and African
origin are disproportionately affected. The highest reported prevalence
is from Martinique. Among white populations, MS (Chap. 444) is far
more common than NMO.
Interestingly, when MS affects individuals of African or Asian
ancestry, there is a propensity for demyelinating lesions to involve
predominantly the optic nerve and spinal cord, an MS subtype termed
opticospinal MS. Some individuals with opticospinal MS are seropositive for AQP4 antibodies, indicating that such cases represent NMOSD.
ASSOCIATED CONDITIONS
Up to 40% of NMO patients have a systemic autoimmune disorder, such
as systemic lupus erythematosus, Sjögren’s syndrome, perinuclear antineutrophil cytoplasmic antibody (p-ANCA)–associated vasculitis, myasthenia
Neuromyelitis Optica
Bruce A. C. Cree, Stephen L. Hauser
TABLE 445-1 Diagnostic Criteria for Neuromyelitis Optica
Spectrum Disorder
Diagnostic Criteria for NMOSD with AQP4-IgG
1. At least 1 core clinical characteristic
2. Positive test for AQP4-IgG using best-available detection method (cell-based
assay strongly recommended)
3. Exclusion of alternative diagnoses
Diagnostic Criteria for NMOSD Without AQP4-IgG or NMOSD with
Unknown AQP4-IgG Status
1. At least 2 core clinical characteristics occurring as a result of one or more
clinical attacks and meeting all of the following requirements:
a. At least 1 core clinical characteristic must be optic neuritis, acute myelitis
with LETM, or area postrema syndrome
b. Dissemination in space (2 or more different clinical characteristics)
c. Fulfillment of additional MRI requirements, as applicable
2. Negative test for AQP4-IgG using best-available detection method or testing
unavailable
3. Exclusion of alternative diagnoses
Core Clinical Characteristics
1. Optic neuritis
2. Acute myelitis
3. Area postrema syndrome: episode of otherwise unexplained hiccups or
nausea or vomiting
4. Acute brainstem syndrome
5. Symptomatic narcolepsy or acute diencephalic clinical syndrome with
NMOSD-typical diencephalic MRI lesions
6. Symptomatic cerebral syndrome with NMOSD-typical brain lesions
Additional MRI Requirements for NMOSD Without AQP4-IgG and
NMOSD with Unknown AQP4-IgG Status
1. Acute optic neuritis: requires brain MRI showing (a) normal findings
or only nonspecific white matter lesions, OR (b) optic nerve MRI with
T2-hyperintense lesion of T1-weighted gadolinium-enhancing lesion
extending over >1/2 optic nerve length or involving optic chiasm
2. Acute myelitis: requires associated intramedullary MRI lesion extending ≥3
contiguous segments (LETM) OR ≥3 contiguous segments of focal spinal cord
atrophy in patients with history compatible with acute myelitis
3. Area postrema syndrome requires associated dorsal medulla/area postrema
lesions
4. Acute brainstem syndrome requires periependymal brainstem lesions
Source: Reproduced with permission from DM Wingerchuk et al: International
consensus diagnostic criteria for neuromyelitis optica spectrum disorders.
Neurology 85:177, 2015.
3478 PART 13 Neurologic Disorders
A B
C D
E F
FIGURE 445-1 Imaging findings in neuromyelitis optica: longitudinally extensive transverse myelitis, optic neuritis, and brainstem involvement. A. Sagittal fluid attenuation
inversion recovery (FLAIR) cervical-spine MRI showing an area of increased signal change on T2-weighted imaging spanning >3 vertebral segments in length. B. Sagittal
T1-weighted cervical-spine MRI following gadolinium-diethylene triamine pentaacetic acid (DPTA) infusion showing enhancement. C. Coronal brain MRI shows hyperintense
signal on FLAIR imaging within the left optic nerve. D. Coronal T1-weighted brain MRI following gadolinium-DPTA infusion shows enhancement of the left optic nerve. E.
Axial brain MRI shows an area of hyperintense signal on T2-weighted imaging within the area postrema (arrow). F. Axial T1-weighted brain MRI following gadolinium-DPTA
infusion shows punctate enhancement of the area postrema (arrow).
gravis, Hashimoto’s thyroiditis, or mixed connective tissue disease. This is
another feature distinct from MS; MS patients rarely have other comorbid
autoimmune diseases with the exception of hypothyroidism. In some
NMO cases, onset may be associated with acute infection with varicella
zoster virus, Epstein-Barr virus, HIV, or tuberculosis. Rare cases appear to
be paraneoplastic and associated with breast, lung, or other cancers.
TREATMENT
Neuromyelitis Optica
Until recently, disease-modifying therapies were not rigorously studied in NMO. Acute attacks are usually treated
with high-dose glucocorticoids (e.g., methylprednisolone
1 g/d for 5–10 days followed by a prednisone taper). Plasma
exchange (typically 5–7 exchanges of 1.5 plasma volumes/exchange)
is used empirically for acute episodes that do not respond to glucocorticoids. Given the unfavorable natural history of untreated NMO,
prophylaxis against relapses is recommended for most patients
and several empiric regimens have been commonly used including: mycophenolate mofetil (1000 mg bid); the B-cell depleting
anti-CD20 monoclonal antibody rituximab (2 g IV Q 6 months); or
a combination of glucocorticoids (500 mg IV methylprednisolone
daily for 5 days; then oral prednisone 1 mg/kg per day for 2 months,
followed by slow taper) plus azathioprine (2 mg/kg per day started
on week 3). Importantly, some therapies with efficacy in MS do
not appear to be useful for NMO. Available evidence suggests that
interferon beta is ineffective and paradoxically may increase the
3479 Neuromyelitis Optica CHAPTER 445
risk of NMO relapses, and based on limited data glatiramer acetate,
fingolimod, natalizumab, and alemtuzumab also appear to be ineffective. These differences highlight the importance of distinguishing NMO from MS.
Three monoclonal antibody therapies have now received regulatory approval for attack prevention in NMO: eculizumab, a terminal
complement inhibitor; inebilizumab, a B-cell depleter; and satralizumab, an IL-6 receptor blocker (Table 445-2).
Eculizumab is a monoclonal antibody that binds to the complement protein C5, inhibiting its cleavage into C5a and C5b and
preventing generation of the terminal complement attack complex
C5b-9. Investigated as add-on therapy in AQP4 seropositive NMO,
eculizumab lengthened the time to first attack by 94%, reduced the
attack rate by 96%, and reduced rates of hospitalization, glucocorticoid, and plasma exchange use. Eculizumab is dosed as follows:
900 mg weekly for the first 4 weeks, followed by 1200 mg for the
fifth dose 1 week later, then 1200 mg every 2 weeks thereafter.
Eculizumab is available only through a restricted program under
a Risk Evaluation and Mitigation Strategy (REMS). Life-threatening and fatal meningococcal infections have occurred in eculizumab-treated patients (Boxed Warning). Eculizumab-treated
patients should be immunized with meningococcal vaccines at
least 2 weeks prior to administering the first dose unless the
risk of delaying eculizumab therapy outweigh the risk of developing a meningococcal infection. Vaccination reduces, but
does not eliminate, the risk of meningococcal infections. All
eculizumab-treated patients must be monitored for early signs of
meningococcal infections and evaluated immediately if infection
is suspected.
Inebilizumab is a humanized monoclonal antibody that binds
to the B-cell-specific surface antigen CD19 and depletes a wide
range of B cells including some plasmablasts as well as a proportion
of plasma cells in secondary lymphoid organs and bone marrow.
Inebilizumab used as monotherapy reduced time to the first NMO
attack by 77% compared to placebo, and also reduced hospitalizations by 78%, disability worsening by 63%, and new MRI lesions by
43%. Inebilizumab is dosed as follows: 300 mg IV infusion followed
2 weeks later by a second 300 mg IV infusion with subsequent doses
of 300 mg infusions every 6 months thereafter.
Inebilizumab is associated with a dose-dependent decline in
serum IgG levels and with neutropenia in some patients.
Satralizumab is a monoclonal antibody that binds to the
interleukin-6 receptor, blocking engagement of IL-6. Satralizumab
was investigated in NMOSD in two trials: one as monotherapy and
the other as add-on therapy. Both AQP4 seropositive and AQP4
seronegative participants were enrolled. In both studies, the time to
first attack was longer with satralizumab treatment compared with
placebo. The risk of attack was reduced by 74% in the monotherapy
study, and in the add-on study by 78%, with satralizumab. Although
both studies recruited substantial numbers of AQP4 seronegative
participants, there was no clinically meaningful impact of satralizumab in the seronegative participants. Satralizumab is administered as follows: a loading dosage of 120 mg by subcutaneous
injection at weeks 0, 2, and 4, followed by a maintenance dosage of
120 mg every 4 weeks.
Screening for hepatitis B virus, tuberculosis, and liver transaminase elevations is required before starting satralizumab.
Hepatic transaminases should be monitored during treatment for
transaminase elevation, and CBC should be monitored during
treatment for neutropenia. Satralizumab is also associated with
weight gain.
MYELIN OLIGODENDROCYTE
GLYCOPROTEIN-ANTIBODY-ASSOCIATED
DISEASE (MOGAD)
Although long considered to be a likely target for antibody-mediated
demyelination, anti-MOG antibodies detected by a cell-based assay
that enables recognition of myelin oligodendrocyte glycoprotein
(MOG) epitopes in a lipid bilayer were recently found to be associated with cases of acute disseminated encephalomyelitis (ADEM)
(Chap. 444) in children, and then with cases of AQP4 seronegative
NMO. Further studies showed that patients who are seropositive for
anti-MOG antibodies are at risk for bilateral, synchronous optic neuritis and myelitis. A clinical feature that can help distinguish ON associated with MOGAD from NMO or MS is the presence of papillitis seen
by funduscopy or orbital MRI. ON associated with MOGAD is typically longitudinally extensive on MRI, and brain MRI can be normal
or show fluffy areas of increased signal change in white or grey matter
structures, similar to NMO. MRI lesions that are typical for MS, including finger-like lesions oriented perpendicular to the ventricular surface
(Dawson fingers) and T1-hypointense lesions, are uncommon. Spinal
cord lesions can be longitudinally extensive or short and sometimes
involve the conus medullaris. Demyelination associated with MOGAD
is sometimes monophasic, as in ADEM, but can also be recurrent. The
CSF may show a pleocytosis with occasional neutrophils. Elevated
intrathecal synthesis of gammaglobulins is atypical: oligoclonal bands
are present in ~6–13% of cases and intrathecal synthesis of anti-MOG
antibodies does not occur. The mechanism of CNS injury in MOGAD
is not established. Studies in MOG-induced experimental autoimmune
encephalomyelitis suggest that anti-MOG antibodies may opsonize
traces of MOG protein in secondary lymphoid tissues, triggering an
encephalitogenic peripheral immune response.
Acute episodes are managed with high-dose glucocorticoids followed by a prednisone taper and sometimes by plasmapheresis, as with
NMO. Brain lesions associated with MOGAD often respond rapidly to
treatment with glucocorticoids and may resolve entirely. Some patients
experience disease recurrence following discontinuation of prednisone
and can become glucocorticoid dependent. Clinical trials have not
been undertaken and there is limited data on other immune-suppressing
medications typically used in NMO. Off-label empiric treatments
include daily prednisone, IVIg, rituximab, and mycophenolate mofetil.
Anti-MOG antibody titers appear to decline either spontaneously or in
the setting of treatment.
Rare cases of relapsing optic nerve and spinal cord disease resembling
NMOSD have recently been recognized in patients with autoantibodies against glial fibrillary acidic protein (GFAP), an astrocyte-specific
protein, although more commonly the disorder presents as a meningoencephalitis resembling acute disseminated encephalomyelitis. GFAP
astrocytopathy is discussed in Chap. 444.
■ FURTHER READING
Cree BAC et al: Inebilizumab for the treatment of neuromyelitis optica
spectrum disorder (N-MOmentum): A double-blind, randomised
placebo-controlled phase 2/3 trial. Lancet 394:1352, 2019.
Hinson SR et al: Autoimmune AQP4 channelopathies and neuromyelitis optica spectrum disorders. Handb Clin Neurol 133:377, 2016.
Marignier R et al: Myelin-oligodendrocyte glycoprotein antibodyassociated disease. Lancet Neurol 20:762, 2021.
Pittock SJ et al: Eculizumab in aquaporin-4-positive neuromyelitis
optica spectrum disorder. N Engl J Med 381:614, 2019.
Traboulsee A et al: Safety and efficacy of satralizumab monotherapy in neuromyelitis optica spectrum disorder: a randomised,
double-blind, multicentre, placebo-controlled phase 3 trial. Lancet
Neurol 19:402, 2020.
Wingerchuk DM et al: International consensus diagnostic criteria for
neuromyelitis optica spectrum disorders. Neurology 85:177, 2015.
TABLE 445-2 Therapeutic Trials for NMO
RISK REDUCTION IN AQP4-
SEROPOSITIVE PATIENTS
Eculizumab (add-on to immune
suppression)
94%, P<0.001
Inolimomab (monotherapy) 78%, P=0.01
Satralizumab (add-on to immune
suppression)
74%, P=0.001
Satralizumab (monotherapy) 77%, P<0.001
3480 PART 13 Neurologic Disorders
Peripheral nerves are composed of sensory, motor, and autonomic
elements. Diseases can affect the cell body of a neuron or its peripheral processes, namely the axons or the encasing myelin sheaths.
Most peripheral nerves are mixed and contain sensory and motor as
well as autonomic fibers. Nerves can be subdivided into three major
classes: large myelinated, small myelinated, and small unmyelinated. Motor axons are usually large myelinated fibers that conduct
rapidly (~50 m/s). Sensory fibers may be any of the three types.
Large-diameter sensory fibers conduct proprioception and vibratory
sensation to the brain, while the smaller-diameter myelinated and
unmyelinated fibers transmit pain and temperature sensation. Autonomic nerves are also small in diameter. Thus, peripheral neuropathies can impair sensory, motor, or autonomic function, either singly
or in combination. Peripheral neuropathies are further classified
into those that primarily affect the cell body (e.g., neuronopathy or
ganglionopathy), myelin (myelinopathy), and the axon (axonopathy). These different classes of peripheral neuropathies have distinct
clinical and electrophysiologic features. This chapter discusses the
clinical approach to a patient suspected of having a peripheral
neuropathy, as well as specific neuropathies, including hereditary
and acquired neuropathies. The inflammatory neuropathies are
discussed in Chap. 447.
GENERAL APPROACH
In approaching a patient with a neuropathy, the clinician has three main
goals: (1) identify where the lesion is, (2) identify the cause, and (3) determine the proper treatment. The first goal is accomplished by obtaining
a thorough history, neurologic examination, and electrodiagnostic and
other laboratory studies (Fig. 446-1). While gathering this information,
seven key questions are asked (Table 446-1), the answers to which help
identify the pattern of involvement and the cause of the neuropathy
(Table 446-2). Despite an extensive evaluation, in approximately half of
patients, no etiology is ever found; these patients typically have a predominately sensory polyneuropathy and have been labeled as having idiopathic or cryptogenic sensory and sensorimotor polyneuropathy (CSPN).
■ INFORMATION FROM THE HISTORY AND
PHYSICAL EXAMINATION: SEVEN KEY
QUESTIONS (TABLE 446-1)
1. What Systems Are Involved? It is important to determine if
the patient’s symptoms and signs are motor, sensory, autonomic, or a
combination of these. If the patient has only weakness without any evidence of sensory or autonomic dysfunction, a motor neuropathy, neuromuscular junction abnormality, or myopathy should be considered.
Some peripheral neuropathies are associated with significant autonomic
nervous system dysfunction. Symptoms of autonomic involvement
include fainting spells or orthostatic lightheadedness; heat intolerance;
or any bowel, bladder, or sexual dysfunction (Chap. 440). There will
typically be an orthostatic fall in blood pressure without an appropriate
increase in heart rate. Autonomic dysfunction in the absence of diabetes
should alert the clinician to the possibility of amyloid polyneuropathy.
Rarely, a pandysautonomic syndrome can be the only manifestation of
a peripheral neuropathy without other motor or sensory findings. The
majority of neuropathies are predominantly sensory in nature.
Section 3 Nerve and Muscle Disorders
446 Peripheral Neuropathy
Anthony A. Amato, Richard J. Barohn
Patient Complaint: ? Neuropathy
History and examination compatible with neuropathy?
Mononeuropathy Mononeuropathy multiplex Polyneuropathy Evaluation of other
disorder or
reassurance and
follow-up
EDx
Is the lesion axonal
or demyelinating?
Is entrapment or
compression present?
Is a contributing systemic
disorder present?
Axonal Demyelinating
with focal
conduction block
Axonal Demyelinating
Consider
vasculitis or
other multifocal
process
Consider
multifocal
form of
CIDP
Subacute
course (months)
Chronic
course (years) Uniform slowing,
chronic
Nonuniform slowing,
conduction block
Decision on need
for surgery (nerve repair,
transposition, or release
procedure)
Possible
nerve
biopsy Test for paraprotein,
HIV, Lyme disease
Review history for toxins;
test for associated
systemic disease or
intoxication
Test for paraprotein,
if negative
Review family
history; examine
family members;
genetic testing
If chronic or
subacute: CIDP
If acute: GBS
Treatment appropriate
for specific diagnosis If tests are
negative, consider
treatment for
CIDP
Treatment appropriate
for specific diagnosis
Genetic counseling if appropriate
Treatment
for CIDP;
see Ch. 447
IVIg or
plasmapheresis;
supportive
care including
respiratory assistance
EDx EDx
Yes No
FIGURE 446-1 Approach to the evaluation of peripheral neuropathies. CIDP, chronic inflammatory demyelinating polyradiculoneuropathy; EDx, electrodiagnostic; GBS,
Guillain-Barré syndrome; IVIg, intravenous immunoglobulin.
3481Peripheral Neuropathy CHAPTER 446
2. What Is the Distribution of Weakness? Delineating the
pattern of weakness, if present, is essential for diagnosis, and in this
regard, two additional questions should be answered: (1) Does the
weakness only involve the distal extremity, or is it both proximal and
distal? and (2) Is the weakness focal and asymmetric, or is it symmetric? Symmetric proximal and distal weakness is the hallmark of
acquired immune demyelinating polyneuropathies, both the acute
form (Guillain-Barré syndrome [GBS]) and the chronic form (chronic
inflammatory demyelinating polyneuropathy [CIDP]) (Chap. 447).
The importance of finding symmetric proximal and distal weakness in
a patient who presents with both motor and sensory symptoms cannot
be overemphasized because this identifies the important subset of
patients who may have a treatable acquired demyelinating neuropathic
disorder (i.e., GBS or CIDP).
Findings of an asymmetric or multifocal pattern of weakness narrow
the differential diagnosis. Some neuropathic disorders may present
with unilateral extremity weakness. In the absence of sensory symptoms and signs, such weakness evolving over weeks or months would
be worrisome for motor neuron disease (e.g., amyotrophic lateral sclerosis [ALS]), but it would be important to exclude multifocal motor
neuropathy that may be treatable (Chap. 447). In a patient presenting
with asymmetric subacute or acute sensory and motor symptoms and
signs, radiculopathies, plexopathies, compressive mononeuropathies,
or multiple mononeuropathies (e.g., mononeuropathy multiplex) must
be considered.
ALS can produce prominent neck extensor weakness (head drop),
tongue and pharyngeal weakness (dysarthria and dysphagia), or
shortness of breath. These focal symmetric weakness patterns can
also be seen in neuromuscular junction disorders (myasthenia gravis,
Lambert-Eaton myasthenic syndrome [LEMS] [Chap. 448]) and some
myopathies, particularly isolated neck extensor myopathy (Chap. 449).
3. What Is the Nature of the Sensory Involvement? The
patient may have loss of sensation (numbness), altered sensation to
touch (hyperpathia or allodynia), or uncomfortable spontaneous sensations (tingling, burning, or aching) (Chap. 25). Neuropathic pain can
be burning, dull, and poorly localized (protopathic pain), presumably
transmitted by polymodal C nociceptor fibers, or sharp and lancinating
TABLE 446-1 Approach to Neuropathic Disorders:
Seven Key Questions
1. What systems are involved?
• Motor, sensory, autonomic, or combinations
2. What is the distribution of weakness?
• Only distal versus proximal and distal
• Focal/asymmetric versus symmetric
3. What is the nature of the sensory involvement?
• Temperature loss or burning or stabbing pain (e.g., small fiber)
• Vibratory or proprioceptive loss (e.g., large fiber)
4. Is there evidence of upper motor neuron involvement?
• Without sensory loss
• With sensory loss
5. What is the temporal evolution?
• Acute (days to 4 weeks)
• Subacute (4–8 weeks)
• Chronic (>8 weeks)
• Monophasic, progressive, or relapsing-remitting
6. Is there evidence for a hereditary neuropathy?
• Family history of neuropathy
• Lack of sensory symptoms despite sensory signs
7. Are there any associated medical conditions?
• Cancer, diabetes mellitus, connective tissue disease or other autoimmune
diseases, infection (e.g., HIV, Lyme disease, leprosy)
• Medications including over-the-counter drugs that may cause a toxic
neuropathy
• Preceding events, drugs, toxins
TABLE 446-2 Patterns of Neuropathic Disorders
Pattern 1: Symmetric proximal and distal weakness with sensory loss
Consider: inflammatory demyelinating polyneuropathy (GBS and CIDP)
Pattern 2: Symmetric distal sensory loss with or without distal weakness
Consider: cryptogenic or idiopathic sensory polyneuropathy (CSPN), diabetes
mellitus and other metabolic disorders, drugs, toxins, familial (HSAN), CMT,
amyloidosis, and others
Pattern 3: Asymmetric distal weakness with sensory loss
With involvement of multiple nerves
Consider: multifocal CIDP, vasculitis, cryoglobulinemia, amyloidosis, sarcoid,
infectious (leprosy, Lyme, hepatitis B, C, or E, HIV, CMV), HNPP, tumor infiltration
With involvement of single nerves/regions
Consider: may be any of the above but also could be compressive
mononeuropathy, plexopathy, or radiculopathy
Pattern 4: Asymmetric proximal and distal weakness with sensory loss
Consider: polyradiculopathy or plexopathy due to diabetes mellitus, meningeal
carcinomatosis or lymphomatosis, sarcoid, amyloid, hereditary plexopathy (HNPP,
HNA), idiopathic
Pattern 5: Asymmetric distal weakness without sensory loss
With upper motor neuron findings
Consider: motor neuron disease
Without upper motor neuron findings
Consider: progressive muscular atrophy, juvenile monomelic amyotrophy
(Hirayama’s disease), multifocal motor neuropathy, multifocal acquired motor
axonopathy
Pattern 6: Symmetric sensory loss and distal areflexia with upper motor neuron
findings
Consider: vitamin B12, vitamin E, and copper deficiency with combined system
degeneration with peripheral neuropathy, chronic liver disease, hereditary
leukodystrophies (e.g., adrenomyeloneuropathy) HSP-plus
Pattern 7: Symmetric weakness without sensory loss
With proximal and distal weakness
Consider: SMA
With distal weakness
Consider: hereditary motor neuropathy (“distal” SMA) or atypical CMT
Pattern 8: Focal midline proximal symmetric weakness
Neck extensor weakness
Consider: ALS
Bulbar weakness
Consider: ALS/PLS, isolated bulbar ALS (IBALS), Kennedy’s syndrome (X-linked,
bulbospinal SMA), bulbar presentation GBS
Diaphragm weakness (SOB)
Consider: ALS
Pattern 9: Asymmetric proprioceptive sensory loss without weakness
Consider causes of a sensory neuronopathy (ganglionopathy):
Cancer (paraneoplastic)
Sjögren’s syndrome
Idiopathic sensory neuronopathy (possible GBS variant)
Cisplatin and other chemotherapeutic agents
Vitamin B6
toxicity
HIV-related sensory neuronopathy
Pattern 10: Autonomic symptoms and signs
Consider neuropathies associated with prominent autonomic dysfunction:
Hereditary sensory and autonomic neuropathy
Amyloidosis (familial and acquired)
Diabetes mellitus
GBS
Idiopathic pandysautonomia (may be a variant of GBS)
Porphyria
HIV-related autonomic neuropathy
Vincristine and other chemotherapeutic agents
Abbreviations: ALS, amyotrophic lateral sclerosis; CIDP, chronic inflammatory
demyelinating polyneuropathy; CMT, Charcot-Marie-Tooth disease; CMV,
cytomegalovirus; GBS, Guillain-Barré syndrome; HIV, human immunodeficiency
virus; HNA, hereditary neuralgic amyotrophy; HNPP, hereditary neuropathy with
liability to pressure palsies; HSAN, hereditary sensory and autonomic neuropathy;
HSP-plus, hereditary spastic paraplegia plus neuropathy; PLS, primary lateral
sclerosis; SMA, spinal muscular atrophy; SOB, shortness of breath.
3482 PART 13 Neurologic Disorders
(epicritic pain), relayed by A-delta fibers. If pain and temperature perception are lost, while vibratory and position sense are preserved along
with muscle strength, deep tendon reflexes, and normal nerve conduction studies (NCS), a small-fiber neuropathy is likely. The most likely
causes of small-fiber neuropathies, when one is identified, are diabetes
mellitus (DM) or glucose intolerance. Amyloid neuropathy should be
considered as well in such cases, but most of these small-fiber neuropathies remain idiopathic despite extensive evaluation.
Severe proprioceptive loss also narrows the differential diagnosis.
Affected patients will note imbalance, especially in the dark. A neurologic examination revealing a dramatic loss of proprioception with
vibration loss and normal strength should alert the clinician to consider a sensory neuronopathy/ganglionopathy (Pattern 9, Table 446-2).
In particular, if this loss is asymmetric or affects the arms more than
the legs, this pattern suggests a non-length-dependent process as seen
in sensory neuronopathies.
4. Is There Evidence of Upper Motor Neuron Involvement?
If the patient presents with symmetric distal sensory symptoms and
signs suggestive of a distal sensory neuropathy, but there is additional
evidence of symmetric upper motor neuron involvement (Chap. 24),
the physician should consider a combined system degeneration with
neuropathy. The most common cause for this pattern is vitamin B12
deficiency, but other etiologies should also be considered (e.g., copper
deficiency, human immunodeficiency virus [HIV] infection, severe
hepatic disease, adrenomyeloneuropathy [AMN]), and hereditary
spastic paraplegia plus a neuropathy.
5. What Is the Temporal Evolution? It is important to determine the onset, duration, and evolution of symptoms and signs. Does
the disease have an acute (days to 4 weeks), subacute (4–8 weeks), or
chronic (>8 weeks) course? Is the course monophasic, progressive, or
relapsing? Most neuropathies are insidious and slowly progressive in
nature. Neuropathies with acute and subacute presentations include
GBS, vasculitis, and radiculopathies related to diabetes or Lyme disease. A relapsing course can be present in CIDP and porphyria.
6. Is There Evidence for a Hereditary Neuropathy? In
patients with slowly progressive distal weakness over many years
with few sensory symptoms yet significant sensory deficits on clinical
examination, the clinician should consider a hereditary neuropathy
(e.g., Charcot-Marie-Tooth disease [CMT]). On examination, the feet
may show high or flat arches or hammer toes, and scoliosis may be
present. In suspected cases, it may be necessary to perform neurologic
and electrophysiologic studies on family members in addition to the
patient.
7. Does the Patient Have Any Other Medical Conditions? It
is important to inquire about associated medical conditions (e.g.,
DM, systemic lupus erythematosus [SLE]); preceding or concurrent
infections (e.g. diarrheal illness preceding GBS); surgeries (e.g., gastric
bypass and nutritional neuropathies); medications (toxic neuropathy),
including over-the-counter vitamin preparations (B6
); alcohol; dietary
habits; and use of dentures (e.g., fixatives contain zinc that can lead to
copper deficiency).
■ PATTERN RECOGNITION APPROACH TO
NEUROPATHIC DISORDERS
Based on the answers to the seven key questions, neuropathic disorders can be classified into several patterns based on the distribution or
pattern of sensory, motor, and autonomic involvement (Table 446-2).
Each pattern has a limited differential diagnosis, and information from
laboratory studies usually permits a final diagnosis to be established.
■ ELECTRODIAGNOSTIC STUDIES
The electrodiagnostic (EDx) evaluation of patients with a suspected
peripheral neuropathy consists of NCS and needle electromyography
(EMG). In addition, studies of autonomic function can be valuable. The
electrophysiologic data can confirm whether the neuropathic disorder
is a mononeuropathy, multiple mononeuropathy (mononeuropathy
multiplex), radiculopathy, plexopathy, or generalized polyneuropathy.
Similarly, EDx evaluation can ascertain whether the process involves
only sensory fibers, motor fibers, autonomic fibers, or a combination
of these. Finally, the electrophysiologic data can help distinguish
axonopathies from myelinopathies as well as axonal degeneration secondary to ganglionopathies from the more common length-dependent
axonopathies.
NCS are most helpful in classifying a neuropathy as due to axonal
degeneration or segmental demyelination (Table 446-3). In general,
low-amplitude potentials with relatively preserved distal latencies,
conduction velocities, and late potentials, along with fibrillations on
needle EMG, suggest an axonal neuropathy. On the other hand, slow
conduction velocities, prolonged distal latencies and late potentials,
relatively preserved amplitudes, and the absence of fibrillations on
needle EMG imply a primary demyelinating neuropathy. The presence
of nonuniform slowing of conduction velocity, conduction block, or
temporal dispersion further suggests an acquired demyelinating neuropathy (e.g., GBS or CIDP) as opposed to a hereditary demyelinating
neuropathy (e.g., CMT type 1).
Autonomic studies are used to assess small myelinated (A-delta)
or unmyelinated (C) nerve fiber involvement. Such testing includes
heart rate response to deep breathing, heart rate, and blood pressure
response to both the Valsalva maneuver and tilt-table testing and quantitative sudomotor axon reflex testing (Chap. 440). These studies are
particularly useful in patients who have pure small-fiber neuropathy or
autonomic neuropathy in which routine NCS are normal.
■ OTHER IMPORTANT LABORATORY
INFORMATION
In patients with generalized symmetric peripheral neuropathy, a standard laboratory evaluation should include a complete blood count,
basic chemistries including serum electrolytes and tests of renal and
hepatic function, fasting blood glucose (FBS), hemoglobin (Hb) A1c,
urinalysis, thyroid function tests, B12, folate, erythrocyte sedimentation
rate (ESR), rheumatoid factor, antinuclear antibodies (ANA), serum
protein electrophoresis (SPEP) and immunoelectrophoresis or immunofixation, and urine for Bence Jones protein. Quantification of the
TABLE 446-3 Electrophysiologic Features: Axonal Degeneration versus
Segmental Demyelination
AXONAL DEGENERATION
SEGMENTAL
DEMYELINATION
Motor Nerve Conduction Studies
CMAP amplitude Decreased Normal (except with CB
or distal dispersion)
Distal latency Normal Prolonged
Conduction velocity Normal Slow
Conduction block Absent Present
Temporal dispersion Absent Present
F wave Normal or absent Prolonged or absent
H reflex Normal or absent Prolonged or absent
Sensory Nerve Conduction Studies
SNAP amplitude Decreased Normal or decreased
Distal latency Normal Prolonged
Conduction velocity Normal Slow
Needle EMG
Spontaneous activity
Fibrillations Present Absent
Fasciculations Present Absent
Motor unit potentials
Recruitment Decreased Decreased
Morphology Long duration, large
amplitude, polyphasic
(if there is reinnervation)
Normal
Abbreviations: CB, conduction block; CMAP, compound motor action potential;
EMG, electromyography; SNAP, sensory nerve action potential.
3483Peripheral Neuropathy CHAPTER 446
concentration of serum-free light chains and the kappa/lambda ratio is
more sensitive than SPEP, immunoelectrophoresis, or immunofixation
to detect a monoclonal gammopathy and therefore should be done if
amyloidosis is suspected. A skeletal survey should be performed in
patients with acquired demyelinating neuropathies and M-spikes to
look for osteosclerotic or lytic lesions. Patients with monoclonal gammopathy should also be referred to a hematologist for consideration
of a bone marrow biopsy. An oral glucose tolerance test is indicated in
patients with painful sensory neuropathies even if FBS and HbA1c are
normal, as the test is abnormal in about one-third of such patients. In
addition to the above tests, patients with a mononeuropathy multiplex
pattern of involvement should have a vasculitis workup, including
antineutrophil cytoplasmic antibodies (ANCAs), cryoglobulins, hepatitis serology, Western blot for Lyme disease, HIV, and occasionally a
cytomegalovirus (CMV) titer.
There are many autoantibody panels (various antiganglioside antibodies) marketed for screening routine neuropathy patients for a treatable condition. These autoantibodies have no proven clinical utility
or added benefit beyond the information obtained from a complete
clinical examination and detailed EDx. A heavy metal screen is also
not necessary as a screening procedure, unless there is a history of
possible exposure or suggestive features on examination (e.g., severe
painful sensorimotor and autonomic neuropathy and alopecia—
thallium; severe painful sensorimotor neuropathy with or without
gastrointestinal [GI] disturbance and Mee’s lines—arsenic; wrist or
finger extensor weakness and anemia with basophilic stippling of red
blood cells—lead).
In patients with suspected GBS or CIDP, a lumbar puncture is
indicated to look for an elevated cerebrospinal fluid (CSF) protein. In
idiopathic cases of GBS and CIDP, CSF pleocytosis is usually absent.
If cells are present, one should consider HIV infection, Lyme disease,
sarcoidosis, or lymphomatous or leukemic infiltration of nerve roots.
Recently, serum IgG4 antibodies to neurofascin and contactin-2 have
been discovered in CIDP with severe sensory ataxia, tremor, and distal
weakness (Chap. 447). These cases are difficult to treat with standard
immunotherapies but may respond to rituximab. Some patients with
GBS and CIDP have abnormal liver function tests. In these cases,
it is important to also check for hepatitis B and C, HIV, CMV, and
Epstein-Barr virus (EBV) infection. In patients with an axonal
GBS (by EMG/NCS) or those with a suspicious coinciding history
(e.g., unexplained abdominal pain, psychiatric illness, significant autonomic dysfunction), it is reasonable to screen for porphyria.
In patients with a severe sensory ataxia, a sensory ganglionopathy
or neuronopathy should be considered. The most common causes of
sensory ganglionopathies are Sjögren’s syndrome (Chap. 361) and a
paraneoplastic neuropathy (Chap. 94). Neuropathy can be the initial
manifestation of Sjögren’s syndrome. Thus, one should always inquire
about dry eyes and mouth in patients with sensory signs and symptoms.
Further, some patients can manifest sicca complex without other manifestations of Sjögren’s syndrome. Thus, patients with sensory ataxia
should be tested for antibodies to SS-A/Ro and SS-B/La, in addition to
the routine ANA. To evaluate a possible paraneoplastic sensory ganglionopathy, antineuronal nuclear antibodies (e.g., anti-Hu antibodies)
should be obtained. These antibodies are most commonly seen in
patients with small-cell carcinoma of the lung but are also present with
breast, ovarian, lymphoma, and other cancers. Importantly, the paraneoplastic neuropathy can precede the detection of the cancer, and detection of these autoantibodies should lead to a search for malignancy.
■ NERVE BIOPSIES
Nerve biopsies are now rarely performed in the evaluation of neuropathies. The primary indication for nerve biopsy is suspicion for amyloid
neuropathy or vasculitis. In most instances, the abnormalities present
on biopsies do not help distinguish one form of peripheral neuropathy
from another (beyond what is already apparent by clinical examination
and the NCS). Nerve biopsies should only be performed when the NCS
are abnormal. The sural nerve is most commonly biopsied because
it is a pure sensory nerve and biopsy will not result in loss of motor
function. In suspected vasculitis, a combination biopsy of a superficial
peroneal nerve (pure sensory) and the underlying peroneus brevis
muscle obtained from a single small incision increases the diagnostic
yield. Tissue can be analyzed to assess for evidence of inflammation,
vasculitis, or amyloid deposition. Semithin plastic sections, teased
fiber preparations, and electron microscopy are used to assess the
morphology of the nerve fibers and to distinguish axonopathies from
myelinopathies.
■ SKIN BIOPSIES
Skin biopsies are sometimes used to diagnose a small-fiber neuropathy.
Following a punch biopsy of the skin in the distal lower extremity,
immunologic staining can be used to measure the density of small
unmyelinated fibers. The density of these nerve fibers is reduced in
patients with small-fiber neuropathies in whom NCS and routine nerve
biopsies are often normal. This technique may allow for an objective
measurement in patients with mainly subjective symptoms. However,
it often adds little to what one already knows from the clinical examination and EDx.
SPECIFIC DISORDERS
■ HEREDITARY NEUROPATHIES
CMT disease is the most common type of hereditary neuropathy
(Pattern 2, Table 446-2). Rather than one disease, CMT is a syndrome
of many genetically distinct disorders (Table 446-4). The various
subtypes of CMT are classified according to the nerve conduction
velocities (NCVs) and predominant pathology (e.g., demyelination
or axonal degeneration), inheritance pattern (autosomal dominant,
recessive, or X-linked), and the specific mutated genes. Type 1 CMT
(or CMT1) refers to inherited demyelinating sensorimotor neuropathies, whereas the axonal sensory neuropathies are classified as CMT2.
By definition, motor conduction velocities in the arms are slowed to
<38 m/s in CMT1 and are >38 m/s in CMT2. However, most cases of
CMT1 actually have motor NCVs between 20 and 25 m/s. CMT1 and
CMT2 usually begin in childhood or early adult life; however, onset
later in life can occur, particularly in CMT2. Both are inherited in an
autosomal dominant fashion, with a few exceptions. CMT3 is an autosomal dominant neuropathy that appears in infancy and is associated
with severe demyelination or hypomyelination. CMT4 is an autosomal
recessive neuropathy that typically begins in childhood or early adult
life. There are no medical therapies for any of the CMTs, but physical
and occupational therapy can be beneficial, as can bracing (e.g., anklefoot orthotics for foot drop) and other orthotic devices.
■ CMT1
CMT1 is the most common form of hereditary neuropathy. Affected
individuals usually present in the first to third decade of life with distal
leg weakness (e.g., foot drop), although patients may remain asymptomatic even late in life. People with CMT generally do not complain
of numbness or tingling, which can be helpful in distinguishing CMT
from acquired forms of neuropathy in which sensory symptoms usually predominate. Although usually asymptomatic, reduced sensation
to all modalities is apparent on examination. Muscle stretch reflexes
are unobtainable or reduced throughout. There is often atrophy of the
muscles below the knee (particularly the anterior compartment), leading to so-called inverted champagne bottle legs.
Motor NCVs are generally in the 20–25 m/s range. Nerve biopsies
usually are not performed on patients suspected of having CMT1,
because the diagnosis usually can be made by less invasive testing (e.g.,
NCS and genetic studies). However, when done, the biopsies reveal
reduced numbers of myelinated nerve fibers with a predilection for loss
of large-diameter fibers and Schwann cell proliferation around thinly
or demyelinated fibers, forming so-called onion bulbs.
CMT1A is the most common subtype of CMT1, representing 70% of
cases, and is caused by a 1.5-megabase (Mb) duplication within chromosome 17p11.2-12 encoding the gene for peripheral myelin protein-22
(PMP-22). This results in patients having three copies of the PMP-22
gene rather than two. This protein accounts for 2–5% of myelin
protein and is expressed in compact regions of the peripheral myelin
sheath. Approximately 20% of patients with CMT1 have CMT1B,
3484 PART 13 Neurologic Disorders
TABLE 446-4 Classification of Charcot-Marie-Tooth Disease and Related Neuropathies
NAME INHERITANCE GENE LOCATION GENE PRODUCT
CMT1
CMT1A AD 17p11.2 PMP-22 (usually duplication of gene)
CMT1B AD 1q21-23 MPZ
CMT1C AD 16p13.1-p12.3 LITAF
CMT1D AD 10q21.1-22.1 ERG2
CMT1E (with deafness) AD 17p11.2 Point mutations in PMP-22 gene
CMT1F AD 8p13-21 Neurofilament light chain
CMT1G AD 8q21 PMP2
CMT1X X-linked dominant Xq13 Connexin-32
HNPP AD 17p11.2 PMP-22
1q21-23 MPZ
CMT dominant-intermediate (CMTDI)
CMT-DIA AD 10q24.1-25.1 ?
CMT-DIB AD 19.p12-13.2 Dynamin 2
CMT-DIC AD 1p35 YARS
CMT-DID
CMT-DIE
CMT-DIF
CMT-DIG
AD
AD
AD
AD
1q22
14q32.33
3q26
8p31
MPZ
IFN-2
GNB4
NEFL
CMT recessive-intermediate (CMT-RI)
CMT-RIA
CMT-RIB
CMT-RIC
CMT-RI D
AR
AR
AR
AR
8q21.1
6q23
1p36
12q24
GDAP1
KARS5
PLEKHG5
COX6A1
CMT2
CMT2A2 (allelic to HMSN VI with optic atrophy) AD 1p36.2 MFN2
CMT2B AD 3q13-q22 RAB7
CMT2B1 (allelic to LGMD 1B) AR 1q21.2 Lamin A/C
CMT2B2 AR and AD 19q13 MED25 for AR; unknown for AD
CMT2C (with vocal cord and diaphragm paralysis) AD 12q23-24 TRPV4
CMT2D (allelic to distal SMA5) AD 7p14 Glycine tRNA synthetase
CMT2E (allelic to CMT 1F) AD 8p21 Neurofilament light chain
CMT2F AD 7q11-q21 Heat-shock 27-kDa protein-1
CMT2G AD 9q31.3-34.2 LRSAM1
CMT2I (allelic to CMT1B) AD 1q22 MPZ
CMT2J AD 1q22 MPZ
CMT2H, CMT2K (allelic to CMT4A) AD 8q13-q21 GDAP1
CMT2L (allelic to distal hereditary motor
neuropathy type 2)
AD 12q24 Heat-shock protein 8
CMT2M AD 16q22 Dynamin-2
CMT2N AD 16q22.1 AARS
CMT2O AD 14q32.31 DYNC1H1
CMT2P AD 9q31.3-34.2 LRSAM1
CMT2P-Okinawa (HSMN2P) AD 3q13-q14 TFG
CMT2Q
CMT2U
CMT2V
CMT2W
CMT2Y
CMT2Z
CMT2X
AD
AD
AD
AD
AD
AD
X-linked
10p14
12q13
17q11
5q31
9p13
22q12
Xq22-24
DHTKD1
MARS
NAGLU
HARS
VCP
MRC2
PRPS1
CMT3 AD 17p11.2 PMP-22
(Dejerine-Sottas disease, congenital
hypomyelinating neuropathy)
AD 1q21-23 MPZ
AR 10q21.1-22.1 ERG2
AR 19q13 Periaxon
(Continued)
3485Peripheral Neuropathy CHAPTER 446
caused by mutations in the myelin protein zero (MPZ). CMT1B is
for the most part clinically, electrophysiologically, and histologically
indistinguishable from CMT1A. MPZ is an integral myelin protein and
accounts for more than half of the myelin protein in peripheral nerves.
Other forms of CMT1 are much less common and also indistinguishable from one another clinically and electrophysiologically.
■ CMT2
CMT2 occurs approximately half as frequently as CMT1, and CMT2
tends to present later in life. Affected individuals usually become symptomatic in the second decade; some cases present earlier in childhood,
whereas others remain asymptomatic into late adult life. Clinically, CMT2
is for the most part indistinguishable from CMT1. NCS are helpful in this
regard; in contrast to CMT1, the velocities are normal or only slightly
slowed. The most common cause of CMT2 is a mutation in the gene for
mitofusin 2 (MFN2), which accounts for ~20–30% of CMT2 cases overall.
MFN2 localizes to the outer mitochondrial membrane, where it regulates
the mitochondrial network architecture by participating in mitochondrial
fusion. The other genes associated with CMT2 are much less common.
■ CMTDI
In dominant-intermediate CMTs (CMTDIs), the NCVs are faster than
usually seen in CMT1 (e.g., >38 m/s) but slower than in CMT2.
■ CMT3
CMT3 was originally described by Dejerine and Sottas as a hereditary
demyelinating sensorimotor polyneuropathy presenting in infancy or
early childhood. Affected children are severely weak. Motor NCVs are
markedly slowed, typically ≤5–10 m/s. Most cases of CMT3 are caused
by point mutations in the genes for PMP-22, MPZ, or ERG-2, which
are also the genes responsible for CMT1.
■ CMT4
CMT4 is extremely rare and is characterized by a severe, childhoodonset sensorimotor polyneuropathy that is usually inherited in an
autosomal recessive fashion. Electrophysiologic and histologic evaluations can show demyelinating or axonal features. CMT4 is genetically
heterogeneous (Table 446-4).
■ CMT1X
CMT1X is an X-linked dominant disorder with clinical features similar
to CMT1 and CMT2, except that the neuropathy is much more severe
in males than in females. CMT1X accounts for ~10–15% of CMT
overall. Males usually present in the first two decades of life with atrophy and weakness of the distal arms and legs, areflexia, pes cavus, and
hammer toes. Obligate female carriers are frequently asymptomatic
TABLE 446-4 Classification of Charcot-Marie-Tooth Disease and Related Neuropathies
NAME INHERITANCE GENE LOCATION GENE PRODUCT
CMT4
CMT4A AR 8q13-21.1 GDAP1
CMT4B1 AR 11q23 MTMR2
CMT4B2 AR 11p15 MTMR13
CMT4C AR 5q23-33 SH3TC2
CMT4D (HMSN-Lom) AR 8q24 NDRG1
CMT4E (congenital hypomyelinating neuropathy) AR Multiple Includes PMP-22, MPZ, and ERG-2
CMT4F AR 19q13.1-13.3 Periaxin
CMT4G AR 10q23.2 HK1
CMT4H AR 12q12-q13 Frabin
CMT4J
CMT4K
AR
AR
6q21
9q34
FIG4
SURF1
HNA AD 17q24 SEPT9
HSAN1A AD 9q22 SPTLC1
HSAN1B AD 3q21 RAB7
HSAN1C AD 14q24.3 SPTLC2
HSAN1D AD 14q21.3 ATL1
HSAN1E AD 19p13.2 DNMT1
HSAN2A AR 12p13.33 PRKWNK1
HSAN2B AR 5p15.1 FAM134B
HSAN2C AR 12q13.13 KIF1A
HSAN2D AR 2q24.3 SCN9A
HSAN3A AR 9q21 IKAP
HSAN3B AR 6p12.1 Dystonin
HSAN4 AR 3q trkA/NGF receptor
HSAN5 AD or AR 1p11.2-p13.2
2q24.3
3p22.2
NGFb
SCN9A
SCN11A
HSAN6 AR 6p12.1 Dystonin
Abbreviations: AARS, alanyl-tRNA synthetase; AD, autosomal dominant; AR, autosomal recessive; ATL, atlastin; CMT, Charcot-Marie-Tooth; DNMT1, DNA methyltransferase 1;
DYNC1HI, cytoplasmic dynein 1 heavy chain 1; ERG2, early growth response-2 protein; FAM134B, family with sequence similarity 134, member B; FIG4, FDG1-related F
actin-binding protein; GDAP1, ganglioside-induced differentiation-associated protein-1; HK1, hexokinase 1; HMSN-P, hereditary motor and sensory neuropathy proximal;
HNA, hereditary neuralgic amyotrophy; HNPP, hereditary neuropathy with liability to pressure palsies; HSAN, hereditary sensory and autonomic neuropathy; IFN2, inverted
formin-2; IKAP, k
B kinase complex-associated protein; LGMD, limb girdle muscular dystrophy; LITAF, lipopolysaccharide-induced tumor necrosis factor α factor; LRSAM1, E3
ubiquitin-protein ligase; MED25, mediator 25; MFN2, mitochondrial fusion protein mitofusin 2 gene; MPZ, myelin protein zero protein; MTMR2, myotubularin-related protein-2;
NDRG1, N-myc downstream regulated 1; PMP-22, peripheral myelin protein-22; PRKWNK1, protein kinase, lysine deficient 1; PRPS1, phosphoribosylpyrophosphate
synthetase 1; RAB7, Ras-related protein 7; SEPT9, Septin 9; SH3TC2, SH3 domain and tetratricopeptide repeats 2; SMA, spinal muscular atrophy; SPTLC, serine
palmitoyltransferase long-chain base; TFG, TRK-fused gene; TrkA/NGF, tyrosine kinase A/nerve growth factor; tRNA, transfer ribonucleic acid; TRPV4, transient receptor
potential cation channel, subfamily V, member 4; WNK1, WNK lysine deficient; YARS, tyrosyl-tRNA synthetase.
Source: Modified from AA Amato, J Russell: Neuromuscular Disorders, 2nd ed. New York, McGraw-Hill, 2016, Table 11-1, pp 265–266.
(Continued)
3486 PART 13 Neurologic Disorders
but can develop signs and symptoms of CMT. Onset in females is
usually after the second decade of life, and the neuropathy is milder in
severity.
NCS reveal features of both demyelination and axonal degeneration.
In males, motor NCVs in the arms and legs are moderately slowed
(in the low to mid 30-m/s range). About 50% of males with CMT1X
have motor NCVs between 15 and 35 m/s with ~80% of these falling
between 25 and 35 m/s (intermediate slowing). In contrast, ~80% of
females with CMT1X have NCVs in the normal range and 20% have
NCVs in the intermediate range. CMT1X is caused by mutations in the
connexin-32 gene. Connexins are gap junction structural proteins that
are important in cell-to-cell communication.
Hereditary Neuropathy with Liability to Pressure Palsies
(HNPP) HNPP is an autosomal dominant disorder related to
CMT1A. While CMT1A is usually associated with a 1.5-Mb duplication in chromosome 17p11.2 that results in an extra copy of PMP-22
gene, HNPP is caused by inheritance of the chromosome with the
corresponding 1.5-Mb deletion of this segment, and thus, affected
individuals have only one copy of the PMP-22 gene. Patients usually
manifest in the second or third decade of life with painless numbness
and weakness in the distribution of single peripheral nerves, although
multiple mononeuropathies can occur (Pattern 3, Table 446-2).
Symptomatic mononeuropathy or multiple mononeuropathies are
often precipitated by trivial compression of nerve(s) as can occur
with wearing a backpack, leaning on the elbows, or crossing one’s
legs for even a short period of time. These pressure-related mononeuropathies may take weeks or months to resolve. In addition, some
affected individuals manifest with a progressive or relapsing, generalized and symmetric, sensorimotor peripheral neuropathy that
resembles CMT.
Hereditary Neuralgic Amyotrophy (HNA) HNA is an autosomal dominant disorder characterized by recurrent attacks of pain,
weakness, and sensory loss in the distribution of the brachial plexus
often beginning in childhood (Pattern 4, Table 446-2). These attacks
are similar to those seen with idiopathic brachial plexitis (see below).
Attacks may occur in the postpartum period, following surgery, or
at other times of stress. Most patients recover over several weeks or
months. Slightly dysmorphic features, including hypotelorism, epicanthal folds, cleft palate, syndactyly, micrognathia, and facial asymmetry,
are evident in some individuals. EDx demonstrate an axonal process.
HNA is genetically heterogeneous but can be caused by mutations in
septin 9 (SEPT9). Septins may be important in formation of the neuronal cytoskeleton and have a role in cell division, but it is not known
how mutations in SEPT9 lead to HNA.
Hereditary Sensory and Autonomic Neuropathy (HSAN)
The HSANs are a very rare group of hereditary neuropathies in which
sensory and autonomic dysfunction predominates over muscle weakness, unlike CMT, in which motor findings are most prominent (Pattern 2,
Table 446-2; Table 446-4). Nevertheless, affected individuals can develop
motor weakness, and there can be overlap with CMT. There are no medical therapies available to treat these neuropathies, other than prevention and treatment of mutilating skin and bone lesions.
Of the HSANs, only HSAN1 typically presents in adults. HSAN1
is the most common of the HSANs and is inherited in an autosomal
dominant fashion. Affected individuals usually manifest in the second through fourth decades of life. HSAN1 is associated with the
degeneration of small myelinated and unmyelinated nerve fibers
leading to severe loss of pain and temperature sensation, deep dermal ulcerations, recurrent osteomyelitis, Charcot joints, bone loss,
gross foot and hand deformities, and amputated digits. Although
most people with HSAN1 do not complain of numbness, they often
describe burning, aching, or lancinating pains. Autonomic neuropathy is not a prominent feature, but bladder dysfunction and reduced
sweating in the feet may occur. HSAN1A, which is most common,
is caused by mutations in the serine palmitoyltransferase long-chain
base 1 (SPTLC1) gene.
OTHER HEREDITARY NEUROPATHIES
(TABLE 446-5)
■ FABRY’S DISEASE
Fabry’s disease (angiokeratoma corporis diffusum) is an X-linked
dominant disorder. Although men are more commonly and severely
affected, women can also manifest symptoms and signs of the disease.
Angiokeratomas are reddish-purple maculopapular lesions that are
usually found around the umbilicus, scrotum, inguinal region, and
perineum. Burning or lancinating pain in the hands and feet often
develops in males in late childhood or early adult life (Pattern 2,
Table 446-2). However, the neuropathy is usually overshadowed by
complications arising from an associated premature atherosclerosis
(e.g., hypertension, renal failure, cardiac disease, and stroke) that often
lead to death by the fifth decade of life. Some patients also manifest
primarily with a dilated cardiomyopathy.
Fabry’s disease is caused by mutations in the α-galactosidase gene
that leads to the accumulation of ceramide trihexoside in nerves
and blood vessels. A decrease in α-galactosidase activity is evident
in leukocytes and cultured fibroblasts. Glycolipid granules may be
appreciated in ganglion cells of the peripheral and sympathetic nervous
systems and in perineurial cells. Enzyme replacement therapy with
α-galactosidase B can improve the neuropathy if patients are treated
early, before irreversible nerve fiber loss develops.
■ ADRENOLEUKODYSTROPHY/
ADRENOMYELONEUROPATHY
Adrenoleukodystrophy (ALD) and AMN are allelic X-linked dominant
disorders caused by mutations in the peroxisomal transmembrane adenosine triphosphate-binding cassette (ABC) transporter gene. Patients
with ALD manifest with central nervous system (CNS) abnormalities.
However, ~30% of patients with mutations in this gene present with the
AMN phenotype that typically manifests in the third to fifth decade
of life as mild to moderate peripheral neuropathy combined with
TABLE 446-5 Rare Hereditary Neuropathies
Hereditary Disorders of Lipid Metabolism
Metachromatic leukodystrophy
Krabbe’s disease (globoid cell leukodystrophy)
Fabry’s disease
Adrenoleukodystrophy/adrenomyeloneuropathy
Refsum’s disease
Tangier disease
Cerebrotendinous xanthomatosis
Hereditary Ataxias with Neuropathy
Friedreich’s ataxia
Vitamin E deficiency
Spinocerebellar ataxia
Abetalipoproteinemia (Bassen-Kornzweig disease)
Disorders of Defective DNA Repair
Ataxia-telangiectasia
Cockayne’s syndrome
Giant Axonal Neuropathy
Porphyria
Acute intermittent porphyria (AIP)
Hereditary coproporphyria (HCP)
Variegate porphyria (VP)
Familial Amyloid Polyneuropathy (FAP)
Transthyretin-related
Gelsolin-related
Apolipoprotein A1-related
Source: Modified from AA Amato, J Russell: Neuromuscular Disorders, 2nd ed.
New York, McGraw-Hill, 2016, Table 12-1, p. 299.
3487Peripheral Neuropathy CHAPTER 446
progressive spastic paraplegia (Pattern 6, Table 446-2) (Chap. 442).
Rare patients present with an adult-onset spinocerebellar ataxia or only
with adrenal insufficiency.
EDx is suggestive of a primary axonopathy with secondary demyelination. Nerve biopsies demonstrate a loss of myelinated and unmyelinated nerve fibers with lamellar inclusions in the cytoplasm of
Schwann cells. Very-long-chain fatty acid (VLCFA) levels (C24, C25,
and C26) are increased in the urine. Laboratory evidence of adrenal
insufficiency is evident in approximately two-thirds of patients. The
diagnosis can be confirmed by genetic testing.
Adrenal insufficiency is managed by replacement therapy; however,
there is no proven effective therapy for the neurologic manifestations
of ALD/AMN. Diets low in VLCFAs and supplemented with Lorenzo’s
oil (erucic and oleic acids) reduce the levels of VLCFAs and increase
the levels of C22 in serum, fibroblasts, and liver; however, several large,
open-label trials of Lorenzo’s oil failed to demonstrate efficacy.
■ REFSUM’S DISEASE
Refsum’s disease can manifest in infancy to early adulthood with the
classic tetrad of (1) peripheral neuropathy, (2) retinitis pigmentosa,
(3) cerebellar ataxia, and (4) elevated CSF protein concentration.
Most affected individuals develop progressive distal sensory loss and
weakness in the legs leading to foot drop by their twenties (Pattern 2,
Table 446-2). Subsequently, the proximal leg and arm muscles may
become weak. Patients may also develop sensorineural hearing loss,
cardiac conduction abnormalities, ichthyosis, and anosmia.
Serum phytanic acid levels are elevated. Sensory and motor NCS
reveal reduced amplitudes, prolonged latencies, and slowed conduction
velocities. Nerve biopsy demonstrates a loss of myelinated nerve fibers,
with remaining axons often thinly myelinated and associated with
onion bulb formation.
Refsum’s disease is genetically heterogeneous but autosomal recessive in nature. Classical Refsum’s disease with childhood or early adult
onset is caused by mutations in the gene that encodes for phytanoylCoA α-hydroxylase (PAHX). Less commonly, mutations in the gene
encoding peroxin 7 receptor protein (PRX7) are responsible. These
mutations lead to the accumulation of phytanic acid in the central
and peripheral nervous systems. Treatment is removal of phytanic
precursors (phytols: fish oils, dairy products, and ruminant fats) from
the diet.
■ TANGIER DISEASE
Tangier disease is a rare autosomal recessive disorder that can present as (1) asymmetric multiple mononeuropathies, (2) a slowly
progressive symmetric polyneuropathy predominantly in the legs, or
(3) a pseudo-syringomyelia pattern with dissociated sensory loss
(i.e., abnormal pain/temperature perception but preserved position/
vibration in the arms [Chap. 442]). The tonsils may appear swollen
and yellowish-orange in color, and there may also be splenomegaly and
lymphadenopathy.
Tangier disease is caused by mutations in the ATP-binding cassette
transporter 1 (ABC1) gene, which leads to markedly reduced levels of
high-density lipoprotein (HDL) cholesterol levels, whereas triacylglycerol levels are increased. Nerve biopsies reveal axonal degeneration
with demyelination and remyelination. Electron microscopy demonstrates abnormal accumulation of lipid in Schwann cells, particularly
those encompassing unmyelinated and small myelinated nerves. There
is no specific treatment.
■ PORPHYRIA
Porphyria is a group of inherited disorders caused by defects in heme
biosynthesis (Chap. 416). Three forms of porphyria are associated
with peripheral neuropathy: acute intermittent porphyria (AIP),
hereditary coproporphyria (HCP), and variegate porphyria (VP). The
acute neurologic manifestations are similar in each, with the exception
that a photosensitive rash is seen with HCP and VP but not in AIP.
Attacks of porphyria can be precipitated by certain drugs (usually those
metabolized by the P450 system), hormonal changes (e.g., pregnancy,
menstrual cycle), and dietary restrictions.
An acute attack of porphyria may begin with sharp abdominal
pain. Subsequently, patients may develop agitation, hallucinations,
or seizures. Several days later, back and extremity pain followed by
weakness ensues, mimicking GBS (Pattern 1, Table 446-2). Weakness
can involve the arms or the legs and can be asymmetric, proximal, or
distal in distribution, as well as affecting the face and bulbar musculature. Dysautonomia and signs of sympathetic overactivity are common
(e.g., pupillary dilation, tachycardia, and hypertension). Constipation,
urinary retention, and incontinence can also be seen.
The CSF protein is typically normal or mildly elevated. Liver
function tests and hematologic parameters are usually normal. Some
patients are hyponatremic due to inappropriate secretion of antidiuretic hormone (Chap. 378). The urine may appear brownish in color
secondary to the high concentration of porphyrin metabolites. Accumulation of intermediary precursors of heme (i.e., d-aminolevulinic
acid, porphobilinogen, uroporphobilinogen, coproporphyrinogen, and
protoporphyrinogen) is found in urine. Specific enzyme activities can
also be measured in erythrocytes and leukocytes. The primary abnormalities on EDx are marked reductions in compound motor action
potential (CMAP) amplitudes and signs of active axonal degeneration
on needle EMG.
The porphyrias are inherited in an autosomal dominant fashion.
AIP is associated with porphobilinogen deaminase deficiency, HCP
is caused by defects in coproporphyrin oxidase, and VP is associated
with protoporphyrinogen oxidase deficiency. The pathogenesis of the
neuropathy is not completely understood. Treatment with glucose and
hematin may reduce the accumulation of heme precursors. Intravenous glucose is started at a rate of 10–20 g/h. If there is no improvement within 24 h, intravenous hematin 2–5 mg/kg per day for 3–14
days should be administered.
■ FAMILIAL AMYLOID POLYNEUROPATHY
Familial amyloid polyneuropathy (FAP) is phenotypically and genetically heterogeneous and is caused by mutations in the genes for
transthyretin (TTR), apolipoprotein A1, or gelsolin (Chap. 112). The
majority of patients with FAP have mutations in the TTR gene. Amyloid deposition may be evident in abdominal fat pad, rectal, or nerve
biopsies. The clinical features, histopathology, and EDx reveal abnormalities consistent with a generalized or multifocal, predominantly
axonal but occasionally demyelinating, polyneuropathy.
Patients with TTR-related FAP usually develop insidious onset of
numbness and painful paresthesias in the distal lower limbs in the third
to fourth decade of life, although some patients develop the disorder
later in life (Pattern 2, Table 446-2). Carpal tunnel syndrome (CTS) is
common. Autonomic involvement can be severe, leading to postural
hypotension, constipation or persistent diarrhea, erectile dysfunction,
and impaired sweating (Pattern 10, Table 446-2). Amyloid deposition
also occurs in the heart, kidneys, liver, and corneas. Patients usually
die 10–15 years after the onset of symptoms from cardiac failure or
complications from malnutrition. Because the liver produces much of
the body’s TTR, liver transplantation has been used to treat FAP related
to TTR mutations. Serum TTR levels decrease after transplantation,
and improvement in clinical and EDx features has been reported.
Both tafamidis meglumine (20 mg daily) and diflunisal (250 mg twice
daily), which prevent misfolding and deposition of mutated TTR,
appear to slow the rate of deterioration in patients with TTR-related
FAP. Recently, two different modes of gene therapy that inhibit hepatic
production of TTR have been shown to improve neurologic function
and quality of life compared to placebo in clinical trials. Inotersen,
an antisense oligonucleotide, is given subcutaneously once a week.
The main side effects are thrombocytopenia and glomerulonephritis.
Patisiran, a small interfering RNA, is give at a dose of 0.3 mg/kg
(up to 30 mg) every 3 weeks. Infusion reactions are common; therefore,
prophylactic corticosteroids, acetaminophen, and an antihistamine
should be administered.
Patients with apolipoprotein A1–related FAP (Van Allen type) usually
present in the fourth decade with numbness and painful dysesthesias in
the distal limbs. Gradually, the symptoms progress, leading to proximal
and distal weakness and atrophy. Although autonomic neuropathy is
3488 PART 13 Neurologic Disorders
not severe, some patients develop diarrhea, constipation, or gastroparesis. Most patients die from systemic complications of amyloidosis (e.g.,
renal failure) 12–15 years after the onset of the neuropathy.
Gelsolin-related amyloidosis (Finnish type) is characterized by the
combination of lattice corneal dystrophy and multiple cranial neuropathies that usually begin in the third decade of life. Over time, a
mild generalized sensorimotor polyneuropathy develops. Autonomic
dysfunction does not occur.
ACQUIRED NEUROPATHIES
■ PRIMARY OR AL AMYLOIDOSIS (SEE CHAP. 112)
Besides FAP, amyloidosis can also be acquired. In primary or AL
amyloidosis, the abnormal protein deposition is composed of immunoglobulin light chains. AL amyloidosis occurs in the setting of multiple
myeloma (MM), Waldenström’s macroglobulinemia, lymphoma, other
plasmacytomas, or lymphoproliferative disorders, or without any other
identifiable disease.
Approximately 30% of patients with AL primary amyloidosis present with a polyneuropathy, most typically painful dysesthesias and
burning sensations in the feet (Pattern 2, Table 446-2). However, the
trunk can be involved, and some patients manifest with a mononeuropathy multiplex pattern. CTS occurs in 25% of patients and may be
the initial manifestation. The neuropathy is slowly progressive, and
eventually, weakness develops along with large-fiber sensory loss. Most
patients develop autonomic involvement with postural hypertension,
syncope, bowel and bladder incontinence, constipation, impotence,
and impaired sweating (Pattern 10, Table 446-2). Patients generally die
from their systemic illness (renal failure, cardiac disease).
The monoclonal protein may be composed of IgG, IgA, IgM, or
only free light chain. Lambda (λ) is more common than κ light chain
(>2:1) in AL amyloidosis. The CSF protein is often increased (with
normal cell count), and thus, the neuropathy may be mistaken for
CIDP (Chap. 447). Nerve biopsies reveal axonal degeneration and
amyloid deposition in either a globular or diffuse pattern infiltrating
the perineurial, epineurial, and endoneurial connected tissue and in
blood vessel walls.
The median survival of patients with primary amyloidosis is <2 years,
with death usually from progressive congestive heart failure or renal
failure. Chemotherapy with melphalan, prednisone, and colchicine, to
reduce the concentration of monoclonal proteins, and autologous stem
cell transplantation may prolong survival, but whether the neuropathy
improves is controversial.
■ DIABETIC NEUROPATHY
DM is the most common cause of peripheral neuropathy in developed
countries. DM is associated with several types of polyneuropathy:
distal symmetric sensory or sensorimotor polyneuropathy, autonomic
neuropathy, diabetic neuropathic cachexia, polyradiculoneuropathies,
cranial neuropathies, and other mononeuropathies. Risk factors for the
development of neuropathy include long-standing, poorly controlled
DM and the presence of retinopathy and nephropathy.
Diabetic Distal Symmetric Sensory and Sensorimotor
Polyneuropathy (DSPN) DSPN is the most common form of
diabetic neuropathy and manifests as sensory loss beginning in the
toes that gradually progresses over time up the legs and into the fingers
and arms (Pattern 2, Table 446-2). When severe, a patient may develop
sensory loss in the trunk (chest and abdomen), initially in the midline
anteriorly and later extending laterally. Tingling, burning, deep aching
pains may also be apparent. NCS usually show reduced amplitudes
and mild to moderate slowing of conduction velocities. Nerve biopsy
reveals axonal degeneration, endothelial hyperplasia, and, occasionally,
perivascular inflammation. Tight control of glucose can reduce the
risk of developing neuropathy or improve the underlying neuropathy.
A variety of medications have been used with variable success to treat
painful symptoms associated with DSPN, including antiepileptic medications, antidepressants, sodium channel blockers, and other analgesics
(Table 446-6).
Diabetic Autonomic Neuropathy Autonomic neuropathy is
typically seen in combination with DSPN. The autonomic neuropathy
can manifest as abnormal sweating, dysfunctional thermoregulation,
dry eyes and mouth, pupillary abnormalities, cardiac arrhythmias,
postural hypotension, GI abnormalities (e.g., gastroparesis, postprandial bloating, chronic diarrhea, or constipation), and genitourinary
dysfunction (e.g., impotence, retrograde ejaculation, incontinence)
(Pattern 10, Table 446-2). Tests of autonomic function are generally
abnormal, including sympathetic skin responses and quantitative
TABLE 446-6 Treatment of Painful Sensory Neuropathies
THERAPY ROUTE DOSE SIDE EFFECTS
First-Line
Lidoderm 5% patch Apply to painful area Up to 3 patches qd Skin irritation
Tricyclic antidepressants (e.g.,
amitriptyline, nortriptyline)
PO 10–100 mg qhs Cognitive changes, sedation, dry eyes and mouth, urinary retention, constipation
Gabapentin PO 300–1200 mg tid Cognitive changes, sedation, peripheral edema
Pregabalin PO 50–100 mg tid Cognitive changes, sedation, peripheral edema
Duloxetine PO 30–60 mg qd Cognitive changes, sedation, dry eyes, diaphoresis, nausea, diarrhea, constipation
Second-Line
Carbamazepine PO 200–400 mg q 6–8 h Cognitive changes, dizziness, leukopenia, liver dysfunction
Phenytoin PO 200–400 mg qhs Cognitive changes, dizziness, liver dysfunction
Venlafaxine PO 37.5–150 mg/d Asthenia, sweating, nausea, constipation, anorexia, vomiting, somnolence, dry
mouth, dizziness, nervousness, anxiety, tremor, and blurred vision as well as
abnormal ejaculation/orgasm and impotence
Tramadol PO 50 mg qid Cognitive changes, gastrointestinal upset
Third-Line
Mexiletine PO 200–300 mg tid Arrhythmias
Other Agents
EMLA cream Apply cutaneously qid Local erythema
2.5% lidocaine
2.5% prilocaine
Capsaicin 0.025–0.075% cream Apply cutaneously qid Painful burning skin
Source: Modified from AA Amato, J Russell: Neuromuscular Disorders, 2nd ed. New York, McGraw-Hill, 2016, Table 22-3, p. 485.
No comments:
Post a Comment
اكتب تعليق حول الموضوع