You are on page 1of 16

This copy is for personal use only. To order printed copies, contact reprints@rsna.

org
1824
SPINE IMAGING

Diagnostic Approach to Intrinsic


Abnormality of Spinal Cord Signal
Intensity
Michael J. Lee, MD
Ryan Aronberg, MD, MS Intramedullary cord hyperintensity at T2-weighted MRI is a com-
Matthew S. Manganaro, MD mon imaging feature of disease in the spinal cord, but it is non-
Mohannad Ibrahim, MD specific. Radiologists play a valuable role in helping narrow the
Hemant A. Parmar, MD differential diagnosis by integrating patient history and laboratory
test results with key imaging characteristics. The authors present an
Abbreviations: ADEM = acute disseminated algorithmic approach to evaluating intrinsic abnormality of spinal
encephalomyelitis, ALS = amyotrophic lateral cord signal intensity (SI), which incorporates clinical evaluation
sclerosis, CSF = cerebrospinal fluid, dAVF =
dural arteriovenous fistula, FLAIR = fluid-at- results, time of onset (acute vs nonacute), cord expansion, and
tenuated inversion recovery, HIV = human im- pattern of T2 SI abnormality. This diagnostic approach provides a
munodeficiency virus, MS = multiple sclerosis,
NMOSD = neuromyelitis optica spectrum disor-
practical framework to aid both trainees and practicing radiologists
der, SACD = subacute combined degeneration, in workup of myelopathy.
SI = signal intensity, STIR = short inversion time
©
inversion-recovery, TM = transverse myelitis RSNA, 2019 • radiographics.rsna.org

RadioGraphics 2019; 39:1824–1839

https://doi.org/10.1148/rg.2019190021

Content Codes: Introduction


From the Department of Radiology, Division of
Myelopathy is a broad term that references the clinical symptoms re-
Neuroradiology, University of Michigan Health lated to spinal cord dysfunction such as motor and sensory changes
System, 1500 E Medical Center Dr, UH B1- and bowel and bladder dysfunction. MRI plays a key role in evalua-
D502, Ann Arbor, MI 48109. Presented as an
education exhibit at the 2018 RSNA Annual tion of suspected myelopathy because it can help identify a cause and
Meeting. Received February 15, 2019; revision delineate the extent of the abnormality.
requested May 13 and received June 27; ac-
cepted July 2. For this journal-based SA-CME Hyperintense intramedullary signal at T2-weighted imaging is
activity, the author M.J.L. has provided disclo- a common and important indicator of myelopathy at MRI (1). T2
sures (see end of article); all other authors, the
editor, and the reviewers have disclosed no rel-
hyperintensity can reflect many processes at the microscopic level,
evant relationships. Address correspondence including edema, blood–spinal cord barrier breakdown, ischemia,
to M.J.L. (e-mail: mjinlee@med.umich.edu). myelomalacia, or cavitation (2). The differential diagnosis includes a
©
RSNA, 2019 large number of diseases that affect the spinal cord. As such, abnor-
mality of intramedullary signal intensity (SI) is somewhat nonspe-
SA-CME Learning Objectives cific and can present a diagnostic dilemma.
After completing this journal-based SA-CME
Clinical evaluation (including patient history, physical examina-
activity, participants will be able to: tion, and laboratory tests) is the cornerstone of workup of suspected
■■Develop a systematic algorithmic ap- spinal cord disease. As such, the radiologist should be aware of the
proach to evaluating intramedullary SI patient’s clinical evaluation results, which greatly influence the dif-
abnormality at T2-weighted spinal MRI. ferential diagnosis. When appropriate, this information is integrated
■■Describe the clinical and imaging fea- into the diagnostic algorithm.
tures of different causes of intrinsic spi-
nal cord T2 SI abnormality with a focus
Extrinsic compression is a common cause of intramedullary T2
on demyelinating disorders. SI abnormality, and excluding this cause is critical during imaging
■■Recognize pitfalls and mimics in evaluation. In cases of extrinsic compression, the cause of abnormal-
evaluation of intrinsic spinal cord SI ity is known and does not pose a diagnostic dilemma. Therefore, this
abnormalities, including those related to review focuses on intrinsic spinal cord SI abnormality that occurs in
artifacts or extrinsic compression.
the absence of an extrinsic compressive lesion. We present a practi-
See rsna.org/learning-center-rg. cal approach to diagnosis when an intrinsic cord SI abnormality is
found. Rather than presenting an exhaustive list of spinal cord dis-
eases, we focus on the common intrinsic disorders of the spinal cord
with special attention to demyelinating conditions.

Algorithmic Approach
The algorithmic approach shown in Figure 1 first focuses on
excluding artifact or extrinsic compression as a cause of cord SI
abnormality. Next, a combination of clinical evaluation results,
RG  •  Volume 39  Number 6 Lee et al  1825

Extrinsic Compression
Teaching Points Once a true (nonartifactual) spinal cord SI abnor-
■■ The algorithmic approach shown in Figure 1 first focuses on
excluding artifact or extrinsic compression as a cause of cord
mality is identified, it is important to evaluate for
SI abnormality. Next, a combination of clinical evaluation the presence of an extrinsic compressive cause.
results, time of onset (acute vs nonacute), cord expansion, Common extrinsic compressive causes include
and pattern of T2 SI abnormality helps the radiologist arrive discogenic myelopathy, ligamentous thickening
at a more specific diagnosis or at least narrow the differential or redundancy from spondylotic change, verte-
diagnosis.
bral body compression fracture with retropulsion,
■■ Once a true (nonartifactual) spinal cord SI abnormality is
traumatic facet dislocation, and malignancy or
identified, it is important to evaluate for the presence of an
extrinsic compressive cause. Common extrinsic compressive
infection of the surrounding spinal column. Usu-
causes include discogenic myelopathy, ligamentous thicken- ally, cord SI alteration is seen focally at or adjacent
ing or redundancy from spondylotic change, vertebral body to the causative feature. However, long-segment
compression fracture with retropulsion, traumatic facet dislo- cord SI abnormality has also been associated with
cation, and malignancy or infection of the surrounding spinal
spondylotic change and may mimic inflammatory
column.
or neoplastic causes (4,5) (Fig 3). While some of
■■ Demyelinating diseases are a common cause of intrinsic spinal
cord SI change.
these causes may manifest in a nonacute setting,
localized cord SI alteration in a patient with acute
■■ Acute infectious myelopathy can be caused by viral, bacterial,
and fungal pathogens. MRI demonstrates diffuse, typically
myelopathic symptoms should be considered to
nonexpansile T2 hyperintensity, with or without SI alterations be acute edema until proved otherwise and may
at dedicated brain imaging. These findings make it essentially necessitate emergent surgical intervention.
indistinguishable from demyelinating conditions, and differ-
entiation is more likely to be made after CSF sampling and
laboratory tests.
Acute versus Nonacute
Symptom Onset
■■ In the nonacute setting, intrinsic SI alteration with associated
focal expansion of the cord suggests a neoplastic process.
Once artifacts and extrinsic compression are ex-
cluded as possible causes of cord SI abnormality,
the remaining cord SI alterations can be consid-
ered intrinsic to the spinal cord. At this point, it is
time of onset (acute vs nonacute), cord expan- essential to know whether the symptom onset is
sion, and pattern of T2 SI abnormality helps acute or nonacute, as this will strongly influence
the radiologist arrive at a more specific diagno- the differential diagnosis.
sis or at least narrow the differential diagnosis.
Ancillary findings such as additional clinical Acute Onset
workup results, extraspinal manifestations, and If the symptom onset is acute, categories of
features on other MR images are appropriate causes to be considered are demyelination, isch-
for inclusion. This algorithmic approach may be emia, and infection. Among these, demyelination
a useful tool for practicing radiologists as well is the most common.
as trainees.
Demyelinating Disease
Artifacts Demyelinating diseases are a common cause
Quality control is the first step in image inter- of intrinsic spinal cord SI change. Demyelinat-
pretation. Although quality control and artifact ing diseases represent a heterogeneous group of
are not the focus of this article, the radiologist diseases with variable clinical manifestation and
should be mindful of the causes of artifact at imaging features. These include multiple sclero-
spinal imaging. These include Gibbs (aka trunca- sis (MS), acute disseminated encephalomyelitis
tion) artifacts seen at high-contrast interfaces, (ADEM), neuromyelitis optica spectrum disorder
respiratory motion, vascular pulsation, cerebro- (NMOSD), and idiopathic transverse myelitis
spinal fluid (CSF) pulsation, and magnetic field (TM). Notably, MS is at least 10 times as com-
inhomogeneity or susceptibility artifact related to mon as the others in this category (Table).
surgical implants (3). Of particular note, Gibbs
artifact can appear as alternating lines of low and Multiple Sclerosis.—MS is a demyelinating disease
high SI extending along the long axis of the spi- of the central nervous system that is mediated by
nal cord, which can mimic a cord SI abnormal- T cells and macrophages and is characterized by
ity or a syrinx (3) (Fig 2). This pattern is caused focal symptomatic lesions in the brain and spinal
by the high-contrast interface of CSF with the cord (1,6). With an incidence of about 3.6 per
spinal cord and can be minimized by increasing 100 000 person-years, MS is the most common
the number of phase-encoding steps, switching demyelinating disease, with a higher incidence in
the frequency- or phase-encoding directions, or females and in populations farther from the equa-
decreasing the field of view (3). tor (7) (Table). The clinical course and severity of
1826  October Special Issue 2019 radiographics.rsna.org

Figure 1.  Algorithmic approach to evaluating T2 spinal cord hyperintensity at MRI. HIV = human im-
munodeficiency virus, Inflamm/Immune-mediated = inflammatory or immune-mediated, Neuro-degen =
neurodegenerative.

Figure 2.  Gibbs (aka truncation)


artifact in two patients. Sagittal MR
images show multiple alternating
light and dark parallel lines (arrow)
at high-contrast interfaces, mim-
icking intrinsic cord SI abnormality
or a syrinx.

the disease can vary greatly, with several clinical 20% of patients will have only spinal cord mani-
variants identified (8). The McDonald criteria are festations (11). MS in the spinal cord commonly
used to diagnose MS by incorporating clinical and affects the cervical region (1). Lesions are typi-
radiologic evidence of multiple attacks dissemi- cally short (ie, <1.5 vertebral body segments) in
nated in space and time (6,9). The criteria include craniocaudal extent, peripheral, and wedge-shaped
the presence of oligoclonal bands in the CSF, or round and affect less than half of the cross-
which is both sensitive and specific for MS (10). sectional area of the cord (1,12) (Figs 4, 5). There
The spinal cord is affected in more than 90% is involvement of both the gray and white matter
of patients with clinically definite MS, and up to in the brain and spinal cord; however, gray matter
RG  •  Volume 39  Number 6 Lee et al  1827

Figure 3.  Spondylotic myelopathy in a 40-year-old man with leg weakness. (a, b) Sagittal short inversion time inver-
sion-recovery (STIR) MR image (a) and MR image obtained after administration of contrast material (b) demonstrate T2
cord hyperintensity (arrow in a) and irregular patchy enhancement (arrowhead in b) secondary to extrinsic compres-
sion from surrounding disk bulge and degenerative change at the level of the most severe narrowing. (c) Follow-up
MR image 14 months after posterior decompression surgery demonstrates significant improvement of the cord edema
with residual focal myelomalacia (arrow).

Clinical Features of Demyelinating Diseases

Parameter MS ADEM NMOSD TM


Incidence (per 3.6 0.4 0.05–0.40 0.001–0.008
100 000
person-years)
Clinical history Multiple episodes Abrupt single episode Visual symptoms Abrupt
Signs of encephalopathy single
Occurs after viral illness or episode
infection
Laboratory test Oligoclonal bands CSF analysis shows pleocytosis Positive serum or CSF NA*
results and no oligoclonal bands aquaporin-4 antibody
No oligoclonal bands
Other neurologic Dissemination in time Symmetric bilateral brain le- Optic nerves affected May be
sequelae and space sions or large brain lesions precursor
Brain lesions in peri- involving deep gray matter or to MS
ventricular and jux- cortex with relative sparing of
tacortical regions periventricular region
*NA = not applicable.

involvement is more evident in the spinal cord concomitant juxtacortical, periventricular, or


than in the brain at routine imaging (1,12,13). infratentorial brain lesions (8) (Fig 5).
In acute or active disease, the lesions can dem-
onstrate contrast enhancement (from transient Acute Disseminated Encephalomyelitis.—
blood–spinal cord barrier breakdown) or cord ADEM typically manifests as an acute mono-
swelling (1,12). In chronic and long-standing phasic illness after viral infection or vaccina-
or progressive disease, there can be spinal cord tion, predominantly occurring in the pediatric
atrophy, which is thought to represent axonal loss population (1,14). The proposed mechanism
(1,11). Many patients with MS have intracranial is development of an autoimmune antibody
manifestations, so it is essential to evaluate for against myelin basic protein (1). It is much less
1828  October Special Issue 2019 radiographics.rsna.org

Figure 4.  Distinguishing imaging


features of demyelinating diseases.
A short lesion is defined as less
than 1.5 vertebral bodies in length,
compared to a long lesion, which
is greater than 1.5 vertebral bodies
in length.

Figure 5.  Spinal cord and intracranial involvement in a 62-year-old woman with long-standing MS. (a, b) Sagittal STIR (a) and axial
T2-weighted (b) MR images of the cervical and upper thoracic spine show areas of patchy and short-segment (<1.5 vertebral body
length) hyperintensity with a peripheral wedge-shaped appearance (arrows). (c) Axial fluid-attenuated inversion-recovery (FLAIR) MR
image of the brain demonstrates areas of bilateral patchy T2 or FLAIR high SI in a pericallosal and periventricular distribution (arrows).
The combined imaging features are typical of a demyelinating disease such as MS.

common than MS, with a reported incidence At spinal imaging, lesions of ADEM may be
of 0.4 per 100 000 person-years (15). ADEM indistinguishable from those of MS, with some
can be differentiated clinically from MS by its potential differences. ADEM lesions are found
monophasic course, signs of encephalopathy, more commonly in the thoracic cord, are usually
and CSF analysis showing pleocytosis without poorly marginated (owing to adjacent edema),
oligoclonal bands (16) (Table). and are larger in cross-sectional area and longer
RG  •  Volume 39  Number 6 Lee et al  1829

Figure 6.  ADEM in a 10-year-old boy with acute onset of weakness. (a) Sagittal T2-weighted MR image demonstrates long-seg-
ment hyperintensity (arrows) extending from the upper to mid thoracic cord without expansion. (b) Axial FLAIR image of the brain
demonstrates additional T2 or FLAIR hyperintensity in the right thalamus (arrowhead). (c) Follow-up axial MR image 6 months later
demonstrates complete resolution of the previously seen hyperintense lesion in the right thalamus.

in craniocaudal extent (although variable in size) natalizumab) have been reported to be ineffective
(1,17,18) (Figs 4, 6). On images obtained during against or even exacerbate the underlying disease
the acute phase, the cord may show mild expan- in patients with NMOSD (24). Because of the
sion and lesions may demonstrate a variable differing disease course and divergent therapeutic
enhancement pattern (1). approach, it has become critical to differentiate
MRI of the brain as well as the spinal cord is NMOSD from MS when possible.
essential and may further help distinguish ADEM At imaging, NMOSD lesions in the spinal
from MS. Intracranial findings may mimic MS, cord are usually longer in craniocaudal extent
but certain features help confirm the diagnosis than those in MS (>1.5 vertebral body) and in-
of ADEM, such as the presence of larger lesions volve the central gray matter of the cord, some-
in the subcortical white matter, involvement of times seen as longitudinally extensive spinal
the deep gray matter structure (basal ganglia and cord lesions (25) (Figs 4, 7). They frequently
thalami) and brainstem, and relative sparing of extend upward into the medulla (26). These
the periventricular region (14,16) (Fig 6). Nota- “bright spotty lesions”—focal internal areas
bly, given the monophasic nature of many cases, of T2 hyperintensity that are at least as bright
follow-up imaging may show resolution (Fig 6c). as CSF with corresponding low SI at T1-
weighted imaging—have recently been shown
Neuromyelitis Optica Spectrum Disorder.— to be highly specific to NMOSD and are seen
NMOSD is a demyelinating disease that predomi- in about one-half of patients (25,26). Contrast
nantly affects the optic nerves and spinal cord, enhancement and cord expansion can be seen
although brain lesions appear to be more common in an acute setting (1).
than previously recognized (1,12,19). The overall
incidence is about 0.05–0.40 per 100 000 person- Idiopathic Acute Transverse Myelitis.—Idio-
years, predominantly affecting females (1,20). pathic acute TM is not a single disease entity but
Laboratory tests in patients with NMOSD a diagnosis of exclusion when there are clinical
are likely to show the presence of the NMO-IgG signs of acute myelitis without a clear identifi-
antibody, a serum autoantibody that reacts to the able cause (27). Reported incidence rates ranging
water channel protein aquaporin-4. The pres- from 0.001 to 0.008 per 100 000 person-years,
ence of the NMO-IgG antibody is approximately with the variation likely owing to differences
70% sensitive and 90% specific for NMOSD. in the definition and advances in diagnostic
(14,21,22). CSF oligoclonal IgG bands are usually techniques over time (28) (Table). Because this
absent (14,23) (Table). entity is rare and is diagnosed from the clinical
The overall prognosis is worse and the physical standpoint, the radiologist should use this term
manifestations are more severe in patients with sparingly or not at all, as a large number of other
NMOSD than in patients with MS (1,6). The causes must be excluded before considering TM
mainstay therapies for MS (eg, interferon-β and in the differential diagnosis.
1830  October Special Issue 2019 radiographics.rsna.org

Figure 7.  NMOSD in a 36-year-old woman. (a, b) Sagittal T2-weighted (a) and contrast-enhanced T1-weighted (b) MR images
demonstrate cord T2 hyperintensity extending from the lower medulla to the C6 level associated with mild cord expansion (arrow
in a) and heterogeneous enhancement (arrow in b). (c) Axial T2-weighted MR image shows hyperintensity (arrow) affecting more
than two-thirds of the cross-sectional area of the cord. (d) MR image shows mild expansion and patchy enhancement of the right
optic nerve (arrowhead).

The imaging features of TM are variable and


nonspecific, ranging from normal to findings simi-
lar to those of NMOSD (29). When imaging find-
ings are present, they are typically long-segment
cervicothoracic lesions affecting more than 50%
of the spinal cord cross-sectional area, with central
spinal cord predominance with or without en-
hancement and mild cord expansion in the acute
setting (1,27) (Figs 4, 8).

Ischemic Myelopathy
Spinal cord infarction is a rare cause of acute Acute arterial compromise is often associ-
myelopathy, accounting for about 6% of cases of ated with plaque-related thrombosis or emboli.
myelopathy (30). As in infarction involving the Other causes include occlusion related to aortic
brain, the onset of symptoms is abrupt and the or cardiac interventions, trauma, systemic
neurologic deficits depend on the vascular terri- arteriopathy, or rarely fibrocartilaginous embo-
tory and the level of cord affected (30). In addition lization (30,32,33). Classically, anterior spinal
to neurologic symptoms, back pain is also com- artery infarct produces T2 hyperintensity in the
mon and is seen in about 70% of patients (30). anterior horns and surrounding white matter,
Spinal cord ischemia can be arterial or ve- forming the “owl’s eye” sign (Fig 9). Posterior
nous. The arterial supply to the spinal cord arises spinal artery infarct produces T2 hyperinten-
from multiple radiculomedullary arteries, which sity that is limited to the dorsal columns and
ultimately form the anterior and posterior spinal posterior horns (31,34). Restricted diffusion at
arteries. The anterior spinal artery perfuses the diffusion-weighted imaging can improve diag-
anterior two-thirds of the spinal cord, and the pos- nostic certainty when cord infarct is suspected
terior spinal arteries supply the posterior one-third (Fig 9) (35,36).
of the spinal cord. In the subacute setting, there may be enhance-
Compromise of the anterior or posterior circu- ment and hemorrhagic conversion (30). Another
lation causes different neurologic sequelae (30). helpful imaging feature is the presence of con-
Anterior spinal artery syndrome causes bilateral comitant vertebral body infarction due to com-
loss of motor and spinothalamic function with mon vasculature shared by the spinal cord and
sparing of the dorsal columns, while posterior vertebral body (30).
spinal artery syndrome results in loss of proprio- Frank venous spinal cord infarction is uncom-
ception and perception of vibration below the level mon, although edema from venous congestion
of the dorsal cord (30,31). is common, and resultant ischemia can lead to
RG  •  Volume 39  Number 6 Lee et al  1831

Figure 8.  Recurrent idiopathic TM in a 60-year-old man with several weeks of worsening bilateral lower
extremity weakness, pain, and numbness that progressed to an inability to walk. He was diagnosed with
recurrent idiopathic TM after an extensive workup was negative for an alternate cause. (a) Sagittal T2-
weighted MR image shows a longitudinally extensive cord hyperintensity extending from the T9 level to
the tip of the conus (arrow). (b) On an axial T2-weighted MR image, the lesion is seen to affect nearly the
entire cross-sectional volume of the spinal cord without associated expansion (arrow).

Figure 9.  Acute cord infarct in a 60-year-old woman after thoracoabdominal aortic aneurysm repair. Axial T2-weighted MR im-
age (a), diffusion-weighted MR image (b), and apparent diffusion coefficient (ADC) map (c) show postoperative changes in the
paraspinal soft tissues (arrows in a). There is abnormal T2 hyperintensity involving the anterior horns of the central gray matter,
demonstrating the owl’s eye sign (arrowhead in a), with a corresponding area of low SI on the ADC map (arrowhead in b and c),
suggesting impeded diffusion from acute spinal cord infarction.

progressive insidious onset of myelopathic symp- thin-section FIESTA (fast imaging employing
toms (30,37). Spinal dural arteriovenous fistula steady-state acquisition) may serve as useful tools
(dAVF) can cause increased venous pressure in indeterminate cases or for further characteriza-
and has a subtle but characteristic appearance at tion (38).
MRI. dAVF usually manifests with poorly defined
T2 hyperintensity and cord enlargement, which Infectious Myelopathy
represent spinal cord edema. The location of SI Acute infectious myelopathy can be caused by
abnormality depends on the site of the dAVF, viral, bacterial, and fungal pathogens (39). MRI
and it is often seen in the thoracic cord extending demonstrates diffuse, typically nonexpansile T2
to the conus medullaris. The dilated perimed- hyperintensity, with or without SI alterations at
ullary vessels manifest as multiple serpentine dedicated brain imaging. These findings make it
flow voids along the surface of the spinal cord essentially indistinguishable from demyelinating
(1,37) (Fig 10). Advanced imaging techniques conditions, and differentiation is more likely to be
such as contrast-enhanced MR angiography or achieved after CSF sampling and laboratory tests.
1832  October Special Issue 2019 radiographics.rsna.org

Figure 10.  dAVF in a 37-year-old man with a 4-month history of pro-


gressive lower extremity dysesthesias, gait unsteadiness, and weakness.
(a, b) Sagittal T2-weighted MR images demonstrate longitudinally exten-
sive abnormal T2 hyperintensity extending from the lower thoracic cord
to the conus medullaris (arrow) with prominent surrounding flow voids
(arrowheads). (c) Image from digital subtraction angiography (DSA)
helps confirm a type 1 spinal dAVF supplied by the left T9 segmental ar-
tery with drainage into the dilated and tortuous posterior coronal venous
plexus. (d) Intraoperative image obtained during T8-T10 laminectomies
demonstrates findings seen on the MR images and DSA image.

Intramedullary spinal cord abscess is a more


serious although rare diagnosis, which has also
been reported as being caused by several patho-
gens. Abscess is characterized by ring enhance-
ment at MRI, which develops approximately 1
week after an acute infection (40). Diffusion re-
striction can be a useful ancillary imaging feature,
similar to in intracranial abscesses (41).

Nonacute Onset
The diseases associated with nonacute myelopathy
are distinct from those that manifest acutely. They
include neoplastic, metabolic, neurodegenerative,
and inflammatory or immune-mediated disease and lar), hemorrhage, and associated cystic changes.
human immunodeficiency virus (HIV) infection. Clinical manifestation of intramedullary neo-
plasms typically involves insidious and progres-
Nonacute Expansile Neoplastic sive neurologic symptoms, with back or neck pain
Myelopathy depending on the tumor location (43). This pain
If the onset of symptoms is subacute or chronic, is typically exacerbated by a recumbent position
the next task is to examine the contour of the and may be related to secondary irritation or
spinal cord to determine if the cord is focally distention of the dura (43). It is important to be
expanded. In the nonacute setting, intrinsic SI aware that nonneoplastic causes, such as ADEM
alteration with associated focal expansion of the or NMOSD, can demonstrate cord expansion,
cord suggests a neoplastic process. Typically, especially on images obtained during the acute
spinal cord expansion is an imaging feature that phase. However, the acuity of symptoms helps
helps distinguish neoplastic from nonneoplastic determine the cause, which underscores the im-
conditions (42) (Fig 11). portance of the clinical evaluation.
In addition to cord expansion, ancillary char- If the diagnosis is still uncertain after spinal
acteristics often seen in intramedullary neoplasm imaging and clinical workup, additional imag-
include enhancement (especially focal or nodu- ing of the brain may be helpful. The presence of
RG  •  Volume 39  Number 6 Lee et al  1833

Figure 11. Neoplastic versus


nonneoplastic causes of intrinsic
spinal cord SI abnormality. The
presence of cord expansion is used
to differentiate between neoplastic
and nonneoplastic causes.

intracranial lesions may indicate an inflammatory change and hemorrhage (42). About 20%–30%
cause. If uncertainty persists, short-term follow- of cases demonstrate the hemosiderin “cap” sign,
up spinal imaging may be helpful, as persistence characterized by a rim of T2 hypointensity at one
or enlargement of the spinal lesion indicates a or both poles of the tumor (42) (Fig 12).
neoplastic process. Hemangioblastoma is a well-demarcated
Neoplastic lesions of the spinal cord and spinal highly vascular nonglial tumor (42). It can appear
column are commonly categorized as intramedul- similar to cerebellar hemangioblastoma, with an
lary or extramedullary. This discussion focuses on avidly enhancing mural nodule with or without
imaging features of intramedullary lesions, which an associated tumor cyst or syrinx formation
can manifest as focal T2 hyperintensity within (42). Classically, internal flow voids and pres-
the cord. Frequently encountered intramedullary ence of a large draining vein are seen; however,
neoplasms include astrocytoma, ependymoma, despite its high vascularity, associated hemor-
and hemangioblastoma. Although far less com- rhage is rare (42). When there are multiple le-
mon, lymphoma and metastases can manifest as sions or additional lesions in the cerebellum, the
intramedullary lesions and could also be consid- diagnosis of von Hippel–Lindau disease should
ered in patients with a history of malignancy. be considered (42,43).
Astrocytoma, the most common glial tumor
in the pediatric population, is an infiltrative glial Nonacute Nonexpansile Myelopathy
tumor often involving multiple vertebral body
levels of the cervical, thoracic, and sometimes the Metabolic Disease.—Several metabolic derange-
entire spinal cord (42,43). Owing to their infiltra- ments can lead to spinal cord SI alteration,
tive pattern of growth, they are typically poorly including various vitamin and mineral deficiencies,
defined lesions with patchy enhancement and a mitochondrial diseases, leukodystrophies, and ge-
large amount of peritumoral edema (42). Spinal netic syndromes. For example, subacute combined
astrocytoma occurs most frequently in young degeneration (SACD) can be seen in the setting
males (mean age of presentation, 29 years) and is of vitamin B12 deficiency and is usually related
associated with neurofibromatosis type 1 (42). to malabsorption or inadequate intake (44). This
Ependymoma is the most common glial tumor entity tends to affects the dorsal columns and
in adults and is often seen in the cervical spinal lateral corticospinal tracts, hence patients present
cord (42). Unlike astrocytoma, it is a sharply de- with paresthesia of the hands and feet with loss of
fined encapsulated tumor and is associated with proprioception, which may progress to gait ataxia
neurofibromatosis type 2 (42,43). Ependymoma and even ataxic paraplegia in severe cases (44).
is usually centrally located, enhances avidly, Sagittal MRI demonstrates nonexpansile T2
and commonly demonstrates peritumoral cystic hyperintensity predominantly involving long
1834  October Special Issue 2019 radiographics.rsna.org

Figure 12.  Cord ependymoma in a 25-year-old woman with a history of neurofibromatosis type 2 who presented with progressive
back pain and leg numbness. Sagittal STIR (a), T1-weighted (b), and contrast-enhanced T1-weighted (c) MR images demonstrate a
heterogeneous mildly enhancing intramedullary lesion in the upper thoracic cord, causing cord expansion (arrow). The mass shows
hemorrhagic products along the inferior aspect (arrowhead in a), demonstrating the hemosiderin cap sign.

Figure 13.  SACD in a 54-year-


old man with progressive sensory
and gait disturbance with mild
cognitive slowing who was found
to have a low serum vitamin B12
level. (a) On a sagittal STIR image,
hyperintensity involving the dorsal
aspect of the cord extends from
C1 to C6 (arrow). (b) Axial T2-
weighted MR image demonstrates
nonexpansile hyperintensity in the
dorsal columns in the inverted V
pattern (arrow). The patient’s neu-
rologic symptoms markedly im-
proved after supplemental vitamin
B12 injections.

segments in the posterior cervical and thoracic ing both upper and lower motor neurons results in
spinal cord without associated enhancement (1). muscle weakness, cramps, fasciculations, and even-
Axial T2-weighted MR images of SACD dem- tual progression to respiratory failure. The diagnosis
onstrate hyperintensity involving bilateral dorsal of ALS is rarely made by using imaging alone, and
columns, classically in an “inverted V” configura- other causes such as acute flaccid paraparesis can
tion (45) (Fig 13). Such typical imaging findings have a similar imaging appearance (52). Nonethe-
in a patient with normal serum vitamin B12 levels less, imaging of the cord in suspected ALS can help
should raise suspicion for alternate causes of confirm the diagnosis, exclude other causes, and
SACD, such as nitrous oxide toxic effects, zinc monitor progression (50,51).
toxic effects, or copper deficiency (46–48). MRI demonstrates T2 hyperintensity involving
the anterolateral columns with or without associ-
Neurodegenerative Disease.—Motor neuron ated spinal cord atrophy. The SI abnormality may
diseases of the spinal cord represent a rare group of be seen to extend cephalad along the cortico-
fatal progressive neurodegenerative diseases, includ- spinal tracts into the intracranial compartment
ing primary lateral sclerosis, spinocerebellar ataxia, (50) (Fig 14). The degree of spinal cord atrophy,
iron neurodegeneration, Friedreich ataxia, and especially gray matter, correlates with the de-
amyotrophic lateral sclerosis (ALS) (39). ALS is the gree of disability at both baseline and follow-up
most common type of motor neuron disease (49). examinations (51).
ALS has an incidence of about two in 100 000
person-years, with a short median survival time Inflammatory and Immune-mediated Disease.—
(50,51). Atrophy of the anterior horn cells affect- The three common multisystem inflammatory and
RG  •  Volume 39  Number 6 Lee et al  1835

Figure 14.  ALS in a 52-year-old man with progressive spastic quadriplegia. (a) Axial T2-weighted MR image shows hyperintensity
in the lateral aspects of the cervical spinal cord (arrows) without enhancement or cord expansion. (b, c) Additional axial MR images
demonstrate T2 or FLAIR hyperintensity in the corticospinal tracts within the cerebral peduncles and lateral aspects of the midbrain and
pons (arrows).

immune-mediated disorders affecting the spinal sive myelopathy including leg paresthesia, motor
cord are systemic lupus erythematosus, Sjögren weakness, and back pain (56). At MRI, there is
disease, and neurosarcoidosis. Manifestations of typically extensive long-segment T2 hyperinten-
these diseases are variable, and often the diagnosis sity. In the initial phase, there may be a variable
will be made by considering the clinical history or degree of enhancement. In later stages, there
any prior nonneurologic manifestations. In gen- may be chronic atrophy or even cystic necrosis
eral, central nervous system involvement in these (55,56) (Fig 16). The ancillary finding of fatty
entities is uncommon, and spinal cord involvement bone marrow replacement in the corresponding
in particular is rare. If the spinal cord is affected, vertebral bodies supports the diagnosis (56).
patients can pre­sent with typical myelopathic
symptoms such as numbness or sphincter dysfunc- HIV Myelopathy.—Despite widespread use of
tion (53). If there is concurrent involvement of the antiretroviral therapy, the incidence of neurologic
brain, patients may present with cranial neuropa- sequelae in patients with HIV infection remains
thy and aseptic meningitis (14). high at around 70% (57). HIV and associated
At MRI, there is usually long-segment nonex- opportunistic infections can affect both the
pansile T2 hyperintensity, which can be seen in all central and peripheral nervous systems (57,58).
three entities. Variable intramedullary enhance- In primary HIV-associated myelopathy, patients
ment can be seen in any of these conditions; how- typically present with progressive spastic parapa-
ever, neurosarcoidosis may have distinguishing fea- resis, ataxia, and loss of sensation.
tures including dorsal spinal cord predominance, The vacuolization within the white matter of
leptomeningeal enhancement, and the trident the cord seen at histologic analysis can be seen
sign—crescentic posterior subpial enhancement at MRI as an area of symmetric nonenhancing
with subtle additional central canal enhancement high SI in the posterior columns. This appearance
(53,54) (Fig 15). All corners of the available im- mimics that of SACD and is possibly related to
ages should be evaluated for extraspinal manifesta- an altered vitamin B12 metabolic pathway (59,60)
tions of these multisystem disorders, such as cystic (Fig 17). However, findings at MRI are often
changes in the salivary glands associated with nonspecific and can vary significantly in patients
Sjögren disease or hilar lymphadenopathy associ- with a clinical diagnosis of HIV myelopathy, likely
ated with neurosarcoidosis (Fig 15). When there owing to the heterogeneous nature of this disease
is persistent diagnostic uncertainty, CSF sampling entity. Imaging features can range from normal
can help distinguish these causes because each to diffuse T2 hyperintensity in the central spinal
condition manifests with specific disease markers. cord with associated cord atrophy (58) (Fig 17).
In addition to multisystem disorders, post-
treatment change after spinal irradiation can Other Abnormalities.—Rare anatomic abnormali-
produce myelitis within the irradiated field ties such as spinal cord herniation and arachnoid
(55). Radiation myelitis has a widely variable webs can be seen at imaging as intramedullary
latent period and manifests as slowly progres- T2 hyperintensity and may progress to syrinx
1836  October Special Issue 2019 radiographics.rsna.org

Figure 15.  Neurosarcoidosis in a 52-year-


old man with lower extremity weakness
and fecal and urinary retention. (a, b) Sag-
ittal (a) and axial (b) T2-weighted MR im-
ages show extensive central T2 hyperinten-
sity (arrows) without expansion extending
from the cervicomedullary junction to the
conus medullaris. (c, d) Sagittal (c) and
axial (d) contrast-enhanced MR images
show associated dorsal pial enhancement
(arrow) and enlarged mediastinal lymph
nodes (arrowheads in d). This combination
of findings is typical for neurosarcoidosis.

Figure 16.  Radiation myelopathy


in a 63-year-old man with multiple
myeloma who presented with pro-
gressive weakness and urinary re-
tention approximately 6 months
after targeted spinal radiation
therapy. (a, b) Sagittal STIR im-
age (a) and axial T2-weighted MR
image (b) show extensive central
T2 hyperintensity (arrow) with-
out thoracic cord expansion in
the prior radiation field. (c) Axial
contrast-enhanced T1-weighted
MR image demonstrates mild
patchy enhancement within the
left hemicord (arrow). The combi-
nation of clinical history and imag-
ing findings is typical of radiation
myelopathy.
RG  •  Volume 39  Number 6 Lee et al  1837

Figure 17.  HIV myelopathy. (a, b) Im-


ages in a 50-year-old man with progres-
sive spastic quadriplegia show diffuse
cord atrophy through visualized seg-
ments of the cervical and upper thoracic
spinal cord (a) with subtle T2 SI involv-
ing the central portion of the spinal cord
(arrowhead in b). (c) Axial T2-weighted
MR image in a different patient with sus-
pected HIV myelopathy demonstrates
hyperintensity in the dorsal columns (ar-
row), mimicking SACD.

Figure 18.  Spinal cord herniation in a


66-year-old man with a history of chronic
back pain and acute onset of thoracic in-
trascapular pain. (a) Sagittal T2-weighted
MR image demonstrates a syrinx extend-
ing from C7 to the level of the T2-T3
disk space (arrow) with adjacent cord SI
abnormality. (b) Axial T2-weighted MR
image shows that the cord appears to be
apposed to the ventral aspect of the dura
with no visible CSF ventral to the spinal
cord (arrow). (c) Sagittal CT myelogram
shows a ventrally displaced spinal cord
that is closely apposed to the dorsal as-
pect of the vertebral bodies throughout
the thoracic spine (arrowheads), with
focal distortion of the posterior cord con-
tour at the T3-T4 level with a transition in
the cord caliber (arrow). (d) Axial CT my-
elogram at the T3-T4 level demonstrates
the center of the cord possibly extending
through the anterior surface of the du-
ral sac (arrow). Intraoperatively, this was
confirmed to be a ventral thoracic dural
defect causing spinal cord herniation.

formation secondary to a disruption of CSF flow ing of the spinal cord with enlargement of the sub-
dynamics (61). These abnormalities appear as arachnoid space dorsal to the cord (62) (Fig 18).
characteristic cord contour distortion at imaging. Arachnoid webs are intradural extramedul-
Spinal cord herniation occurs when the spinal cord lary arachnoid tissue that crosses over the dorsal
herniates through a postsurgical or idiopathic dural surface of the spinal cord (61). These result in a
defect. Imaging shows characteristic anterior kink- cord contour distortion that appears similar to
1838  October Special Issue 2019 radiographics.rsna.org

Figure 19.  Arachnoid web in a 47-year-


old man with a history of progressive para-
paresis and lower extremity numbness.
(a) Sagittal T2-weighted MR image dem-
onstrates focal intramedullary abnormal
SI with cord distortion at the T3-T4 level
adjacent to slight cord expansion (arrow).
(b) Sagittal CT myelogram demonstrates
relative expansion of the cord at the T4
level (arrow) with focal cord thinning at
the T3-T4 level (arrowhead), correspond-
ing to the cord abnormality seen on the
MR image. (c) Axial CT myelogram shows
marked thinning with anterior displace-
ment of the cord at the T3-T4 level (ar-
row). Intraoperatively, this was confirmed
to be related to arachnoiditis with webs
without evidence of cord herniation.

cord herniation (Fig 19). Occasionally, the distor-


tion causes indentation of the dorsal spinal cord,
known as the scalpel sign (61). Both cord hernia-
tion and arachnoid web are potentially curable
with surgical intervention, but they are frequently
overlooked diagnoses (61,62). The excellent spa-
tial resolution of images acquired using FIESTA
(fast imaging employing steady-state acquisi-
tion) sequences at MRI may improve detection
(63,64). In equivocal cases, CT myelography can
help localize the dural defect and conventional
myelography shows real-time movement of CSF,
so that other causes of intradural filling defect
such as arachnoid cyst can be excluded (62).
elopathy: a review of structural changes and measurement
techniques. Neurosurg Focus 2016;40(6):E5.
Conclusion 3. Taber KH, Herrick RC, Weathers SW, Kumar AJ, Schomer DF,
Hayman LA. Pitfalls and artifacts encountered in clinical MR
Spinal cord SI abnormality at MRI is a com- imaging of the spine. RadioGraphics 1998;18(6):1499–1521.
monly encountered finding in spinal cord disease 4. Bae YJ, Lee JW, Park KS, et al. Compressive myelopathy: mag-
and poses diagnostic challenges owing to the netic resonance imaging findings simulating idiopathic acute
transverse myelopathy. Skeletal Radiol 2013;42(6):793–802.
broad differential diagnosis and variable appear- 5. Kelley BJ, Erickson BJ, Weinshenker BG. Compressive
ances at imaging. Our algorithmic approach myelopathy mimicking transverse myelitis. Neurologist
that combines clinical evaluation, acute versus 2010;16(2):120–122.
6. Kearney H, Miller DH, Ciccarelli O. Spinal cord MRI in
nonacute time of onset, cord expansion, and pat- multiple sclerosis: diagnostic, prognostic and clinical value.
tern of T2 SI abnormality provides a framework Nat Rev Neurol 2015;11(6):327–338.
for radiologists to help narrow their differential 7. Alonso A, Hernán MA. Temporal trends in the incidence
of multiple sclerosis: a systematic review. Neurology
diagnosis in imaging evaluation of myelopathy. 2008;71(2):129–135.
8. Filippi M, Rocca MA. MR imaging of multiple sclerosis.
Acknowledgments.—The authors would like to thank Danielle Radiology 2011;259(3):659–681.
Dobbs and Vanessa Allen for the illustrations. 9. Thompson AJ, Banwell BL, Barkhof F, et al. Diagnosis of
multiple sclerosis: 2017 revisions of the McDonald criteria.
Lancet Neurol 2018;17(2):162–173.
Disclosures of Conflicts of Interest.—M.J.L. Activities related to 10. Bernitsas E, Khan O, Razmjou S, et al. Cerebrospinal fluid
the present article: disclosed no relevant relationships. Activities humoral immunity in the differential diagnosis of multiple
not related to the present article: honorarium from Contemporary sclerosis. PLoS One 2017;12(7):e0181431.
Diagnostic Radiology for writing an article. Other activities: dis- 11. Lycklama G, Thompson A, Filippi M, et al. Spinal-cord MRI
closed no relevant relationships. in multiple sclerosis. Lancet Neurol 2003;2(9):555–562.
12. Bou-Haidar P, Peduto AJ, Karunaratne N. Differential diag-
nosis of T2 hyperintense spinal cord lesions: part B. J Med
References Imaging Radiat Oncol 2009;53(2):152–159.
1. Grayev AM, Kissane J, Kanekar S. Imaging approach to the 13. Geurts JJ, Barkhof F. Grey matter pathology in multiple
cord T2 hyperintensity (myelopathy). Radiol Clin North Am sclerosis. Lancet Neurol 2008;7(9):841–851.
2014;52(2):427–446. 14. Cho TA, Bhattacharyya S. Approach to Myelopathy. Con-
2. Nouri A, Martin AR, Mikulis D, Fehlings MG. Magnetic tinuum (Minneap Minn) 2018;24(2, Spinal Cord Disor-
resonance imaging assessment of degenerative cervical my- ders):386–406.
RG  •  Volume 39  Number 6 Lee et al  1839

15. Leake JA, Albani S, Kao AS, et al. Acute disseminated 40. Murphy KJ, Brunberg JA, Quint DJ, Kazanjian PH. Spinal
encephalomyelitis in childhood: epidemiologic, clinical and cord infection: myelitis and abscess formation. AJNR Am J
laboratory features. Pediatr Infect Dis J 2004;23(8):756–764. Neuroradiol 1998;19(2):341–348.
16. Young NP, Weinshenker BG, Lucchinetti CF. Acute dis- 41. Dörflinger-Hejlek E, Kirsch EC, Reiter H, Opravil M, Kaim
seminated encephalomyelitis: current understanding and AH. Diffusion-weighted MR imaging of intramedullary spinal
controversies. Semin Neurol 2008;28(1):84–94. cord abscess. AJNR Am J Neuroradiol 2010;31(9):1651–1652.
17. Lee YJ. Acute disseminated encephalomyelitis in children: 42. Koeller KK, Rosenblum RS, Morrison AL. Neoplasms of
differential diagnosis from multiple sclerosis on the basis of the spinal cord and filum terminale: radiologic-pathologic
clinical course. Korean J Pediatr 2011;54(6):234–240. correlation. RadioGraphics 2000;20(6):1721–1749.
18. Rossi A. Imaging of acute disseminated encephalomyelitis. 43. Samartzis D, Gillis CC, Shih P, O’Toole JE, Fessler RG.
Neuroimaging Clin N Am 2008;18(1):149–161, ix. Intramedullary Spinal Cord Tumors. I. Epidemiology, Patho-
19. Wang KY, Chetta J, Bains P, et al. Spectrum of MRI brain physiology, and Diagnosis. Global Spine J 2015;5(5):425–435.
lesion patterns in neuromyelitis optica spectrum disorder: a 44. Ravina B, Loevner LA, Bank W. MR findings in subacute com-
pictorial review. Br J Radiol 2018;91(1086):20170690. bined degeneration of the spinal cord: a case of reversible cervical
20. Marrie RA, Gryba C. The incidence and prevalence of myelopathy. AJR Am J Roentgenol 2000;174(3):863–865.
neuromyelitis optica: a systematic review. Int J MS Care 45. Kumar A, Singh AK. Teaching NeuroImage: inverted V sign
2013;15(3):113–118. in subacute combined degeneration of spinal cord. Neurology
21. Wang Y, Wu A, Chen X, et al. Comparison of clinical 2009;72(1):e4.
characteristics between neuromyelitis optica spectrum dis- 46. Baer AH, McKnight CD, Ibrahim M, Parmar HA. Imaging
orders with and without spinal cord atrophy. BMC Neurol of the Spinal Cord: Classic Syndromes and Non-neoplastic
2014;14(1):246. Lesions. Neurographics 2015;5(6):238–253.
22. Lennon VA, Wingerchuk DM, Kryzer TJ, et al. A serum 47. Kumar N. Copper deficiency myelopathy (human swayback).
autoantibody marker of neuromyelitis optica: distinction from Mayo Clin Proc 2006;81(10):1371–1384.
multiple sclerosis. Lancet 2004;364(9451):2106–2112. 48. Winston GP, Jaiser SR. Copper deficiency myelopathy and
23. Jarius S, Paul F, Franciotta D, et al. Cerebrospinal fluid find- subacute combined degeneration of the cord: why is the
ings in aquaporin-4 antibody positive neuromyelitis optica: phenotype so similar? Med Hypotheses 2008;71(2):229–236.
results from 211 lumbar punctures. J Neurol Sci 2011;306(1- 49. Rezania K, Roos RP. Spinal cord: motor neuron diseases.
2):82–90. Neurol Clin 2013;31(1):219–239.
24. Jarius S, Wildemann B, Paul F. Neuromyelitis optica: clini- 50. Agosta F, Chiò A, Cosottini M, et al. The present and the
cal features, immunopathogenesis and treatment. Clin Exp future of neuroimaging in amyotrophic lateral sclerosis. AJNR
Immunol 2014;176(2):149–164. Am J Neuroradiol 2010;31(10):1769–1777.
25. Yonezu T, Ito S, Mori M, et al. “Bright spotty lesions” on spinal 51. Paquin ME, El Mendili MM, Gros C, Dupont SM, Cohen-
magnetic resonance imaging differentiate neuromyelitis optica Adad J, Pradat PF. Spinal Cord Gray Matter Atrophy in
from multiple sclerosis. Mult Scler 2014;20(3):331–337. Amyotrophic Lateral Sclerosis. AJNR Am J Neuroradiol
26. Pekcevik Y, Mitchell CH, Mealy MA, et al. Differentiating 2018;39(1):184–192.
neuromyelitis optica from other causes of longitudinally 52. Maloney JA, Mirsky DM, Messacar K, Dominguez SR,
extensive transverse myelitis on spinal magnetic resonance Schreiner T, Stence NV. MRI findings in children with acute
imaging. Mult Scler 2016;22(3):302–311. flaccid paralysis and cranial nerve dysfunction occurring during
27. Jacob A, Weinshenker BG. An approach to the diagnosis of the 2014 enterovirus D68 outbreak. AJNR Am J Neuroradiol
acute transverse myelitis. Semin Neurol 2008;28(1):105–120. 2015;36(2):245–250.
28. Berman M, Feldman S, Alter M, Zilber N, Kahana E. Acute 53. Soni N, Bathla G, Pillenahalli Maheshwarappa R. Imaging
transverse myelitis: incidence and etiologic considerations. findings in spinal sarcoidosis: a report of 18 cases and review
Neurology 1981;31(8):966–971. of the current literature. Neuroradiol J 2019;32(1):17–28.
29. Scotti G, Gerevini S. Diagnosis and differential diagnosis of 54. Zalewski NL, Krecke KN, Weinshenker BG, et al. Central
acute transverse myelopathy: the role of neuroradiological in- canal enhancement and the trident sign in spinal cord sar-
vestigations and review of the literature. Neurol Sci 2001;22(8 coidosis. Neurology 2016;87(7):743–744.
suppl 2):S69–S73. 55. Bou-Haidar P, Peduto AJ, Karunaratne N. Differential
30. Vargas MI, Gariani J, Sztajzel R, et al. Spinal cord ischemia: diagnosis of T2 hyperintense spinal cord lesions: part A. J
practical imaging tips, pearls, and pitfalls. AJNR Am J Neu- Med Imaging Radiat Oncol 2008;52(6):535–543.
roradiol 2015;36(5):825–830. 56. Khan M, Ambady P, Kimbrough D, et al. Radiation-Induced
31. Novy J, Carruzzo A, Maeder P, Bogousslavsky J. Spinal cord Myelitis: Initial and Follow-Up MRI and Clinical Features
ischemia: clinical and imaging patterns, pathogenesis, and in Patients at a Single Tertiary Care Institution during 20
outcomes in 27 patients. Arch Neurol 2006;63(8):1113–1120. Years. AJNR Am J Neuroradiol 2018;39(8):1576–1581.
32. Bansal S, Brown W, Dayal A, Carpenter JL. Posterior spinal 57. Bilgrami M, O’Keefe P. Neurologic diseases in HIV-infected
cord infarction due to fibrocartilaginous embolization in a patients. Handb Clin Neurol 2014;121:1321–1344.
16-year-old athlete. Pediatrics 2014;134(1):e289–e292. 58. Chong J, Di Rocco A, Tagliati M, Danisi F, Simpson DM,
33. Mascalchi M, Cosottini M, Ferrito G, Salvi F, Nencini P, Atlas SW. MR findings in AIDS-associated myelopathy.
Quilici N. Posterior spinal artery infarct. AJNR Am J Neu- AJNR Am J Neuroradiol 1999;20(8):1412–1416.
roradiol 1998;19(2):361–363. 59. Sartoretti-Schefer S, Blättler T, Wichmann W. Spinal MRI in
34. Masson C, Pruvo JP, Meder JF, et al. Spinal cord infarc- vacuolar myelopathy, and correlation with histopathological
tion: clinical and magnetic resonance imaging findings findings. Neuroradiology 1997;39(12):865–869.
and short term outcome. J Neurol Neurosurg Psychiatry 60. Thurnher MM, Post MJ, Jinkins JR. MRI of infections and
2004;75(10):1431–1435. neoplasms of the spine and spinal cord in 55 patients with
35. Kranz PG, Amrhein TJ. Imaging Approach to Myelopathy: AIDS. Neuroradiology 2000;42(8):551–563.
Acute, Subacute, and Chronic. Radiol Clin North Am 61. Reardon MA, Raghavan P, Carpenter-Bailey K, et al.
2019;57(2):257–279. Dorsal thoracic arachnoid web and the “scalpel sign”: a
36. Goh C, Phal PM, Desmond PM. Neuroimaging in distinct clinical-radiologic entity. AJNR Am J Neuroradiol
acute transverse myelitis. Neuroimaging Clin N Am 2013;34(5):1104–1110.
2011;21(4):951–973, x. 62. Parmar H, Park P, Brahma B, Gandhi D. Imaging of idiopathic
37. Krings T, Lasjaunias PL, Hans FJ, et al. Imaging in spinal spinal cord herniation. RadioGraphics 2008;28(2):511–518.
vascular disease. Neuroimaging Clin N Am 2007;17(1):57–72. 63. Ferré JC, Carsin-Nicol B, Hamlat A, Carsin M, Morandi
38. Krings T, Geibprasert S. Spinal dural arteriovenous fistulas. X. MR imaging features of idiopathic thoracic spinal cord
AJNR Am J Neuroradiol 2009;30(4):639–648. herniations using combined 3D-fiesta and 2D-PC Cine
39. Granados Sánchez A, García Posada L, Ortega Toscano C, techniques. J Neuroradiol 2005;32(2):125–130.
López López A. Diagnostic Approach to Myelopathies. Rev 64. Goetti R, Wille D, Kretzschmar U, Klein A, Scheer I. Idio-
Colomb Radiol 2011;22(3):1–21. http://www.webcir.org/ pathic spinal cord herniation: first reported case in a child.
revistavirtual/articulos/diciembre11/colombia/col_ingles_a.pdf. JAMA Neurol 2013;70(1):125–126.

TM
This journal-based SA-CME activity has been approved for AMA PRA Category 1 Credit . See rsna.org/learning-center-rg.

You might also like