You are on page 1of 21

PRIMER

Traumatic spinal cord injury


Christopher S. Ahuja1, Jefferson R. Wilson1, Satoshi Nori2, Mark R. N. Kotter3,
Claudia Druschel4, Armin Curt5,6 and Michael G. Fehlings1,7
Abstract | Traumatic spinal cord injury (SCI) has devastating consequences for the physical, social and
vocational well-being of patients. The demographic of SCIs is shifting such that an increasing proportion
of older individuals are being affected. Pathophysiologically, the initial mechanical trauma (the primary
injury) permeabilizes neurons and glia and initiates a secondary injury cascade that leads to progressive
cell death and spinal cord damage over the subsequent weeks. Over time, the lesion remodels and is
composed of cystic cavitations and a glial scar, both of which potently inhibit regeneration. Several animal
models and complementary behavioural tests of SCI have been developed to mimic this pathological
process and form the basis for the development of preclinical and translational neuroprotective and
neuroregenerative strategies. Diagnosis requires a thorough patient history, standardized neurological
physical examination and radiographic imaging of the spinal cord. Following diagnosis, several
interventions need to be rapidly applied, including haemodynamic monitoring in the intensive care unit,
early surgical decompression, blood pressure augmentation and, potentially, the administration of
methylprednisolone. Managing the complications of SCI, such as bowel and bladder dysfunction,
the formation of pressure sores and infections, is key to address all facets of the patient’s injury experience.

Spinal cord injury (SCI) is defined as damage to the the most important intervention we can deliver. For SCI
­spinal cord (FIG. 1) that temporarily or permanently that cannot be prevented, the development of effective
causes changes in its function. SCI is divided into trau- ­treatments becomes crucially important 2.
matic and non-traumatic aetiologies1. Traumatic SCI The past three decades have marked an exciting time
occurs when an external physical impact (for e­ xample, for the field, as numerous neuroprotective and neurore-
a motor vehicle injury, fall, sports-related injury or generative therapies have been translated from preclinical
violence) acutely damages the spinal cord, whereas studies into clinical trials. Although undoubtedly impres-
non-traumatic SCI occurs when an acute or chronic dis- sive, further progress will require a concerted effort to
ease process, such as a tumour, infection or degenerative better understand the pathophysiological c­ ascade of SCI,
disc disease, generates the primary injury. the limitations in translating data obtained from animal
In traumatic SCI, the primary insult damages cells models and how to apply combinatorial treatments to
and initiates a complex secondary injury cascade, which this complex, multifaceted disease process.
cyclically produces the death of neurons and glial cells, This Primer provides new researchers with a succinct,
ischaemia and inflammation. This cascade is followed up‑to‑date foundation on SCI. Discussed herein are key
by changes in the organization and structural architec- aspects of epidemiology, pathophysiology and patient
ture of the spinal cord, including the formation of a glial presentation, relevant to both translational researchers
scar and cystic cavities. The glial scar and cystic cavities, and basic scientists. The Primer also provides an over-
in combination with poor endogenous remyelination view of important in‑practice and upcoming thera­peutic
and axonal regrowth, mean that the spinal cord has a strategies, including medical, surgical and cell-based
poor intrinsic recovery potential, such that SCI causes treatments, and concludes with the current outlook for
Correspondence to M.G.F. ­permanent neurological deficits. patients and future directions of the field.
Toronto Western Hospital, SCIs have devastating physical, social and vocational
West Wing 4th floor, consequences for patients and their families, and a loss Epidemiology
399 Bathurst Street, Toronto,
Ontario M5T 2S8, Canada.
of independence and persistently increased lifelong Incidence and prevalence
michael.fehlings@uhn.ca mortality rates are the hallmarks of SCI. Furthermore, Epidemiological data on SCI are often divided into trau-
the direct costs for the care of patients with SCI are matic and non-traumatic aetiologies, suggesting impor-
Article number: 17018
doi:10.1038/nrdp.2017.18 stagger­ing at US$1.1–4.6 million per patient over their tant epidemiological distinctions1. However, data are
Published online 27 Apr 2017 lifetime, which underscores the role of prevention as most often reported by individual national or provincial

NATURE REVIEWS | DISEASE PRIMERS VOLUME 3 | ARTICLE NUMBER 17018 | 1


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
PRIMER

Author addresses Mortality


Although the survival of patients with traumatic SCI
1
Division of Neurosurgery, Department of Surgery, has improved over time, patients continue to have
University of Toronto, Toronto, Ontario, Canada. mortality rates that exceed those of age-matched con-
2
Department of Orthopedic Surgery, Keio University, trols14. Estimates for acute in‑hospital mortality range
Tokyo, Japan.
from 4% to 17%, then after hospital discharge, annual
3
Department of Clinical Neuroscience, University of
Cambridge, Cambridge, UK. mortality rates remain persistently high, with 3.8%
4
Centrum für Muskuloskeletale Chirurgie, of patients dying in the first year after injury, 1.6% in
Universitätsmedizin Berlin, Berlin, Germany. the second year and then 1.2% for every year there­
5
Spinal Cord Injury Center, Balgrist University Hospital, after. The risk of mortality increases with more-severe
Zurich, Switzerland. injuries, higher injury levels (that is, cervical SCIs are
6
Ordinarius for Paraplegiology, University of Zurich, associated with higher mortality than lumbar SCIs),
Zurich, Switzerland. increasing patient age, the presence of multisystem
7
Toronto Western Hospital, West Wing 4th floor, trauma and higher-­energy injury mechanisms. Despite
399 Bathurst Street, Toronto, Ontario M5T 2S8, Canada. modern medical care, patients with traumatic SCI have
a significantly reduced lifespan. For example, the life
databases, which make generalizations between countries expectancy after SCI for an individual 40 years of age is
difficult. In addition, data are often retrospective and lowered to 23 years after cervical level 5 (C5)–C8 injury,
based on treatment codes or surgical procedures, which 20 years after C1–C4 injury and 8.5 years if they are
fail to capture the true incidence and prevalence of SCI. ventilator dependent 2.
The incidence of SCI varies worldwide3 (FIG.  2).
Among developed regions, the incidence of traumatic Mechanisms/pathophysiology
SCI is higher in North America (39 cases per mil- Acute injury phase
lion individuals) than in Australia (16 cases per ­million Traumatic SCI is pathophysiologically divided into
individuals) or western Europe (15 cases per mil- primary and secondary injuries and can be temporally
lion individuals), owing to higher rates of violent crime divided into the acute (<48 hours), subacute (48 hours
and self-harm 4. By comparison, the prevalence of to 14 days), intermediate (14 days to 6 months) and
non-traumatic SCI has been estimated as 1,227 cases chronic (>6 months) phases (FIG. 3). The initial traumatic
per million individuals in Canada and 364 cases per event (that is, the primary injury) produces immediate
million individuals in Australia; reliable data from other mechanical disruption and dislocation of the vertebral
­countries are not available5,6. column, which causes compression or transection of
Traumatic SCI occurs more commonly in males the spinal cord. This focal region of damage injures
(79.8%) than in females (20.2%)7. The age profile of neurons and oligodendrocytes (that is, the myelinating
individuals with a traumatic SCI has a bimodal distrib­ cell type of the central nervous system (CNS)), disrupts
ution; one peak is between 15 and 29 years of age and the the vascu­lature and compromises the blood–spinal cord
second, smaller but growing peak is in those >50 years barrier. Together, these events immediately initiate a sus-
of age8,9. In the United States, the proportion of patients tained secondary injury cascade, which leads to further
with traumatic SCI >60 years of age increased from damage to the spinal cord and neurological dysfunction.
4.6% in 1970 to 13.2% in 2008 (REFS 10,11). This trend This damage can often be in excess of that caused by the
is continuing in parallel with the ageing population of primary injury.
the world7. Secondary cellular changes during the acute phase
Traffic accidents are the primary cause of all trau- of injury, such as cell dysfunction and death, are caused
matic SCIs in North America and accounted for 38% of by cell permeabilization, pro-apoptotic signalling and
injuries between 2010 and 2014, although this number is ischaemic injury due to the destruction of the micro-
gradually declining 7. Falls are typically the second-most vascular supply of the spinal cord within minutes of
common cause of traumatic SCIs and accounted for injury 15,16. In addition, blood vessel injury can cause
31% of injuries between 2010 and 2014, followed by severe haemorrhages, which can expose the cord to
sports-related injuries, which account for 10–17% of an influx of inflammatory cells, cytokines and vaso-
traumatic SCIs9,11. High-energy impacts, such as traffic active peptides. Indeed, increases in the level of pro-­
accidents and sport-related injuries, are more common inflammatory cytokines, such as tumour necrosis factor
in younger individuals, whereas low-energy impacts, (TNF) and IL‑1β, are evident in the spinal cord within
such as falls, disproportionately occur in individuals minutes of injury 17. This parallels the arrival of inflam-
>60 years of age, in whom underlying spinal degener­ matory cells (such as macrophages, neutrophils and
ative changes, such as degenerative cervical myelopathy, lymphocytes) into the spinal cord, which remain in the
are common9,11. Indeed, the incidence of cervical SCI cord well beyond the subacute phase. The subsequent
for the general population (0.13 per thousand-years)12 overwhelming inflammatory response in the acute and
is much lower than for patients with degenerative cervi- subacute phases of injury, combined with the disrupted
cal myelopathy (12.33 per thousand-years)13. Overall, in blood–spinal cord barrier, progressively add to spinal
the general population, traumatic SCI occurs most fre- cord swelling. Swelling can lead to further mechanical
quently at the level of the cervical spine (~60%), followed compression of the cord, which can extend for multiple
by thoracic (32%) and lumbosacral (9%)7. spinal segments and worsen the injury.

2 | ARTICLE NUMBER 17018 | VOLUME 3 www.nature.com/nrdp


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
PRIMER

Subacute injury phase excito­toxicity releases ATP, DNA and potassium, which
In the acute-to-subacute period, ischaemia and excito- can activ­ate microglial cells. Activated microglia, in
toxicity contribute to a loss of intracellular and extra- addition to other inflammatory cells such as activated
cellular ionic homeostasis, with a key mediator of cell macrophages, polymorphonuclear cells and lympho-
death being intracellular calcium dysregulation in both cytes, infiltrate the injury site, where they propagate
neurons and glia. Data from animal models indicate the inflammatory response and contribute to ongoing
that a high intracellular calcium concentration activates apoptosis of neurons and oligodendrocytes. Phagocytic
calpains, which can cause mitochondrial dysfunction inflammatory cells can clear myelin debris at the injury
and cell death18,19. Furthermore, ongoing necrosis of site, but can also induce further damage to the spinal cord
neurons and glia due to ischaemia, inflammation and through the release of cytotoxic by‑products, including

a Ventral corticospinal tract c d


C2
Dorsal column Tectospinal tract
C2
Vestibulospinal tract
C3
C3 C4
Lateral Reticulospinal tract T2
C4
spinothalamic T3
tract Lateral T4
Brainstem C5
corticospinal C1 C6 C8 T5
tract C2 T6
Ventral Rubrospinal C3 T7
spinothalamic Sensory tract C7 T8
C4 Dorsum
tract root Spinal T9
C5 T10 C6
cord
C6 T11 T1
C7 S3 T12 L1
C8 Vertebra S4–S5
Spinal
root T1
T2 Palm
L2 L2
T3
S2
T4
T5 L3 L3
T6
T7
Motor T8
root L4 L4
T9
T10 L5 L5
b Posterior spinal
arteries T11
T12 S1
Spinal nerves L1
e
C5 Elbow flexors
L2 C6 Wrist extensors
Cauda C7 Elbow extensors
Disc L3 equina C8 Finger flexors
L4
T1 Finger abductors
L5
Vertebra L2 Hip flexors
S1
L3 Knee extensors
S2 L4 Ankle dorsiflexors
Spinal cord S3 L5 Long toe extensors
S4
Anterior spinal S5 S1 Ankle plantar flexors
artery
Figure 1 | Anatomy of the spinal column. a | The spinal cord itself is cord by the spinal arteries, which are located anteriorly and posteriorly and
organized into grey matter (which contains neuronal cell bodies) and white Nature Reviews
branch to perfuse the spinal cord parenchyma. | Disease
The spinal cordPrimers
is also
matter (which contains myelinated axons). The white matter can be surrounded by a protective layer of cerebrospinal fluid contained within the
further subdivided into several ascending or descending tracts, which pachymeninges. c–e | Each segmental region of the spinal cord (part c)
are composed of bundles of axons that originate from and project to innervates a specific region of the skin (part d), muscle (part e) or organ
specific regions in the brain and periphery. These tracts transmit specific group. Damage to the spinal cord can result in partial or complete loss of
information, such as sensory information (for example, temperature or itch) function below the level of the injury. Note that part e describes the
or motor information. Spinal nerve roots enter the spinal cord and either ‘key muscles’ as described in the International Standards for Neurological
convey sensory information to the spinal cord (through the sensory or dorsal Classification of Spinal Cord Injury. Parts a–c are adapted with permission
root) or convey motor information to the periphery (through the motor or from REF. 218, Macmillan Publishers Limited. Part d and part e are adapted
ventral root). b | The vertebral column encircles the spinal cord in protective with permission from the American Spinal Injury Association: International
bone and ligament, which, in humans, is segmented into 7 cervical, Standards for Neurological Classification of Spinal Cord Injury, revised
12 thoracic, 5 lumbar and 5 sacral vertebrae. Blood is supplied to the spinal 2011; Atlanta, GA, Revised 2011, Updated 2015.

NATURE REVIEWS | DISEASE PRIMERS VOLUME 3 | ARTICLE NUMBER 17018 | 3


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
PRIMER

Incidence Incidence
(country) (state or province)
0–10 10–20
10–20 20–30
20–30 30–40
30–40 40–50
40–50 50–60
60–70
70–80
Lack accurate
epidemiological data
States or provinces

Figure 2 | Annual incidence of spinal cord injury across reported countries, states orNature
provinces and regions.
Reviews | Disease Primers
The annual incidence of spinal cord injuries varies depending on geographical region. Adapted with permission from
REF. 3, Dove Press.

free radicals (for example, O2−, hydrogen perox­ide and Intermediate–chronic injury phase
peroxynitrite). These reactive oxygen species cause As the acute inflammatory response subsides, the s­ pinal
lipid peroxidation, DNA oxidative damage and protein cord lesion evolves through dynamic intermediate
oxidation, which cause additional necrotic and delayed through to chronic phases that are marked by attempts
apoptotic cell death, further contributing to the harsh at remyelination, vascular reorganization, alterations in
post-injury microenvironment 20,21. the composition of the extracellular matrix (ECM) and
High levels of the neurotransmitter glutamate are remodelling of neural circuits19.
released from dying neurons and astrocytes and are
poorly reabsorbed by surviving astrocytes22,23. This causes Cystic cavitations. In humans, the overwhelming cell
NMDA (N-methyl-d‑aspartate), AMPA (α‑amino‑­ death and degeneration in the acute phase of injury pro-
3‑hydroxy‑5‑methyl‑4‑isoxazole propionic acid) and motes the ex vacuo (that is, loss of tissue volume) forma-
kainate receptor overactivation, which, when combined tion of cystic cavities, which contain extracellular fluid,
with the loss of ATP-dependent ion pump functions and thin bands of connective tissue and macrophages26. The
subsequent resultant sodium dysregulation, can lead to cystic cavities coalesce to become a formidable barrier
excitotoxic cell death24,25. Neuronal death due to excito­ to directed axonal regrowth and are a poor substrate for
toxicity, as well as the other insults discussed above, cell migration27,28.
­cyclically propagates the secondary injury cascade19.
The impaired autoregulatory capacity of the injured Glial scar. Studies using animal models have shown a
cord vasculature, in addition to the systemic effects perilesional zone around the cystic cavities, in which
of SCI (such as hypotension and respiratory failure; reactive astrocytes proliferate and tightly interweave
see Clinical manifestations section), can contribute to their processes, creating an inhibitory mesh-like array.
­ongoing ischaemia that persists for days to weeks after In the acute phase, signalling from activated microglia,
injury. Prolonged ischaemia contributes to further astrocytes and macrophages causes the secretion of
neuro­nal and glial (predominantly oligodendrocyte) ECM proteins that are inhibitory to axonal growth, such
cell death and the propagation of the injury. The multi- as chondroitin sulfate proteoglycans (CSPGs), tenascin
ple causes of cell death that occur during the acute and and NG2 proteoglycan (also known as chondroitin sul-
subacute phases of SCI can produce greater damage than fate proteoglycan 4), which condense with astrocytes
the original primary injury and form the basis for the to form the glial scar. The glial scar potently restricts
neuroprotective interventions (see below). axon regeneration (that is, the repair or regrowth of

4 | ARTICLE NUMBER 17018 | VOLUME 3 www.nature.com/nrdp


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
PRIMER

existing neural pathways, or the development of de novo macro­phages, lymphocytes and astrocytes, and is inhib-
pathways) and anatomical plasticity by inhibiting ited by the presence of EphrinB3 in myelin debris42,43,
neurite outgrowth29,30. as well as molecules within the glial scar 36,44,45. This
Oligodendrocyte progenitor cells that express NG2 could lead to poor remyelination post-injury, which in
proteoglycan migrate to the lesion site and associate turn impairs functional recovery 46.
with dystrophic axons (that is, swollen, injured axons
that can be found in the damaged CNS). Pericytes also Endogenous attempts at repair
proliferate after SCI, giving rise to stromal cells that Contrary to historical dogma, endogenous mech-
might deposit numerous ECM proteins31. Furthermore, anisms exist for at least partial regeneration of the
fibroblasts can infiltrate the perilesional region, particu- injured spinal cord. CNS neurons exhibit both anatom-
larly after breaks in the glial layer that separates neural ical and synaptic plasticity, which might contribute to
tissue from the meninges26, to replace the ECM with ongoing functional recovery for years after injury 47,48.
fibrous connective tissue and dense collagen deposits. Furthermore, neural precursor cell pools, which are
Together, these ECM and cellular changes represent a mostly found in the ependymal layers of the central
marked physical and biochemical barrier to regener­ canal, as well as widely distributed oligodendrocyte
ation. However, not all aspects of the scar pose an inhib- precursor cells, can generate neurons, oligodendro-
itory barrier 32; complete attenuation of astrocytes in cytes and astrocytes (including reactive astrocytes)49,50.
the glial scar results in impaired regeneration, as astro- These cells might have both helpful and detrimental
gliosis in the acute–subacute phases is respons­ible for roles throughout the post-injury regenerative process.
isolating the injury site, to reduce the spread of cyto- Exploiting these endogenous mechanisms by increas-
toxic mol­ecules and inflammatory cells into adjacent, ing the recruitment of pro-­regenerative cells51, prod­
­uninjured parenchyma33,34. Furthermore, peri­lesional ucing a microenvironment that is more conducive to
astrocytes might provide local trophic support and cell migration and neurite outgrowth52, and/or shifting
promote neovascularization35. This dual role of the glial the balance towards pro-­regenerative cell phenotypes53
scar ­continues to be investigated. are some of the ­exciting areas of ongoing research.
These and other mechanisms can be supplemented by
Adult CNS myelin. Even if regenerative efforts are the neuro­protective and neuroregenerative strategies
able to overcome spinal cord lesions, properties of the discussed later, but barriers to regeneration still exist,
adult mammalian CNS can still limit neurite regrowth. meaning additional therapies to remove or overcome
For example, molecules present in myelin are potent these ­barriers are necessary.
inhib­itors of axon regeneration, and several molecules
released by degenerating oligodendrocytes can con- Animal models
tribute to the failure of regeneration. These mol­ecules Animal models have contributed to our understanding
include neurite outgrowth inhibitor A (Nogo‑A), of the pathophysiology of SCI and have been useful
oligodendrocyte-­myelin glycoprotein (OMgp) and for the preclinical testing of new therapies. The ideal
myelin-­associated glycoprotein (MAG), which can all animal model should anatomically and pathophysio-
bind to the Nogo receptor and p75 neurotrophin recep- logically resemble human SCI, require minimal train-
tor (p75NTR; also known as TNF receptor super­family ing, be inexpensive and produce consistent results. Rat
member 16) to activate RHOA and Rho-associate pro- models are the most commonly used for SCI research
tein kinase (ROCK), which causes growth cone collapse, and are well established and inexpensive, and the
neurite retraction and increases the risk of apoptosis36. injury response is similar to that observed in humans
CSPGs in the glial scar can also activate the Nogo recep- (including the production of cystic cavities, glial scar
tor, in addition to the membrane-bound protein tyrosine formation and changes in the ECM) (BOX 1). However,
phosphatase-θ (PTPθ), to trigger growth cone collapse differences in size, molecular signalling, anatomy and
via the RHO–ROCK pathway 37. Interestingly, individ- the recovery potential following SCI have made direct
ual knockout of Nogo, MAG and OMgp showed limited translation challenging 54. Numerous therapies in SCI
effects on axon regeneration in vivo, potentially owing to and other CNS fields (such as stroke) have, unfortu-
the synergistic inhibitory activity of myelin-­associated nately, been ineffective when translated to humans
proteins and CSPGs on axonal regeneration38,39. This from small animals, owing to their inherent biological
continues to be an area of active investigation. differences. Large animal models, such as non-human
primates, overcome some of these barriers, but substan-
Attempts at remyelination. Although severe SCI can tial differences in cost and unique housing requirements
destroy substantial portions of the spinal cord white make their use less common and even they are unable
matter, a surviving subpial rim of demyelinated axons to exactly mimic human SCI55. However, larger animal
can persist in rodent models of injury 40. These neurons models can form an important intermediary model
are susceptible to subsequent injury and progressive to confirm results from rodents by providing relevant
Wallerian degeneration (that is, an ordered process of safety, biodistribution and technical feasibility data56,57.
axonal death)40,41. In theory, oligodendrocyte precursor Furthermore, testing novel therapies in multi­p le
cells can differentiate into mature oligodendrocytes ­species is an important approach to bolster pre­clinical
and remyelinate these axons. However, r­ emyelination evidence before commencing clinical trials, as is now
requires a coordinated inflammatory response by ­recommended for stroke therapies58.

NATURE REVIEWS | DISEASE PRIMERS VOLUME 3 | ARTICLE NUMBER 17018 | 5


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
PRIMER

a Blood–spinal Mechanical Compromised Diagnosis, screening and prevention


cord barrier injury vascular supply Clinical manifestations
Haemorrhage Release
disruption
Pro-inflammatory of ATP and Fractures of the spinal column are often described
Axon disruption cytokine release potassium by their vertebral level, but the neurological injury is
described by the spinal cord level at which the nerve
roots emerge. The discrepancy between the two becomes
increasingly apparent in the mid-to-low regions of the
thoracic spinal cord, where a fracture at thoracic level 8
(T8) might cause a neurological SCI at T12 and a ­fracture
at T12 might cause a SCI at sacral level 1 (S1).
The clinical manifestations of SCI depend on the
level of neurological injury and the amount of preserved
­spinal cord tissue. SCI can result in the partial or com-
plete loss of sensorimotor function below the level of the
injury. Depending on the level of the injury, this can lead
to compromised respiratory function (including hyper-
Necrotic Pro-apoptotic capnia, hypoxaemia and poor secretion clearance59,60);
cell death signalling for example, injuries above C5 disrupt innervation to the
Inflammatory
cell infiltration Oedema Demyelinated axon diaphragm, injuries above T11 disrupt innervation to the
intercostal chest muscles and injuries above lumbar level 1
(L1) can disrupt innervation to the abdominal muscles.
In addition to disruption of sensorimotor func-
b Vessel Cystic Inflammatory Release of
tion, SCI can affect the sympathetic nervous system, as
thrombosis cavitation cell infiltration oxygen free radicals
preganglionic sympathetic neurons originate in the spinal
Axonal Fibroblast Haematoma
dieback infiltration cord, between T1 and L2. SCI can reduce sympathetic
outflow from the spinal cord, which results in a loss
of basal vascular tone below the level of injury (FIG. 4).
In addition, high thoracic or cervical injuries can lead to
severe hypotension and bradycardia (that is, neurogenic
shock, see below)61,62. The loss of innervation to second-
ary lymphatic organs (such as the spleen) can induce
secondary immunodeficiency (also known as immune
paralysis), which can increase the susceptibility to infec-
tions (for example, urinary tract infections and pneumo-
nia)63. These systemic manifestations of CNS injury are
the leading causes of early mortality in patients with SCI.

Inhibitory Figure 3 | Pathophysiology of traumatic spinal cord


proteoglycan injury. a | The initial mechanical trauma to the spinal cord
deposition Vasospasm initiates a secondary injury cascade that is characterized in
Microglial Ongoing Further
infiltration oedema Astrogliosis demyelination the acute phase (that is, 0–48 hours after injury) by oedema,
haemorrhage, ischaemia, inflammatory cell infiltration, the
release of cytotoxic products and cell death. This secondary
injury leads to necrosis and/or apoptosis of neurons and
c Wallerian Inhibitory Perilesional Coalescence Restricted regenerative glial cells, such as oligodendrocytes, which can lead to
degeneration proteoglycan scar astrogliosis of cystic cell migration demyelination and the loss of neural circuits. b | In the
cavitation subacute phase (2–4 days after injury), further ischaemia
occurs owing to ongoing oedema, vessel thrombosis and
vasospasm. Persistent inflammatory cell infiltration causes
further cell death, and cystic microcavities form, as cells
and the extracellular architecture of the cord are damaged.
In addition, astrocytes proliferate and deposit extracellular
matrix molecules into the perilesional area. c | In the
intermediate and chronic phases (2 weeks to 6 months),
axons continue to degenerate and the astroglial scar
matures to become a potent inhibitor of regeneration.
Cystic cavities coalesce to further restrict axonal regrowth
and cell migration. Republished with permission of
AlphaMed Press, from Concise review: bridging the gap:
novel neuroregenerative and neuroprotective strategies
Limited Restricted in spinal cord injury, Ahuja, C. S. & Fehlings, M., Stem Cells
Limited oligodendrocyte Schwann cell axonal Plasticity of injured Transl Med. 5, 7, 2016, permission conveyed through
remyelination remyelination regrowth and uninjured neurons Copyright Clearance Center, Inc.
Nature Reviews | Disease Primers

6 | ARTICLE NUMBER 17018 | VOLUME 3 www.nature.com/nrdp


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
PRIMER

Spinal shock. Spinal shock is defined as a temporary and is also typified by hypotension, bradycardia, wide
clinical state of flaccid paralysis post-SCI, including the pulse pressure and warm pink extremities. Neurogenic
loss of motor, sensory, autonomic and reflex function at shock is most clinically relevant with a neurological
or below the level of injury. Spinal shock is commonly level of injury above T6, as these injuries prevent central
confused with neurogenic shock (which is a hypo­tensive impulses reaching the mid-thoracic spinal cord, which is
state caused by loss of sympathetic outflow). Spinal where the sympathetic splanchnic nerves (which have
shock can affect the accuracy of the initial neurologi- an important role in maintaining vascular tone) arise.
cal examination, which is used to define the severity of Importantly, in injuries above T6, the sympathetic
SCI. However, understanding when a patient no longer outflow to the cardiac pacemaker can also be affected.
has spinal shock is problematic and has been the sub- Neurogenic shock is estimated to occur in up to 20%
ject of controversy 64. However, the theory to which of patients with cervical level injuries, and bradycardia
most individ­uals subscribe describes spinal shock as a occurs in nearly all patients with severe cervical injuries
four-phase progression, from an initial stage of areflexia during the acute phase66,67.
or hypo­reflexia to the later stage of the return of deep
­tendon reflexes and hyper-reflexia. Diagnosis
After any traumatic injury, first responders rapidly assess
Neurogenic shock. Hypotension post-SCI has several patients in the field and attempt resuscitation en route to
causes, including hypovolaemia secondary to blood loss, the hospital. During this period, the advanced trauma
the distributive pooling of venous blood within para- life support protocols dictate initial care, which includes
lysed atonic musculature and bradycardia. Hypotension airway, breathing and circulation support, along with the
can also be caused by vasodilation secondary to loss of immobilization of the potentially injured and unstable
sympathetic tone65,66, which produces neurogenic shock spinal column using a rigid cervical collar and back-
board. Although individual hospital approaches vary,
most patients with trauma will undergo a gross neuro-
Box 1 | Animal models of spinal cord injury logical examination (which includes a voluntary motor
The type and location of the spinal cord injury (SCI) are key factors in the development and sensory examination of each limb and a rectal
of a clinically relevant and translatable animal model. The anatomical and examination) and spinal imaging (using, for example,
pathophysiological differences between the cervical and thoracic spinal regions are X‑ray or CT imaging) if a SCI is suspected. Concerns on
substantial and should be considered. Furthermore, the choice of species and type clinical examination or early radiographic imaging are
of model might be useful to answer different questions57,211–213. followed by advanced ­imaging and detailed neurological
Models of traumatic SCI ­examinations (see below).
• Contusion models: inflict transient, acute injuries through weight-drop,
or electromagnetic or pneumatic impactors Imaging. Plain X‑ray, CT and MRI are the most com-
• Compression models: inflict prolonged, acute injuries through calibrated
monly used radiological tools when investigating dam-
clip-compression and forceps, among others age to the spine and SCI. Anterior–posterior (AP) and
• Transection models involving unilateral (partial) or bilateral (complete) lesions
lateral cervical spine X‑ray, AP chest and AP pelvis
X‑rays are performed in the trauma room. Although
The ideal behavioural outcome is rapidly assessable, inexpensive, requires minimal not particularly sensitive for the identification of subtle
training, has good intra-rater and inter-rater reliability and causes minimal distress
fractures involving the cervical spine, X‑rays are useful
in the animal. Several established behavioural outcomes exist for mice and rats.
to detect gross fracture dislocation injuries that are often
Locomotor function associated with SCI. It is essential to ensure the adequacy
• Open-field locomotor assessment: used to assess sequential locomotor recovery of of any X‑rays with visualization of the rostral half of the
the hindlimb when used in combination with a locomotor recovery scale, such as the T1 vertebrae.
Basso, Beattie and Bresnahan (BBB) scale CT has largely supplanted X‑ray for the diagnosis of
• Digital systems: used to quantify cadence, walk time, stride length and stride width bone injuries in patients with trauma68,69. With respect
similar to the clinical GaitRite analysis system (CIR Systems, Inc.) to the spine, some authors (namely, M.G.F.) currently
• Ladder, rope or wire grid test: used to assess coordinated locomotion perform, and recommend, a high-resolution fine-cut CT
• Staircase test: used to measure forelimb reaching and grasping of the cervical to lumbar spine. CT angiography can also
Limb strength be performed to evaluate the bilateral vertebral ­arteries
• Inclined plane test: used to indirectly assess trunk stability, proprioception or in certain cervical injuries. AOSpine has also devel-
sensation and unilateral limb strength oped subaxial cervical70 (C3–C7) and thoracolumbar 71
• Forelimb grip strength: used to provide quantitative readouts of the peak force (T1–L5) classification systems to standardize nomen­
of the forelimb clature of bony and ligamentous spinal injuries. These
systems convey key information on the fracture pattern
Sensory function
(for example, compression injury and trans­lational
• Tail-flick test: used to assess nociception in response to temperature injury), including adjacent structure involvement
• Von Frey filaments (thin nylon strands of varying diameter): used to assess sensory (for example, facets, ligaments and vertebral artery)
preservation and allodynia (pain sensation for normally non-painful stimuli) with modifiers for neurological status (for example,
in response to mechanical stimuli
­incomplete SCI and complete SCI).
• The Hargreaves assay: used to assess sensory preservation and allodynia in response Although extremely sensitive for diagnosing a frac-
to temperature
ture or dislocation of the spine, CT is less effective at

NATURE REVIEWS | DISEASE PRIMERS VOLUME 3 | ARTICLE NUMBER 17018 | 7


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
PRIMER

evaluating the integrity of soft tissue structures, such as The timing of MRI can be crucial with respect to the
intervertebral discs, ligaments, the spinal cord and nerve treatment of patients with SCI and cervical facet dis­
roots, but MRI is well suited for assessing these struc- location. MRI before closed reduction (that is, correct-
tures72. Specifically, when evaluating for ligament or ing the dislocation with the use of traction) enables the
verte­bral disc injury, the T2‑weighted short-tau inver- detection of disc herniation, which, if present, can lead
sion recovery (STIR) sequence enables the identification to a deterioration in neurological status, although this
of injury-related oedema and tissue disruption. MRI can has been disputed74,75. However, MRI can, depending
identify spinal cord transection and can ­evaluate for the on the institution, substantially delay time to decom-
presence of oedema and/or haemorrhage73. pression of the spinal cord and involves the additional
transfer of a patient with a highly unstable spine. On the
basis of the existing evidence, the most recent iteration
of the American Association of Neurological Surgeons
and Congress of Neurological Surgeons (AANS/CNS)
Glossopharyngeal
guidelines for the management of cervical SCI recom-
nerve mends MRI before performing an open reduction (that
Cartoid ↓ Heart rate is, realignment of the broken bones following surgery
Medulla

baroreceptors and cardiac to expose the bones) or a closed reduction in an uncon-


Aortic arch output
scious or uncooperative patient; if a disc herniation
baroreceptor
is identified, the guidelines recommend an anterior
Preserved approach to remove the disc before reduction76.
Cervical

baroreceptor The role of MRI is rapidly evolving, and advanced


afferent input microstructural techniques that can quantify physio­
Parasympathetic
outflow to the logical changes at a cellular level and assess axon integrity
Injury Vagus heart is unaffected (for example, diffusion tensor imaging), myelination (for
nerve by the injury
example, myelin water transfer) and the presence of key
metabolites related to ischaemia, cell loss or gliosis (for
Decreased
example, magnetic resonance spectroscopy)30,77 are likely to
Sympathetic outflow

sympathetic
outflow to see increased integration in the care of patients with SCI.
Thoracic

the heart
Electrophysiology. Numerous electrophysiological stud-
ies have been evaluated for predicting outcome and for
tracking and monitoring recovery over time after trau-
Sympathetic
efferents matic SCI. Electrophysiology is an attractive tool as it
does not require the patient to be conscious or commu-
Sacral Lumbar

Decreased sympathetic
Spinal outflow to the vascular nicative. Several parameters have been studied (BOX 2),
cord system leads to a loss which can be used to derive measures of physiological
of vasular tone, increased
blood pooling and and anatomical function, such as conduction time to
decreased venous motor neurons, cortical and spinal inhibition, spinal
return to the heart cord excitability (such as the H‑reflex) and sensory
Parasympathetic impairment, among others. Although interesting as a
outflow from
Decreased muscle tone research tool, electrophysiological measurements have
S2–S4
reduces venous return
not consistently demonstrated added value in predicting
outcome in awake and alert patients with SCI78. However,
electrophysiological measurements might provide insight
into the mechanisms underlying the functional recov-
ery of the patient (for example, regeneration, plasticity
Figure 4 | Cervical and high thoracic spinal cord injuries disrupt the outflow of the or adaptation), which could be of benefit as the field
sympathetic nervous system. Injuries in the cervical and Nature
highReviews Disease
thoracic|cord can Primers
disrupt ­develops, such as for patient selection for clinical trials79.
the sympathetic outflow (blue line) to the heart and the peripheral vascular system,
while preserving baroreceptor inputs (red line) and parasympathetic output (green line). Classification of SCI. Perhaps the most significant
As a result, parasympathetic innervation to the heart dominates in patients with cervical advancement related to our ability to diagnose and clas-
and upper thoracic injuries, which causes bradycardia and decreased cardiac output. sify SCI over the past few decades has been the develop­
This is further compounded by the loss of peripheral muscular and vascular tone, ment of the International Standards for Neurological
which promotes a redistribution of blood to the periphery with reduced venous return. Classification of Spinal Cord Injury (ISNCSCI)80,81.
Consequently, patients often experience hypotensive symptoms, particularly with The ISNCSCI has been uniformly adopted by SCI clin-
exertion or upright positioning. The parasympathetic–sympathetic imbalance can also
ical communities and serves as the main measure of
allow unchecked reflex spinal sympathetic stimulation as a consequence of noxious
triggers (such as bladder distension or pressure sores), which leads to sudden peripheral ­neurological outcome in clinical trials.
vasoconstriction and acute hypertension. As a response, parasympathetic outflow The ISNCSCI comprises three central neurological
above the injury level increases, leading to vasodilation, headaches, sweating and summary scores: American Spinal Injury Association
sinus congestion. This dangerous acute syndrome is known as autonomic dysreflexia. (ASIA) motor score (which grades muscle power from
S2–S4, sacral levels 2–4. each myotome (that is, a group of muscles innervated by

8 | ARTICLE NUMBER 17018 | VOLUME 3 www.nature.com/nrdp


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
PRIMER

Box 2 | Electrophysiological recordings Brown-Séquard syndrome is most commonly


observed in individuals with penetrating traumatic SCI,
Electrophysiological recordings have examined several parameters, including secondary to gunshot and knife wounds. Deficits in
motor-evoked potentials (MEPs), somatosensory-evoked potentials (SSEPs), patients with Brown-Séquard syndrome include loss of
dermatomal SSEPs, electromyography (EMG), nerve conduction studies (NCSs) and motor function, light touch, proprioception and vibra-
sympathetic skin response (SSR). MEPs assess the integrity of descending motor tracts,
tion sensation ipsilateral to the injury, and loss of pain
through a central impulse (usually through transcranial magnetic motor cortex
stimulation), which is then detected by electrodes in peripheral muscle. The amplitude and temperature sensation contralateral to the injury 87.
of the MEP signal, but not the signal latency, correlates with improved motor function Anterior and posterior cord syndromes are rarely
post-injury79. EMGs can examine subtle changes in voluntary muscle contraction to observed in isolation in the context of traumatic SCI,
track recovery214. NCSs provide detailed conduction velocities of nerves to better but are more frequent in patients with non-traumatic
distinguish between ventral horn or anterior root injuries and pyramidal tract injuries215. SCI of vascular aetiology 87.
SSEPs assess the integrity of ascending sensory tracts, usually in the dorsal columns,
through the temporal summation of electroencephalogram (EEG) signal, after the Prognosis
stimulation of a peripheral sensory nerve. SSEPs performed in the acute-phase Neurological recovery. Neurological recovery in patients
post-injury can predict long-term neurological and functional outcomes, such as with SCI is typically observed within the first 6 months
future ambulation216. However, SSEPs were no better than acute clinical examination
after injury, but continued improvements can be seen
in predicting ambulation216. In another study, in the subacute phase of injury,
the combination of lower extremity motor scores and tibial SSEPs provided the most up to 5 years later 88,89. The prognosis for neurologi-
accurate prediction of ambulation compared with the use of either variable alone. cal recovery is variable and depends primarily on the
Finally, the integrity of the sympathetic system can be tested by measurement of initial severity of neurological injury; a more severe
the electrical potential generated between skin sweat glands (SSRs)217. degree of initial injury portends a worsened prognosis
at 1 year 90,91. The neurological level of injury can also
determine neurological recovery; in general, thoracic
one spinal nerve root), the ASIA sensory score (which injuries (particularly complete injuries) are associated
assesses light touch and pinprick sensation in 28 derma­ with reduced potential for motor recovery compared
tomes (that is, an area of skin innervated by one with injuries in the cervical or lumbar spinal cord. This
­spinal nerve root) from C2 to S4 or S5) and the ASIA is thought to exist because neurological recovery is more
Impairment Scale grade (which is used to determine the difficult to ­clinically detect in the thoracic region92,93.
grade of SCI and encompasses the extent of remaining Functional outcomes, in particular, the ability to
sensorimotor function; BOX 3)80. We recommend that walk, are of interest to patients. In general, patients
ISNCSCI examination is performed at the time of acute with ASIA Impairment Scale grade A injuries are gen-
hospital admission as soon as reasonably possible to serve erally predicted to have a <5% chance of walking 1 year
as a baseline for comparison throughout follow‑up. post-injury, regardless of the neurological level of
The ISNCSCI has substantial evidence of both intra- injury 94. Ambulatory rates are substantially higher for
rater and inter-rater reliability 82,83 and correlates with patients with incomplete injuries, but are variable and
other clinical, radiological and electrophysio­logical depend on the initial level of neurological injury 94.
proxies for injury severity. Going forward, work is
needed to better define the clinical relevance of sensori- Predicting neurological recovery. Several tools have
motor changes on the ISNCSCI to establish how many been developed to predict neurological recovery in
points of gain are to be considered ‘clinically important’. patients with SCI. One rule by van Middendorp et al.95
primarily relies on acute clinical examination features
SCI syndromes and can accurately predict long-term walking potential.
SCI patterns can broadly be defined as either complete Other tools, such as that developed by Wilson et al.96,
or incomplete. A third category, discomplete, describes use age, neurological examination and MRI features and
those with clinically complete injuries but persistent evi- can accurately predict the likelihood of long-term func-
dence of subclinical (for example, electrophysiological) tional independence, and Pavese et al.97 have generated
brain–muscle connectivity 84. For incomplete injuries, two simple models to predict urinary continence and
several patterns of neurological deficit are associated complete bladder emptying at 1 year after injury using
with SCI syndromes, including central cord syndrome, motor, sensory and spinal cord independent measure
Brown-Séquard syndrome (also known as hemi-cord (SCIM) subscale scores. Each of these might function
syndrome), anterior cord syndrome and posterior as useful tools in the future to help clinicians to estimate
cord syndrome (FIG. 5). long-term prognosis in the acute setting.
Central cord syndrome is the most common incom-
plete SCI syndrome and accounts for 15–25% of trauma­ Management
tic SCIs85. Most commonly, central cord syndrome is ‘Time is spine’ has emerged as a central concept in the
diagnosed in elderly patients with pre-existing cervical management of any patient with SCI. Similar to other
spondylosis and stenosis who present after a fall that acute CNS insults, functional neural tissue is progres-
results in cervical hyperextension86. Central cord syn- sively lost during the hours after SCI, making it crucially
drome is character­ized by a disproportionate motor important to rapidly diagnose patients and implement
impairment of the upper limbs, rather than the lower neuroprotective interventions during the acute injury
limbs, in addition to bladder dysfunction and varying phase. These treatments have the potential to substan-
degrees of sensory loss85. tially alter the long-term functional recovery of patients

NATURE REVIEWS | DISEASE PRIMERS VOLUME 3 | ARTICLE NUMBER 17018 | 9


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
PRIMER

Box 3 | ASIA Impairment Scale injury, intra-abdominal injury, thoracic injuries, pelvic
or long bone fractures and facial trauma. In all cases,
The American Spinal Injury Association (ASIA) Impairment Scale grade is a global transfers of care should be expedited to reduce diagno-
measure of injury severity and is largely based on the concept of sacral sparing (that is, sis and intervention times, and the transfer of patients
some degree of maintained perineal sensation, voluntary anal contraction and/or great to a specialized SCI care ­centre is recommended by
toe flexion indicating an incomplete lesion). The scale is used to determine the
AANS/CNS guidelines76.
grade of spinal cord injury (SCI), which ranges from ASIA Impairment Scale grade A
(the most severe injury with complete sensorimotor loss) to ASIA Impairment Scale
grade E (the least severe injury with no neurological deficit). Medical management
Haemodynamics. In the ICU, one of the most essential
Grade A
components of acute SCI management is the mainten­
Sensory or motor function below the neurological level (that is, the lowest segment
where sensorimotor function is normal on both sides) of injury, including absent sacral
ance of adequate spinal cord perfusion, through the
function (that is, no voluntary anal contraction, no great toe flexion, and no perineal, avoidance of systemic hypotension and support of
genital, anal pinprick or light touch sensation). mean arterial pressure. Hypotension is common post-
SCI; on the basis of findings from predominantly retro­
Grade B
Sensory but not motor function is preserved below the neurological level of injury,
spective clinical studies, the 2013 AANS/CNS SCI
including the distal sacral segments (S4–S5). No motor function is present more guidelines recommend avoiding systemic hypotension
than three levels below the neurological level, on either side of the body. (maintaining a systolic blood pressure of <90 mmHg)
and maintaining a mean arterial pressure between 85 and
Grade C
90 mmHg for the first 7 days post-injury 76,98. In addition,
Motor function below the neurological level of injury (including the distal sacral
segments) is preserved, with more than half of the key muscles (that is, elbow oxygen saturation should be maintained at ≥90% and
flexors and extensors, wrist extensors, finger flexors and abductors, hip flexors, prophylaxis to prevent deep venous ­thrombosis should
knee extensors, ankle dorsiflexors, long toe extensors and ankle plantar flexors) having be administered as soon as possible.
a grade of <3 on the ASIA motor score (against gravity without additional resistance).
Grade D Methylprednisolone sodium succinate. Historically,
Motor function below the neurological level of injury (including the distal sacral the most contentious issue surrounding the medical
segments) is preserved with more than half of the key muscles having a grade of ≥3 manage­ment of SCI is the suitability of the adminis-
(antigravity) on the ASIA motor score. tration of high-dose intravenous methylprednisolone
Grade E
sodium succinate (MPSS) in the acute phase of injury.
Neurologically intact patients (that is, sensorimotor function is normal in all segments) In preclinical evaluations, MPSS showed substantial
who previously had deficits secondary to a suspected SCI. promise as a neuroprotective agent 99–101. Subsequent
clinical evaluation of MPSS led to the completion
of three large randomized clinical trials (that is, the
and to provide meaningful improvements in quality of National Acute Spinal Cord Injury Studies (NASCIS)).
life (QOL). Management of patients with SCI is com- The second NASCIS study probably had the largest
plex and involves multiple stages of care, which can often impact on clinical practice102–104, and compared a high-
continue for years after the initial injury. dose 24‑hour infusion of MPSS with placebo, or with
naloxone. In the primary analysis, no significant dif-
Prehospital and hospital care setting ference in neurological recovery was observed between
For any patient with suspected spinal trauma and/or patients treated with MPSS or those who received
traumatic SCI, complete immobilization of the cranio– placebo. However, in a secondary analysis (involving
spinal axis should be maintained. In the prehospital set- patients treated ≤8 hours post-SCI), MPSS administra-
ting, this should involve transport with the use of a rigid tion resulted in a 5‑point increase in ASIA motor scores
spine board and application of a cervical collar. After at 6 months follow‑up compared with placebo admin-
rapid transport to the hospital, precautions, including istration103. In a 2012 Cochrane review, data from two
flat bedrest with a cervical collar, should be maintained other confirmatory randomized studies using the same
until confirmation or restitution of spinal stability. dose of MPSS were collated with the data from the sec-
Current AANS/CNS SCI guidelines state that ond NASCIS; overall, administration of a high-dose
manage­ment of acute patients with SCI, particularly 24‑hour infusion of MPSS results in a 4‑point increase
those with complete cervical injuries, should be per- in ASIA motor scores at long-term follow‑up compared
formed in an intensive care unit (ICU) with continuous with no treatment or placebo105. Regarding adverse
cardiac, haemodynamic and respiratory monitoring 76. effects, weak trends towards an increased incidence
Indeed, the existing low-quality (that is, from non-­ of gastrointestinal haemorrhage and wound infection
randomized, retrospective observational studies) clin- were noted with MPSS, but this did not achieve signifi­
ical evidence suggests that admission to an ICU, with cance. In line with these findings, a large proportion
the early identification and management of systemic of the spine surgery community began the routine
complications of SCI (including hypoxia, hypotension, administration of high-dose MPSS for patients with
pulmonary dysfunction and cardiovascular instability), SCI arriving at the hospital within 8 hours of injury 106.
has a role in reducing secondary injury and facilitat- However, numerous criticisms of this practice, and of
ing improved recovery 30,41. Care in the ICU is more the supporting body of literature, have emerged over the
important when considering concomitant injuries years107. Namely, critics have pointed to the potential for
that can accompany SCI, including traumatic brain increased complications, the use of subgroup analyses

10 | ARTICLE NUMBER 17018 | VOLUME 3 www.nature.com/nrdp


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
PRIMER

in the second NASCIS to prove effect, small positive guidelines recommended a 24‑hour administration of
effect sizes and methodological limitations in the two MPSS, started within the first 8 hours after injury, as
NASCIS II confirmatory trials, as arguments against the a treatment option109. As noted in recent written com-
routine use of MPSS within 8 hours. mentary, as well as in debate presentations at recent
Balancing the available perspectives and evidence, international neurosurgery meetings, this change in
the latest AANS/CNS SCI guidelines (that is, the 2013 recommendation has placed the clinician in a somewhat
guidelines) recommend against the use of MPSS for precarious position110,111. An upcoming 2017 AOSpine
SCI, arguing that the evidence of harm is more consist- guideline in the Global Spine Journal will suggest a
ent than the evidence of potential benefit 76,108. However, 24‑hour infusion of MPSS be offered to patients within
the stance adopted by the authors of these guidelines 8 hours of acute SCI as a treatment option. Ultimately,
has been somewhat controversial given that, despite the authors of this review feel that decisions surround-
no new prospective, randomized data on the topic in ing MPSS therapy should be left to the physician
the interval, the 2002 version of the AANS/CNS SCI involved, balancing the potential for benefit with the
potential for ­complications, given the characteristics of
the ­presenting patient.
a Dorsal column Lateral corticospinal tract*
(fine touch and Decompressive surgery. Surgical intervention is an
proprioception) Reticulospinal tract‡ essential cornerstone of the acute treatment for patients
with spinal trauma and acute SCI (FIG. 6). Overall, sur-
Lateral spinothalamic Rubrospinal tract‡
gery aims to realign the spinal column, re‑establish
tract (temperature
and pain) Vestibulospinal tract‡ ­spinal stability and decompression (that is, relief of bony
Tectospinal tract‡
or ligamentous compression) of the spinal cord. Surgery
Ventral spinothalamic tract typically involves open reduction and decompression
(crude touch and itch) Ventral corticospinal tract* paired with an instrumented fusion (for example, using
implanted metal hardware) to stabilize the spinal col-
b c umn in an anatomical position. The extent of surgery
is tailored to the anatomical site, as well as the severity
and extent of injury.
From a biological perspective, ongoing compres-
sion of the spinal cord is thought to exacerbate local
spinal cord ischaemia, thereby potentiating secondary
injury 112,113. Thus, decompressing the spinal cord early
after SCI should help to limit the zone of injury and
improve clinical outcomes. Indeed, evidence from a
d e systematic review and a meta-analysis of preclinical
studies showed that a longer duration of spinal cord
compression was typically associated with worsened
outcomes (including neurobehavioural recovery and
blood flow disturbances114. However, clinical class I
randomized evidence supporting the efficacy of early
surgical decompression is still lacking. That being said,
several prospective, non-randomized studies have sup-
ported the safety and efficacy of surgical decompres-
Figure 5 | Spinal cord injury syndromes. The major descending motor tracts
Nature Reviews arePrimers
| Disease in sion, including one study that noted an increased odds
yellow and the major ascending sensory tracts are in blue (part a), as also depicted of a ≥2 grade improvement in the ASIA Impairment
in FIG. 1a. The patterns of sensorimotor loss exhibited in patients with spinal cord injury
Scale grade with early (within 24 hours) decompression
(SCI) syndromes can be explained by damage to specific spinal cord tracts with sparing
of other tracts. For example, the disproportionate motor impairment of the upper limbs compared with late (>24 hours) decompressive surgery
compared with the lower limbs in patients with central cord syndrome (part b) might be in patients with cervical SCIs. In addition, data from
explained by the complete, non-selective injury to the corticospinal tract (which is this study showed a trend towards a reduced incidence
thought to transmit impulses related to fine hand and finger movements), but the of acute in‑hospital complications in the early surgery
preservation of the extrapyramidal tracts (which are thought to control gross leg and group, but imbalances between the treatment groups
proximal arm movements). In addition, the different levels of sensorimotor, pain and might have influenced outcomes. Other studies have
temperature loss in patients with Brown-Séquard syndrome (that is, the contralateral shown an association between early decompressive sur-
pain and temperature loss are detected several levels below that of the ipsilateral gery and significantly greater improvements in ASIA
sensorimotor loss) can be explained by the decussation of the lateral spinothalamic tract motor scale recovery 115; specifically, in patients with
over several spinal segments (part c). Anterior cord syndrome (part d) results in complete
ASIA Impairment Scale grade A injuries (BOX 3), reduced
motor paralysis due to damage to the corticospinal tract, loss of pain and temperature
sensation secondary to damage of the spinothalamic tract, but preservation of length of hospital stay, complication rates and health
light-touch sensation and proprioception (as the dorsal columns are generally care costs116. In another study, very early decompression
preserved by this injury). Posterior cord syndrome (part e) results in the reverse, with loss (≤8 hours) was associated with significant improvement
of light touch and proprioception but preservation of motor function, and pain and/or in 1 year SCIM scores and ASIA Impairment Scale
temperature sensation. grades117. No international clinical guideline regarding

NATURE REVIEWS | DISEASE PRIMERS VOLUME 3 | ARTICLE NUMBER 17018 | 11


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
PRIMER

a b Neuropathic arthropathy. Neuropathic or Charcot joint


arthropathy (that is, the slow progressive destruction of
a joint) can lead to deformity, overlying skin ulceration
and potentially fatal infections. The loss of sensation
that is common after SCI allows repeated micro­traumas
to go unnoticed, which promotes bone resorption121.
Furthermore, autonomic dysregulation can cause
hyperaemia of denervated joints, which promotes fur-
ther bone resorption. This arthropathy can occur in
any joint, including the hips, knees, ankles, shoulders,
elbows, spine and small joints.
Charcot arthropathy of the spine is often diag-
nosed 10–15 years after SCI and presents as deformity,
para­doxical pain (below the sensory level of injury),
c d a deterior­ation in neurological function and/or ­audible
sounds with movement. Treatment might be conserva-
tive, such as clinical and radiographic follow‑up, sympto-
matic (for example, treatment with analgesics or bracing)
or ­surgical (such as vertebral fusion)122.

Spasticity. Spasticity is the velocity-dependent increase in


muscle tone with exaggerated deep tendon reflexes that
results from injury to upper motor neurons. Spasticity
affects 65–78% of individuals with chronic SCI (>1 year
post-injury) and can substantially affect mobilization,
activities of daily living and sleep. Furthermore, spasti­city
can contribute to other local and systemic complications
of SCI, including the development of pressure ulcers,
Figure 6 | Surgical decompression and realignment ofNature the injured spinal
Reviews cord. Primers
| Disease contractures, fractures and cardiorespiratory decondition-
Arrows mark the cervical level 5 (C5)–C6 where the injury is centred. a | Preoperative CT ing 123. Treatment of spasticity may include physical ther-
imaging demonstrates a severe C5–C6 fracture dislocation (arrow), with compromise of apy, systemic pharmacological treatments (for example,
the central spinal canal. b | Preoperative MRI shows ongoing compression of the spinal clonidine or γ-aminobutyric acid (GABA)-ergic drugs,
cord (arrow) and a bright T2‑weighted signal in the surrounding ligaments that is
such as diazepam and baclofen), intrathecal pharmaco­
suggestive of disruption. c | Following surgery, including cervical traction, surgical
decompression and instrumented fusion anterior and posterior metal hardware can
logical treatments (for example, an intrathecal baclofen
be seen on the CT, and the restoration of appropriate spinal alignment. d | Successful pump), local botulinum toxin ­injections or ­surgery
decompression of the spinal cord can be seen on the postoperative MRI. (for example, tendon release surgery)123.

Systemic complications
the timing of decompressive surgery exists118. However, Several chronic systemic complications can substan-
one guideline supported by AOSpine has recently been tially affect the QOL of patients and their functional
completed and will be published in the Global Spine independence.
Journal in early 2017.
Cardiovascular. Analogous to changes observed dur-
Local complications ing the acute period of injury, chronic cervical or
Syringomyelia. Post-traumatic syringomyelia occurs thoracic SCI compromises sympathetic outflow from
in ~3% of patients with SCI and is characterized as a the CNS, which can lead to hypotension124 (FIG. 4). As
longitudinal fluid-filled cavity that can span many a result, ~60% of patients experience symptomatic
segments of the spinal cord and can lead to progres- orthostatic (or postural) hypotension (for example,
sive myelopathy that occurs months to years after SCI dizziness, weakness and syncope)125. These symptoms
(FIG. 7). Syringomyelia is distinct from the more com- occur consistently initially but gradually resolve over
mon post-injury cystic cavitations, which are smaller weeks to months, although they can persist for longer
and localized to the injury site. The pathophysiology in some patients125. Treatment includes the use of lower
of post-traumatic syringomyelia is not known but extremity compression stockings, abdominal binding or
might involve a one-way valve that gradually leads to medical management, including volume augmentation
intra­p arenchymal cerebrospinal fluid and/or inter­ (such as the use of hydration, salt tablets or fludrocorti-
stitial fluid accumulation119. Treatment depends on the sone) and/or peripheral vasoconstriction (for example,
clin­ical presentation and progression of symptoms120; with ­midodrine, ephedrine or droxidopa)126.
asymptomatic patients are monitored with serial clinical
and MRI examinations, whereas progressively, sympto- Autonomic dysreflexia. Autonomic dysreflexia is an
matic patients might undergo surgical decompression urgent condition that most commonly occurs in patients
by ­connecting the fluid cavity to the intrathecal space. with injuries at or above T6 (particularly, in those with

12 | ARTICLE NUMBER 17018 | VOLUME 3 www.nature.com/nrdp


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
PRIMER

complete injuries). Autonomic dysreflexia is caused Respiratory. Paralysis of the phrenic nerve, intercostal
by the presence of a noxious stimulus below the level muscles and/or abdominal muscles leads to reduced
of injury (such as bladder distension, bowel impaction lung capacity, ineffective cough and accelerated fatigue
or pressure sores), which causes a reflex overstimula- with respiratory demand129. As a consequence, patients
tion of spinal sympathetic neurons, leading to vaso­ experience recurrent pneumonia, atelectasis (that is,
constriction and dangerous acute hypertension127. As a alveolar collapse) and pleural effusion (fluids around
response, para­sympathetic outflow increases above the the lungs), and are more likely to have sleep apnoea and
injury level and sympathetic outflow can be inhibited, respiratory failure130. Whereas long-term rehabilitation,
depending on the injury level, which leads to vasodila- which promotes cardiorespiratory conditioning, may
tion, headache, sweating and sinus congestion. Prompt be beneficial, the respiratory defects themselves restrict
treatment requires upright positioning of the patient, rehabilitation capacity and long-term independence.
removal of the triggering stimulus and pharmacological Owing to this, respiratory complications are the lead-
anti-­hypertensives for refractory cases128. Episodes of ing cause of mortality in patients with chronic SCI.
life-threatening autonomic dysreflexia can occur in both In individ­uals with high cervical injuries, or those with
acute and chronic stages of injury, ­making long-term poor respiratory reserve, lifelong ventilator dependency
prevention key by avoiding noxious stimuli (for example, can also result 131,132.
by frequent bowel and bladder care and r­ epositioning to
avoid pressure sores). Secondary immunodeficiency. As previously men-
tioned, the disruption of CNS input to immune organs
can result in the systemic dysfunction of macrophages,
a b T cells, B cells and natural killer cells in a process known
as immune paralysis. The clinical manifestation of this is
an increased susceptibility to infections, such as pneumo-
nia, urinary tract infections and wound infections133,134.
Although the cause of immune paralysis continues to
be investigated, cervical or high thoracic injuries have
C2
been shown to cause interruption of the sympathetic
C3 innervation of lymphatic organs and are associated with
rapid splenic atrophy 133. No accepted management for
C4 c
­secondary immunodeficiency exists.
C5
C6
Genitourinary and gastrointestinal. Dysfunction of the
genitourinary and gastrointestinal systems increases care
C7 requirements, the risk of infection and can be a source
T1
of substantial social and psychological stress in patients
with SCI. Injuries at or above L1–L2 interrupt innerva-
T2 tion of the detrusor, or the bladder muscle, and urinary
d e sphincters, which can cause an inability to empty the
T3 bladder, acontractile bladder, urinary incontinence and
T4 recurrent infections135,136. Management includes urethral
catheterization every few hours, the surgical creation
T5 of a urinary stoma, injections of botulinum toxin and
T6
pharmacological therapies (such as anticholinergics or
α-blockers)124.
T7
The neurological level of injury can also substantially
T8 affect sexual function. For example, injuries above T11
T9
f can affect psychogenic arousal (that is, erection or vaginal
lubrication as a result of arousal in the brain) with pres-
T10 ervation of reflexive arousal (that is, erection or vaginal
lubrication as a result of genital stimulation) and the abil-
T11 ity to orgasm. Conus injuries (that is, injuries in the sacral
segment) can interfere with reflexive arousal but preserve
T12 psychogenic arousal. T12–L2 injuries with sacral segment
sparing can preserve all sexual functions137.
Approximately 39% of patients with SCI report that
Figure 7 | Post-spinal cord injury syrinx. T2‑weighted MRI of the cervical (parts a–c)
Nature Reviews | Disease Primers bowel dysfunction significantly reduces their QOL124. SCI
and thoracic (parts d–f) spine in sagittal (part a and part d) and axial (parts b, c, e and f)
planes shows a post-traumatic syrinx within the spinal cord parenchyma (white arrows) can interrupt the voluntary control of the anal sphincter
and kyphosis (that is, forward bending) of the thoracic spine at the initial site of the (causing faecal retention) and/or the parasympathetic
spinal cord injury (SCI; black arrow in part d). The syrinx extends well beyond the innervation of the bowel (in patients with lumbosacral
mid-thoracic site of the SCI into the high cervical spinal cord, which probably causes injury). Both cases lead to constipation, an increased risk
upper limb pain. of infection and stress for patients. Treatments can range

NATURE REVIEWS | DISEASE PRIMERS VOLUME 3 | ARTICLE NUMBER 17018 | 13


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
PRIMER

from dietary fibre intake, digital rectal stimulation or dis­ Physical rehabilitation is focused on regaining func-
impaction and the use of suppositories, to ­implantation tion, enhancing any remaining function and prevent-
of an electrical stimulator or colostomy 138,139. ing complications. Key components of rehabilitation
are strength training, cardiovascular-focused exercise,
Pressure sores. Pressure sores cause pain, increased care respir­atory conditioning, transfer or mobility training
requirements and can be life-threatening if not promptly and stretching to prevent muscle contractures (that is,
treated. Sores most commonly occur on the buttocks the permanent shortening of muscle). The patient’s
(31%), lateral thighs (26%), sacrum (18%), feet (7%) and progress helps to dictate the level of ongoing care that
ankles (4%)124. Prevention of pressure sores requires daily is needed in the community and the use of assistive
inspection and cleaning of the skin, but also a relief of devices for daily living. Further high-quality (that is,
the pressure on each region every few hours. Once a sore level 1–2) trials of physical rehabilitation are required
develops, diligent aseptic technique, debridement, dress- to validate the intuitive efficacy and compare specific
ing and nutritional support are vital to halt progression to treatment modalities144. Interestingly, physical rehabili-
life-threatening and limb-­threatening infections140. tation can induce significant changes in cellular signal-
ling and growth factor expression145. Early mobilization
Neurogenic heterotopic ossification. Approximately increases endogenous growth factor levels (such as
10–53% of patients with chronic SCI form ectopic bone insulin-like growth factor 1) and axon regeneration in
in the connective tissue around joints, in a process called animal ­models145. However, in clinical practice, venti-
neurogenic heterotopic ossification. This ossification lator dependence, poor vascular tone, neuropathic and
occurs most commonly in the large joints (for example, somatic pain, psychosocial challenges and resource
the hips, knees, elbows or shoulders), tends to develop limitations in acute care institutions can make early
within months of the SCI and presents with localized mobilization challenging 30. These important clinical
pain, redness, low-grade fever and increased spasticity 141. barriers are often overlooked but represent formidable
The exact cause of neurogenic heterotopic ossification is ­overarching challenges to recovery.
not known, but it might involve a combination of local, Weight-supported locomotor training (WSLT)
humoral and neuro-immunological factors. Management uses assisted devices (such as Hocoma’s Lokomat and
can include physical therapy, pharmacological therapy HealthSouth’s AutoAmbulator) and therapists to dynam-
(such as bisphosphonates and/or NSAIDs), radiation ically support the patient’s weight while they attempt
therapy or surgical resection of the ossification142. locomotion on a treadmill or open ground. The therapy
seeks to enhance the remaining connectivity between
Neuropathic pain. Neuropathic pain is experienced regions above the SCI and the locomotor central p ­ attern
by up to 40% of patients with chronic SCI, has a mean generator (that is, a region of neurons that, when activ­
onset of 1.2 years after injury and can have a substan- ated, can initiate locomotion in the absence of sensory
tial effect on the psychological well-being and QOL input or input from the brain) with the spinal cord.
of patients. The mechanism underlying injury-level WSLT has been shown to improve assisted mobility,
pain is thought to be sprouting of spinal cord fibres cardiorespiratory status and to prevent pressure sores
around damaged nerve roots, leading to inappro­priate and joint-related complications of SCI. A randomized,
activ­ation of primary afferent fibres and the initi­ation single-blind trial (n = 146) comparing 12 weeks of WSLT
of pain by normally non-noxious stimuli (that is, allo­ versus similar intensity physical rehabilitation found no
dynia). Below-injury-level pain is hypothesized to significant difference in outcomes, but both groups had
occur owing to a loss of spinal and supraspinal inhib­ improvements in locomotion at 6 months, highlighting
itory signalling combined with the potentiation of brain the importance of intensive rehabilitation146.
pain-responsive areas. Neuropathic pain can be treated Occupational therapy focuses on integrating adaptive
pharmacologically (for example, using antidepressants, devices into people’s daily lives to maximize functional
anti­convulsants and/or opioids), surgically (such as independence at home and at work. Devices might
implanted spinal cord stimulators, deep brain stimulators include wheelchairs, lifts, braces, orthoses, environmen-
and dorsal root entry zone lesioning) or through non-­ tal control units (such as lights, television or phones),
allopathic treatments (such as a­ cupuncture, ­massage and bathroom equipment (such as showering or toileting),
­cognitive–­behavioural therapy)143. vehicle modifications for driving and others147. The US
Department of Health and Human Services maintains a
Rehabilitation database (www.AbleData.com) of accessibility devices to
Rehabilitation requires an interdisciplinary approach help inform patients.
involving nurses, physicians, dieticians, psychologists,
physiotherapists, social workers, recreation therapists, Functional electrical stimulation
speech therapists, orthotists and child life specialists. Functional electrical stimulation (FES) uses small
Rehabilitation can have significant effects on long-term pulses of current to activate muscles and has been
health by helping patients to recover as much function as successfully used in the upper extremities for eating,
possible, prevent secondary complications, understand gripping and writing. In the lower extremities, FES has
the extent of their injuries, cope with loss of indepen­ been connected to a wheeled walker for ambulation (for
dence and address other practical challenges, such as example, the Parastep by Sigmedics, Inc.) and to station­
vocational and financial concerns. ary ­bicycles (for example, ERGYS 3 by Therapeutic

14 | ARTICLE NUMBER 17018 | VOLUME 3 www.nature.com/nrdp


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
PRIMER

Alliances, Inc. and RT300 by Restorative Therapies). Economic impact


FES can also be surgically implanted with electrodes on The financial burden of SCI on patients, their famil­
the anterior sacral nerve roots to provide patients with ies and society is substantial. Direct health care costs
controllable bowel or bladder function. Typically, the and living expenses vary substantially based on the
implanted sacral nerve stimulator, such as the Vocare geo­graphical region and the age, survival and injury
Bladder System (Finetech Medical), requires surgical severity of individual patients. For example, in the
lesioning of dorsal sensory roots to improve continence, United States, the lifetime cost for providing care to a
but an open-label pilot study is under way to assess the patient 25 years of age with an ASIA Impairment Scale
system in patients with SCI without nerve sacrifice, with grade A injury is ~$2.3 million for thoracic injuries,
results expected by 2018 (NCT02978638). Importantly, ~$3.5 million for C5–C8 injuries and ~$4.7 million
FES can also enhance neuroplasticity and decrease the for C1–C4 injuries over the course of the patient’s life-
systemic complications of chronic SCI in patients148. time. In addition, i­ndirect costs (including lost wages
In addition, FES-based exercises can double the oxy- and benefits) are estim­ated at ~$72,000 per year 2. Even
gen uptake, triple the ventilation rate and improve the small improvements in function, such as mobility and
overall muscle to fat ratio in the body 149,150. FES is an manual d ­ exterity, can substantially reduce these costs,
actively researched field, with the next generation of highlighting the ­economic importance of the ‘time is
devices integrating more-advanced closed-loop feed- spine’ concept.
back systems, greater MRI compatibility and novel
stimulation programmes to reduce adverse effects and Outlook
improve efficacy 151. Improving translation
Although numerous treatments have generated pos-
Quality of life itive results in preclinical models of SCI, translation
QOL in patients with SCI is most often defined by the to patients has been challenging. Typically, preclinical
ability of patients to be independent of assisted care and studies use animal models with highly standardized inju-
to hold meaningful employment152. The most frequently ries, treatment paradigms and assessment techniques in
used subjective measure of QOL is the Satisfaction ­animals that are genotypically and pheno­typically simi-
with Life Scale (SWLS), and the most commonly used lar, which is in contrast to the heterogeneity of patients.
objective measure is the 36‑Item Short-Form Health As a result, an effective therapeutic approach from a
Survey (SF‑36)153. A new scale to assess QOL in patients ­single animal model might only be translatable to a sub-
with SCI is the SCI-QOL, a patient-reported outcome set of individuals within a clinical trial that often assesses
measure comprising 18 domains, including metrics for a wide array of patients. This requires higher recruit-
­physical, emotional and social aspects154. ment to sufficiently power the study and often necessi­
tates controversial subgroup analyses167. To overcome
Factors associated with QOL this, one strategy is to embark on clinical trials only
Patients with SCI have been shown to have a lower QOL after a treatment has demonstrated efficacy in multi­ple
than the general population in a meta-analysis155. Out animal models and species. Although this decreases the
of functional impairment (the loss or abnormality of number of potentially translatable therapies, it might
anatomical function), disability (a functional limitation identify the highest yield treatments. Another strategy
for specific activities) and handicap (a disadvantage is to narrow the inclusion criteria of studies using clin-
in acting in a certain role), handicap is most strongly ical factors and biomarkers, which can provide insight
associ­ated with QOL in patients with SCI. Other studies into the underlying pathophysiology 168. Together, these
have indicated that the severity and level of injury is sig- approaches might decrease variability in clinical trials
nificantly associated with QOL (that is, individuals with (and recruitment requirements) while increasing the
higher-level and more-severe injuries showed signifi­ statistical power of the trial.
cantly lower QOL)156,157. However, conflicting results
have been reported in other studies158,159. Other factors Common data elements
that are associated with QOL include advanced age and The SCI field needs high-quality large-scale data sets to
lower QOL160,161, and for both functional and psycho­ better understand the heterogeneity between patients,
logical outcome, lengthier duration of SCI is associated as this affects their response to treatment and our ability
with a more positive assessment of QOL160,162. to predict outcomes. Generating these data sets can be
Social support, as indicated by marriage or cohabit­ logistically challenging as patients present emergently
ation, and employment have a positive effect on QOL and require complex care169, but several registries have
after SCI163,164. A higher level of education165 and the been developed, including the North American Clinical
ability to walk without assistance161 were associated with Trials Network SCI Registry 170, the International Spinal
higher QOL scores. SCI-related morbidities, including Cord Society SCI Data Sets171 and the National Spinal
neuro­pathic pain, spasticity and bladder, bowel and Cord Injury Statistical Center database172, among o ­ thers.
­sexual dysfunction were all associated with a lower Large clinical trials have also contributed patient records,
QOL 166. In  general, carefully designed studies are but data elements need to be standardized to harmo-
required to give us a better understanding of how we nize data sets and to draw meaningful conclusions.
can better prognosticate and inform long-term strategies Towards this goal, the National Institute of Neurological
to improve QOL for those with SCI. Disorders and Stroke (NINDS) within the US NIH has

NATURE REVIEWS | DISEASE PRIMERS VOLUME 3 | ARTICLE NUMBER 17018 | 15


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
PRIMER

Table 1 | Selected planned or ongoing trials in patients with spinal cord injury increase in the levels of hepatic enzymes — was reported.
A larger multicentre efficacy trial is currently ongoing
Treatment Stage ClinicalTrials.gov (NCT01828203).
identifier or refs
Riluzole (a sodium channel blocker) has improved
Pharmacological neurobehavioural and pathological outcomes in animal
Minocycline* Phase III NCT01828203 models of SCI and is thought to prevent continuous
Riluzole* Phase IIb/III NCT01597518 activation of neuronal voltage-gated sodium chan-
nels, preventing cellular swelling and death, in addi-
Granulocyte colony-stimulating factor* Phase I/II 219,220
tion to reducing excitotoxicity 176. Data from a phase I
Cethrin ‡
Phase II/III NCT02669849 trial showed an improvement in ASIA motor scores in
Anti-Nogo-A antibody‡ Phase II NCT00406016 patients with cervical level injuries, 90 days after riluzole
Procedural treatment, compared with non-treated patients matched
from a historical registry cohort 177. Three patients had
Systemic hypothermia Phase II/III 221,222
temporary borderline severe increases in the levels of
Cerebrospinal fluid drainage Phase II NCT02495545 liver enzymes, but no serious adverse events were attrib-
Blood pressure augmentation Phase II NCT02495545 uted to the drug. Currently, a phase II/III multicentre,
Neuromodulation randomized trial (RISCIS) is enrolling patients and is
supported by AOSpine (NCT01597518).
Spinal cord stimulation Phase I NCT02592668 Basic fibroblast growth factor (bFGF; also known as
Deep brain stimulation Phase I NCT02006433 fibroblast growth factor 2) is an important mediator of
Cell-based strategies angiogenesis, has a key role as a morphogen in embryo­
logical development and is used in vitro to maintain
Oligodendrocyte precursor cells Phase I/II NCT02302157
pluripotency of many cells types, including neural stem
Schwann cells Phase I NCT01739023 cells178. In animal models, bFGF can promote neuro-
Umbilical cord-derived stem cells Phase III NCT02481440 protection against excitotoxicity and can reduce injury
Bone marrow-derived mesenchymal stem cells Phase II NCT02570932 mediated by free radicals179,180. A structural analogue
to bFGF (SUN13837) has been assessed in a phase I/II
Bioengineering
­randomized trial with results pending.
Robotic exoskeletons Phase I NCT02322125 Finally, the use of systemic hypothermia as a potential
Functional peripheral electrical stimulation Phase I/II NCT01479777 neuroprotective strategy is under clinical investigation in
Implantable bioengineered scaffolds or matrices Phase III NCT02138110 a phase II/III study: the ARCTIC trial. Hypothermia can
*Denotes neuroprotective studies. ‡Denotes neuroregenerative studies.
decrease the basal metabolic rate of the CNS after injury
and provides an anti-inflammatory effect 181. Systemic
intravascular cooling to 33 °C after acute hospital admis-
developed a set of common data elements for SCI. The sion in patients with complete SCI is safe and associated
2014 common data elements are classified along a spec- with increased rates of ASIA Impairment Scale grade
trum according to their use and validation in SCI and are conversion compared with historical controls182.
grouped by field, including demographics, care, electro­
diagnostics, functional, imaging, neuro­logical, pain, Neuroregenerative treatments. The RHOA pathway can
QOL and psychological. Upcoming studies and registries negatively affect axonal and neurite growth, and mol­
should apply these elements to their data collection for ecules that activate this pathway are upregulated follow-
the ultimate benefit of all patients. ing SCI183. A specific bacterially derived toxin, known
as VX‑210, can inhibit RHOA-mediated inhibition of
Current clinical studies axonal growth, leading to enhanced regeneration and
The past several decades have seen a flurry of preclinical improved behavioural outcomes in rodent models184.
SCI research that has given rise to a host of promising Cethrin (Alseres Pharmaceuticals), a recombinant ver-
therapeutic advances, each of which are in various stages sion of VX‑210, showed promise in preclinical studies
of clinical development 60,173. Pharmaco­logical agents and no serious drug-related adverse events were noted
that are currently being investigated can broadly be in a phase I/IIa dose-escalation study in patients with
classified as either neuroprotective or n
­ euroregenerative ASIA Impairment Scale grade A cervical and thoracic
(TABLE 1). injuries185. Although this study was uncontrolled, ASIA
motor score recovery at 12 months was superior to his-
Neuroprotective treatments. Minocycline (a structural torical recovery rates185. A phase IIb/III study is under
analogue of the antibiotic tetracycline) can induce way (NCT02669849).
neuroprotection in animal models of SCI, presumably As previously mentioned, Nogo‑A is found in CNS
through reducing oligodendrocyte apoptosis and by myelin and presumably has a role in preventing the for-
reducing local inflammation174,175. A phase II place- mation of new functional connections post-SCI. Anti-
bo-controlled, randomized study showed an improve- Nogo‑A antibodies have shown promise in promoting
ment of 6 points in the ASIA motor score in 1 year axonal regeneration in preclinical SCI studies186, and
after delivery of minocycline for 7 days compared with a phase I study has been completed, with a phase II
­placebo, and only one adverse event — a transient ­placebo-controlled European trial under way187.

16 | ARTICLE NUMBER 17018 | VOLUME 3 www.nature.com/nrdp


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
PRIMER

Biomaterials are under intense investigation, as they Further efforts at cell transplantation include the
can be engineered to mimic the architecture of lost ECM transplantation of human embryonic stem cell-­derived
in the spinal cord and can structurally support cell migra- oligodendrocyte progenitor cells (such as the Geron
tion and axonal regrowth. A phase III trial in thoracic trial), but this trial was discontinued for financial r­ easons.
SCI, entitled INSPIRE, is under way in the United States, Fortunately, renewed funding has allowed Asterias
with a biodegradable Neuro-Spinal Scaffold (InVivo Biotherapeutics, Inc. to restart the study of these cells
Therapeutics) to assess the safety and improvements in as a phase I/IIa dose-escalation study, in which their
ASIA Impairment Scale grade, motor scores and sensory product, AST‑OPC1, is transplanted into the subacutely
scores (NCT02138110). injured cervical spinal cord (NCT02302157). Other cell
types that are under clinical investigation include human
Cellular transplantation. Transplantation of various Schwann cells and umbilical cord blood mononuclear
cell types to repair the injured spinal cord is an exciting cells, among others. One phase I/II trial involving umbili-
therapeutic concept188 and addresses the extensive loss of cal cord blood mononuclear cells found that the addition
tissue caused by SCI that cannot be replaced by endogen­ of intensive locomotor training to cell-based therapy can
ous repair processes. In addition, transplanted cells can significantly enhance functional recovery in patients with
replace lost cells, modulate the injury environment and chronic injuries205.
stimulate synergistic regenerative programmes173. Any Biotechnology-led trials include the recently termin­
specific cell type might have one or more of these actions, ated testing of a human fetal neural stem cell product
which remains an area of active investigation189. (HuCNS-SCs; StemCells, Inc.) and an ongoing trial by
The various cell types that have been assessed in pre- Neuralstem of transplanted NSI‑566, which are stem
clinical studies include neural stem or precursor cells, cells derived from the human fetal spinal cord. Other
oligo­dendrocyte precursor cells, olfactory ensheathing possible therapies include adult autologous stem cells
cells (OECs), Schwann cells and umbilical cord mesen- (RhinoCyte, Inc.), although this therapy is still at pre-
chymal stem cells, among others173,190. Cell transplanta- clinical stages, and human glial-restricted progenitor
tion into the transected cord has been shown to promote product (Q‑cells; Q Therapeutics). Numerous novel
the recovery of motor function, including coordinated conventional drugs, such as anti-Nogo-A antibodies, are
walking 191, paw use and climbing 192, in addition to also currently being evaluated in clinical trials (TABLE 1).
improved bladder function193 and phrenic nerve activ-
ity 194 in ­animal models. Importantly, neural precursor Neuromodulation, robotics and future directions. Several
cells and adult olfactory tissue are also effective when neuromodulatory approaches, involving the focused
transplanted 1 month after SCI in rats, which is a time delivery of electrical current to the CNS, are under study
point that is considered to model stable, chronic SCI for the treatment of SCI. Specifically, spinal cord stimula-
in humans. tion using surgically implanted electrodes in the epidural
Mechanistically, transplanted cells can improve space over the conus medullaris, has improved functional
regeneration by promoting axonal growth (observed and locomotion-related outcomes in patients with chronic
with OECs), remyelinating denuded axons them- SCI206. This trial is no longer active. In addition, although
selves (observed with Schwann cells and oligodendro- not under investigation in the clinic, preclinical studies
cytes, among others) and supporting remyelination have shown that stimulation of deep brain centres in the
by endogen­ous oligodendrocytes195,196. In addition, region of the mesencephalic locomotor region resulted in
­factors that are secreted by transplanted cells can bene­ substantial improvement in functional deficit in rat SCI
ficially modulate the environment and promote axon models207. Finally, neuroprosthetic brain–computer inter-
regeneration197,198. faces have successfully restored upper limb function in a
Several trials have tested the safety and preliminary paralysed patient; in this case, the device was implanted
efficacy of cell transplantation in patients with SCI. The into the motor cortex of a patient with a complete cervi-
first human trial confirmed the safety of transplanta- cal injury and stimulated specific hand and wrist muscle
tion of purified OECs into the spinal cord199. However, groups, which allowed functional control of motor out-
subsequently studies that transplanted mucosal ­tissue, put without transmission through the spinal cord208. The
as opposed to purified OECs, reported conflicting next steps are to reduce the movement-retraining process
results200,201. Other trials have investigated other trans- and to make this technology feasible for everyday use.
planted cells, including OECs and olfactory nerve fibro- The use of robotics is also beginning to have a more
blasts, Schwann cells and a combination of OECs and substantive role in granting patients with SCI the abil-
Schwann cells202,203. A systematic review of the use of ity regain functionality. In 2014, the US FDA approved
OECs in SCI supported the positive findings found in the first robotic exoskeleton (ReWalk; ReWalk Robotics,
other trials204. More recently, data from a phase I trial Inc.) for use in patients with paraplegia, which fits around
reported motor and sensory improvements and no seri- the legs and back of patients to facilitate sitting, standing
ous adverse events 1 year post-transplantation of auto­ and walking 209,210. Other devices include Indego (Parker
logous mucosal OECs and olfactory nerve fibroblasts into Hannifin Corporation), Ekso (Ekso Bionics), REX (Rex
the spinal cords of patients with AISA Impairment Scale Bionics) and Hybrid Assistive Limb (Cyberdyne, Inc.)210.
grade A injuries (n = 6)202. However, large sample sizes It is anticipated that, as technology improves, robotics
and long follow‑up periods will be required to confirm will be used in conjunction with the discussed biological
safety and efficacy 200. treatments to help optimize outcomes in the long term.

NATURE REVIEWS | DISEASE PRIMERS VOLUME 3 | ARTICLE NUMBER 17018 | 17


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
PRIMER

1. Noonan, V. K., Dvorak, M. F. & Fehlings, M. G. in Na+‑dependent glutamate transport. J. Neurosci. 19, 50. Barnabe-Heider, F. et al. Origin of new glial cells in
Critical Care in Spinal Cord Injury (ed. Fehlings, M. G.) RC16 (1999). intact and injured adult spinal cord. Cell Stem Cell 7,
6–20 (Future Medicine Ltd, 2013). 25. Li, S. & Stys, P. K. Mechanisms of ionotropic glutamate 470–482 (2010).
2. [No authors listed.] Spinal cord injury facts and figures receptor-mediated excitotoxicity in isolated spinal cord 51. Mao, Y., Nguyen, T., Sutherland, T. & Gorrie, C. A.
at a glance. J. Spinal Cord Med. 37, 117–118 (2014). white matter. J. Neurosci. 20, 1190–1198 (2000). Endogenous neural progenitor cells in the repair
3. Singh, A., Tetreault, L., Kalsi-Ryan, S., Nouri, A. 26. Norenberg, M. D., Smith, J. & Marcillo, A. The of the injured spinal cord. Neural Regen. Res. 11,
& Fehlings, M. G. Global prevalence and incidence pathology of human spinal cord injury: defining the 1075–1076 (2016).
of traumatic spinal cord injury. Clin. Epidemiol. 6, problems. J. Neurotrauma 21, 429–440 (2004). 52. Bradbury, E. J. et al. Chondroitinase ABC promotes
309–331 (2014). 27. Tator, C. H. Update on the pathophysiology and functional recovery after spinal cord injury. Nature
A comprehensive overview of the global pathology of acute spinal cord injury. Brain Pathol. 5, 416, 636–640 (2002).
epidemiology of traumatic SCI divided by data 407–413 (1995). 53. Li, G. et al. Mdivi‑1 inhibits astrocyte activation
available from geographical regions. 28. Milhorat, T. H., Capocelli, A. L., Anzil, A. P., and astroglial scar formation and enhances axonal
4. Cripps, R. A. et al. A global map for traumatic spinal Kotzen, R. M. & Milhorat, R. H. Pathological basis regeneration after spinal cord injury in rats.
cord injury epidemiology: towards a living data of spinal cord cavitation in syringomyelia: analysis of Front. Cell. Neurosci. 10, 241 (2016).
repository for injury prevention. Spinal Cord 49, 105 autopsy cases. J. Neurosurg. 82, 802–812 (1995). 54. Tator, C. H. Review of treatment trials in human spinal
493–501 (2011). 29. McKeon, R. J., Schreiber, R. C., Rudge, J. S. & Silver, J. cord injury. Neurosurgery 59, 957–987 (2006).
5. Noonan, V. K. et al. Incidence and prevalence of spinal Reduction of neurite outgrowth in a model of glial 55. Zhang, N., Fang, M., Chen, H., Gou, F. & Ding, M.
cord injury in Canada: a national perspective. scarring following CNS injury is correlated with the Evaluation of spinal cord injury animal models. Neural
Neuroepidemiology 38, 219–226 (2012). expression of inhibitory molecules on reactive Regen. Res. 9, 2008–2012 (2014).
6. New, P. W., Farry, A., Baxter, D. & Noonan, V. K. astrocytes. J. Neurosci. 11, 3398–3411 (1991). 56. Nout, Y. S. et al. Animal models of neurologic
Prevalence of non-traumatic spinal cord injury in 30. Ahuja, C. S., Martin, A. R. & Fehlings, M. Recent disorders: a nonhuman primate model of spinal
Victoria, Australia. Spinal Cord 51, 99–102 (2013). advances in managing a spinal cord injury secondary cord injury. Neurotherapeutics 9, 380–392 (2012).
7. Chen, Y., He, Y. & DeVivo, M. J. Changing to trauma. F1000Res. 5 (F1000 Faculty Rev), 1017 57. Kwon, B. K. et al. Large animal and primate models
demographics and injury profile of new traumatic (2016). of spinal cord injury for the testing of novel therapies.
spinal cord injuries in the United States, 1972–2014. 31. Cregg, J. M. et al. Functional regeneration beyond Exp. Neurol. 269, 154–168 (2015).
Arch. Phys. Med. Rehabil. 97, 1610–1619 (2016). the glial scar. Exp. Neurol. 253, 197–207 (2014). 58. Fisher, M. et al. Update of the stroke therapy
A cross-sectional analysis of longitudinal data 32. Silver, J. & Miller, J. H. Regeneration beyond the glial academic industry roundtable preclinical
that highlights key trends in the demographics scar. Nat. Rev. Neurosci. 5, 146–156 (2004). recommendations. Stroke 40, 2244–2250 (2009).
of individuals with traumatic SCIs. An in‑depth review of the glial scar, ECM 59. Tator, C. H. Acute spinal cord injury: a review of recent
8. van den Berg, M. E., Castellote, J. M., proteoglycans and the behaviour of regenerating studies of treatment and pathophysiology. Can. Med.
Mahillo-Fernandez, I. & de Pedro-Cuesta, J. Incidence neurons in the injured CNS microenvironment. Assoc. J. 107, 143–145 (1972).
of spinal cord injury worldwide: a systematic review. 33. Wanner, I. B. et al. Glial scar borders are formed by 60. Wilson, J. R., Forgione, N. & Fehlings, M. G. Emerging
Neuroepidemiology 34, 184–192 (2010). newly proliferated, elongated astrocytes that interact therapies for acute traumatic spinal cord injury. CMAJ
9. Lenehan, B. et al. The epidemiology of traumatic to corral inflammatory and fibrotic cells via 185, 485–492 (2013).
spinal cord injury in British Columbia, Canada. Spine STAT3‑dependent mechanisms after spinal cord injury. 61. Guha, A., Tator, C. H. & Rochon, J. Spinal cord blood
37, 321–329 (2012). J. Neurosci. 33, 12870–12886 (2013). flow and systemic blood pressure after experimental
10. Devivo, M. J. Epidemiology of traumatic spinal cord 34. Anderson, M. A. et al. Astrocyte scar formation aids spinal cord injury in rats. Stroke 20, 372–377 (1989).
injury: trends and future implications. Spinal Cord 50, central nervous system axon regeneration. Nature 62. Guha, A. B. & Tator, C. H. Acute cardiovascular effects
365–372 (2012). 532, 195–200 (2016). of experimental spinal cord injury. J. Trauma 28,
11. DeVivo, M. J. & Chen, Y. Trends in new injuries, 35. Rolls, A., Shechter, R. & Schwartz, M. The bright side 481–490 (1988).
prevalent cases, and aging with spinal cord injury. of the glial scar in CNS repair. Nat. Rev. Neurosci. 10, 63. Schwab, J. M., Zhang, Y., Kopp, M. A., Brommer, B.
Arch. Phys. Med. Rehabil. 92, 332–338 (2011). 235–241 (2009). & Popovich, P. G. The paradox of chronic
12. Wu, J. C. et al. Effects of age, gender, and socio- 36. Filbin, M. T. Myelin-associated inhibitors of axonal neuroinflammation, systemic immune suppression,
economic status on the incidence of spinal cord injury: regeneration in the adult mammalian CNS. Nat. Rev. autoimmunity after traumatic chronic spinal cord
an assessment using the eleven-year comprehensive Neurosci. 4, 703–713 (2003). injury. Exp. Neurol. 258, 121–129 (2014).
nationwide database of Taiwan. J. Neurotrauma 29, 37. Forgione, N. & Fehlings, M. G. Rho–ROCK inhibition in 64. Ko, H., Dittuno, J., Graziani, V. & Little, J. The pattern
889–897 (2012). the treatment of spinal cord injury. World Neurosurg. of reflex recovery during spinal shock. Spinal Cord 37,
13. Wu, J. C. et al. Epidemiology of cervical spondylotic 82, e535–e539 (2014). 402–409 (1999).
myelopathy and its risk of causing spinal cord injury: 38. Lee, J. K. et al. Assessing spinal axon regeneration 65. Ploumis, A., Yadlapalli, N., Fehlings, M. G., Kwon, B. K.
a national cohort study. Neurosurg. Focus 35, E10 and sprouting in Nogo-, MAG-, and OMgp-deficient & Vaccaro, A. R. A systematic review of the evidence
(2013). mice. Neuron 66, 663–670 (2010). supporting a role for vasopressor support in acute SCI.
14. Krause, J., Sternberb, M., Lottes, S. & Maides, J. 39. Geoffroy, C. G. & Zheng, B. Myelin-associated Spinal Cord 48, 356–362 (2010).
Mortality after spinal cord injury: an 11‑year inhibitors in axonal growth after CNS injury. 66. Lehmann, K. G., Lane, J. G., Piepmeier, J. M.
prospective study. Arch. Phys. Med. Rehabil. 78, Curr. Opin. Neurobiol. 27, 31–38 (2014). & Batsford, W. P. Cardiovascular abnormalities
815–821 (1997). 40. Fehlings, M. G. & Tator, C. H. The relationships among accompanying acute spinal cord injury in humans:
15. LaPlaca, M. C., Simon, C. M., Prado, G. R. the severity of spinal cord injury, residual neurological incidence, time course and severity. J. Am. Coll.
& Cullen, D. K. CNS injury biomechanics and function, axon counts, and counts of retrogradely Cardiol. 10, 46–52 (1987).
experimental models. Prog. Brain Res. 161, 13–26 labeled neurons after experimental spinal cord injury. 67. Guly, H. R., Bouamra, O., Lecky, F. E. & Trauma Audit
(2007). Exp. Neurol. 132, 220–228 (1995). and Research Network. The incidence of neurogenic
16. Choo, A. M. et al. Contusion, dislocation, and 41. Tator, C. H. & Fehlings, M. G. Review of the secondary shock in patients with isolated spinal cord injury in
distraction: primary hemorrhage and membrane injury theory of acute spinal cord trauma with the emergency department. Resuscitation 76, 57–62
permeability in distinct mechanisms of spinal cord emphasis on vascular mechanisms. J. Neurosurg. 75, (2008).
injury. J. Neurosurg. Spine 6, 255–266 (2007). 15–26 (1991). 68. Acheson, M., Livingston, R., Richardson, M.
17. Pineau, I. & Lacroix, S. Proinflammatory cytokine 42. Kotter, M. R., Li, W. W., Zhao, C. & Franklin, R. J. & Stimac, G. High-resolution CT scanning in the
synthesis in the injured mouse spinal cord: multiphasic Myelin impairs CNS remyelination by inhibiting evaluation of cervical spine fractures: comparison
expression pattern and identification of the cell types oligodendrocyte precursor cell differentiation. with plain film examinations. AJR Am. J. Roentgenol.
involved. J. Comp. Neurol. 500, 267–285 (2007). J. Neurosci. 26, 328–332 (2006). 148, 1179–1185 (1987).
18. Schanne, F. A., Kane, A. B., Young, E. E. & Farber, J. L. 43. Syed, Y. A. et al. Antibody-mediated neutralization 69. Woodring, J. & Lee, C. Limitations of cervical
Calcium dependence of toxic cell death: a final of myelin-associated EphrinB3 accelerates CNS radiography in the evaluation of acute cervical trauma.
common pathway. Science 206, 700–702 (1979). remyelination. Acta Neuropathol. 131, 281–298 J. Trauma 34, 32–39 (1993).
19. Kwon, B. Pathophysiology and pharmacologic (2016). 70. Vaccaro, A. R. et al. AOSpine subaxial cervical
treatment of acute spinal cord injury. Spine J. 4, 44. Kotter, M. R., Setzu, A., Sim, F. J., Van Rooijen, N. spine injury classification system. Eur. Spine J. 25,
451–464 (2004). & Franklin, R. J. Macrophage depletion impairs 2173–2184 (2016).
20. Dizdaroglu, M., Jaruga, P., Birincioglu, M. oligodendrocyte remyelination following lysolecithin- 71. Vaccaro, A. R. et al. AOSpine thoracolumbar spine
& Rodriguez, H. Free radical-induced damage to DNA: induced demyelination. Glia 35, 204–212 (2001). injury classification system: fracture description,
mechanisms and measurement. Free Radic. Biol. Med. 45. Bieber, A. J., Warrington, A., Pease, L. R. & neurological status, and key modifiers. Spine 38,
32, 1102–1115 (2002). Rodriguez, M. Humoral autoimmunity as a mediator 2028–2037 (2013).
21. Hausmann, O. N. Post-traumatic inflammation of CNS repair. Trends Neurosci. 24, S39–S44 (2001). 72. Lammertse, D. et al. Neuroimaging in traumatic spinal
following spinal cord injury. Spinal Cord 41, 369–378 46. Jeffery, N. D. & Blakemore, W. F. Locomotor deficits cord injury: an evidence-based review for clinical
(2003). induced by experimental spinal cord demyelination practice and research. J. Spinal Cord Med. 30,
22. Liu, M. et al. Necroptosis, a novel type of programmed are abolished by spontaneous remyelination. Brain 205–214 (2007).
cell death, contributes to early neural cells damage 120, 27–37 (1997). 73. Miyanji, F., Furlan, J., Aarabi, B., Arnold, P.
after spinal cord injury in adult mice. J. Spinal Cord 47. Lynskey, J. V., Belanger, A. & Jung, R. Activity- & Fehlings, M. Acute cervical traumatic spinal cord
Med. 38, 745–753 (2015). dependent plasticity in spinal cord injury. J. Rehabil. injury: MR imaging findings correlated with neurologic
23. Wang, Y. et al. Necroptosis inhibitor necrostatin‑1 Res. Dev. 45, 229–240 (2008). outcome — prospective study with 100 consecutive
promotes cell protection and physiological function 48. Raineteau, O. & Schwab, M. E. Plasticity of motor patients. Radiology 243, 820–827 (2007).
in traumatic spinal cord injury. Neuroscience 266, systems after incomplete spinal cord injury. Nat. Rev. 74. Vaccaro, A. R. et al. Magnetic resonance evaluation
91–101 (2014). Neurosci. 2, 263–273 (2001). of the intervertebral disc, spinal ligaments, and spinal
24. Li, S., Mealing, G. A., Morley, P. & Stys, P. K. Novel 49. Meletis, K. et al. Spinal cord injury reveals cord before and after closed traction reduction of
injury mechanism in anoxia and trauma of spinal multilineage differentiation of ependymal cells. cervical spine dislocations. Spine 24, 1210–1217
cord white matter: glutamate release via reverse PLoS Biol. 6, e182 (2008). (1999).

18 | ARTICLE NUMBER 17018 | VOLUME 3 www.nature.com/nrdp


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
PRIMER

75. Nakashima, H. et al. Posterior approach for cervical 97. Pavese, C. et al. Prediction of bladder outcomes after 119. Brodbelt, A. R. & Stoodley, M. A. Post-traumatic
fracture-dislocations with traumatic disc herniation. traumatic spinal cord injury: a longitudinal cohort syringomyelia: a review. J. Clin. Neurosci. 10, 401–408
Eur. Spine J. 20, 387–394 (2011). study. PLoS Med. 13, e1002041 (2016). (2003).
76. Resnick, D. K. Updated guidelines for the 98. Ryken, T. C. et al. The acute cardiopulmonary 120. Schurch, B., Wichmann, W. & Rossier, A. B.
management of acute cervical spine and spinal cord management of patients with cervical spinal cord Post‑traumatic syringomyelia (cystic myelopathy):
injury. Neurosurgery 72 (Suppl. 3), 1 (2013). injuries. Neurosurgery 72 (Suppl. 2), 84–92 (2013). a prospective study of 449 patients with spinal cord
Widely accepted guidelines by the AANS/CNS 99. Braughler, J. & Hall, E. Effects of multi-dose injury. J. Neurol. Neurosurg. Psychiatry 60, 61–67
for the management of acute cervical SCIs. methylprednisolone sodium succinate administration (1996).
77. Cadotte, D. W. & Fehlings, M. G. Will imaging on injured cat spinal cord neurofilament degradation 121. Alpert, S. W., Koval, K. J. & Zuckerman, J. D.
biomarkers transform spinal cord injury trials? and energy metabolism. J. Neurosurg. 61, 290–295 Neuropathic arthropathy: review of current knowledge.
Lancet Neurol. 12, 843–844 (2013). (1984). J. Am. Acad. Orthop. Surg. 4, 100–108 (1996).
78. Curt, A. & Dietz, V. Electrophysiological recordings 100. Hall, E. D. & Braughler, J. M. Glucocorticoid 122. Aebli, N., Potzel, T. & Krebs, J. Characteristics and
in patients with spinal cord injury: significance for mechanisms in acute spinal cord injury: a review and surgical management of neuropathic (Charcot) spinal
predicting outcome. Spinal Cord 37, 157–165 therapeutic rationale. Surg. Neurol. 18, 320–327 arthropathy after spinal cord injury. Spine J. 14,
(1999). (1982). 884–891 (2014).
79. Curt, A., Van Hedel, H. J., Klaus, D., Dietz, V. 101. Hall, E. D. & Braughler, J. M. Effects of intravenous 123. Adams, M. M. & Hicks, A. L. Spasticity after spinal
& Group, E.‑S. S. Recovery from a spinal cord injury: methylprednisolone on spinal cord lipid peroxidation cord injury. Spinal Cord 43, 577–586 (2005).
significance of compensation, neural plasticity, and Na+ + K+)-ATPase activity. Dose–response 124. Sezer, N., Akkus, S. & Ugurlu, F. G. Chronic
and repair. J. Neurotrauma 25, 677–685 (2008). analysis during 1st hour after contusion injury complications of spinal cord injury. World J. Orthop. 6,
80. Kirshblum, S. C. et al. Reference for the 2011 revision in the cat. J. Neurosurg. 57, 247–253 (1982). 24–33 (2015).
of the International Standards for Neurological 102. Bracken, M. et al. Administration of 125. Claydon, V. E., Steeves, J. D. & Krassioukov, A.
Classification of Spinal Cord Injury. J. Spinal Cord methylprednisolone for 24 or 48 hours or tirilazad Orthostatic hypotension following spinal cord injury:
Med. 34, 547–554 (2011). mesylate for 48 hours in the treatment of acute spinal understanding clinical pathophysiology. Spinal Cord 44,
The updated ISNCSCI for the assessment and cord injury. Results of the Third National Acute Spinal 341–351 (2006).
reassessment of patients with traumatic SCI. Cord Injury Randomized Controlled Trial. National 126. Krassioukov, A., Eng, J. J., Warburton, D. E., Teasell, R.
81. American Spinal Injury Association. ISNCSCI worksheet. Acute Spinal Cord Injury Study. JAMA 277, & Spinal Cord Injury Rehabilitation Evidence Research
ASIA http://asia-spinalinjury.org/information/downloads/ 1597–1604 (1997). Team. A systematic review of the management of
(2000). 103. Bracken, M. B. et al. A randomized, controlled trial orthostatic hypotension after spinal cord injury.
82. Marino, R. J., Jones, L., Kirshblum, S., Tal, J. of methylprednisolone or naloxone in the treatment Arch. Phys. Med. Rehabil. 90, 876–885 (2009).
& Dasgupta, A. Reliability and repeatability of the of acute spinal-cord injury. Results of the Second 127. Krassioukov, A., Warburton, D. E., Teasell, R.
motor and sensory examination of the international National Acute Spinal Cord Injury Study. N. Engl. & Eng, J. J. A systematic review of the management
standards for neurological classification of spinal cord J. Med. 322, 1405–1411 (1990). of autonomic dysreflexia after spinal cord injury.
injury. J. Spinal Cord Med. 31, 166–170 (2008). 104. Bracken, M. B. et al. Efficacy of methylprednisolone in Arch. Phys. Med. Rehabil. 90, 682–695 (2009).
83. Savic, G., Bergstrom, E. M., Frankel, H. L., acute spinal cord injury. JAMA 251, 45–52 (1984). 128. Blackmer, J. Rehabilitation medicine: 1. Autonomic
Jamous, M. A. & Jones, P. W. Inter-rater reliability of 105. Bracken, M. B. Steroids for acute spinal cord injury. dysreflexia. CMAJ 169, 931–935 (2003).
motor and sensory examinations performed according Cochrane Database Syst. Rev. 18, CD001046 129. Linn, W. S., Adkins, R. H., Gong, H. Jr & Waters, R. L.
to American Spinal Injury Association standards. (2012). Pulmonary function in chronic spinal cord injury:
Spinal Cord 45, 444–451 (2007). A systematic review and meta-analysis of the a cross-sectional survey of 222 southern California
84. Sherwood, A. M., Dimitrijevic, M. R. & McKay, W. B. best-available randomized trials that assess steroid adult outpatients. Arch. Phys. Med. Rehabil. 81,
Evidence of subclinical brain influence in clinically therapy for acute traumatic SCI. 757–763 (2000).
complete spinal cord injury: discomplete SCI. 106. Eck, J. C., Nachtigall, D., Humphreys, S. C. 130. Brown, R., DiMarco, A. F., Hoit, J. D. & Garshick, E.
J. Neurol. Sci. 110, 90–98 (1992). & Hodges, S. D. Questionnaire survey of spine Respiratory dysfunction and management in spinal
85. Schneider, R. C., Cherry, G. & Pantek, H. The surgeons on the use of methylprednisolone for acute cord injury. Respir. Care 51, 853–868 (2006).
syndrome of acute central cervical spinal cord injury; spinal cord injury. Spine 31, E250–E253 (2006). 131. Winslow, C. & Rozovsky, J. Effect of spinal cord injury
with special reference to the mechanisms involved in 107. Hurlbert, R. Methylprednisolone for acute spinal on the respiratory system. Am. J. Phys. Med. Rehabil.
hyperextension injuries of cervical spine. J. Neurosurg. cord injury: an inappropriate standard of care. 82, 803–814 (2003).
11, 546–577 (1954). J. Neurosurg. Spine 93, 1–7 (2000). 132. DeVivo, M. J., Black, K. J. & Stover, S. L. Causes of
86. Fehlings, M. G. et al. The aging of the global 108. Hurlbert, R. J. et al. Pharmacological therapy for death during the first 12 years after spinal cord injury.
population: the changing epidemiology of disease and acute spinal cord injury. Neurosurgery 72 (Suppl. 2), Arch. Phys. Med. Rehabil. 74, 248–254 (1993).
spinal disorders. Neurosurgery 77, S1–S5 (2015). 93–105 (2013). 133. Brommer, B. et al. Spinal cord injury-induced immune
87. McKinley, W., Santos, K., Meade, M. & Brooke, K. 109. Hadley, M. et al. Pharmacological therapy after deficiency syndrome enhances infection susceptibility
Incidence and outcomes of spinal cord injury clinical acute cervical spinal cord injury. Neurosurgery 50, dependent on lesion level. Brain 139, 692–707
syndromes. J. Spinal Cord Med. 30, 215–224 (2007). 563–572 (2002). (2016).
88. Burns, A. & Ditunno, J. Establishing prognosis and 110. Fehlings, M. G., Wilson, J. R. & Cho, N. 134. Ulndreaj, A., Chio, J. C., Ahuja, C. S. & Fehlings, M. G.
maximizing functional outcomes after spinal cord Methylprednisolone for the treatment of acute Modulating the immune response in spinal cord injury.
injury. Spine 26, S137–S145 (2001). spinal cord injury: counterpoint. Neurosurgery Expert Rev. Neurother. 16, 1127–1129 (2016).
89. Kirshblum, S., Millis, S., McKinley, W. & Tulsky, D. Late 61 (Suppl. 1), 36–42 (2014). 135. Benevento, B. T. & Sipski, M. L. Neurogenic bladder,
neurologic recovery after traumatic spinal cord injury. 111. Hurlbert, R. J. Methylprednisolone for the treatment neurogenic bowel, and sexual dysfunction in people
Arch. Phys. Med. Rehabil. 85, 1811–1817 (2004). of acute spinal cord injury: point. Neurosurgery 61 with spinal cord injury. Phys. Ther. 82, 601–612
90. Fawcett, J. et al. Guidelines for the conduct of clinical (Suppl. 1), 32–35 (2014). (2002).
trials for spinal cord injury as developed by the ICCP 112. Carlson, G. D. et al. Early time-dependent 136. Taweel, W. A. & Seyam, R. Neurogenic bladder in spinal
panel: spontaneous recovery after spinal cord injury decompression for spinal cord injury: vascular cord injury patients. Res. Rep. Urol. 7, 85–99 (2015).
and statistical power needed for therapeutic trials. mechanisms of recovery. J. Neurotrauma 14, 137. Hess, M. J. & Hough, S. Impact of spinal cord injury
Spinal Cord 45, 190–205 (2007). 951–962 (1997). on sexuality: broad-based clinical practice intervention
91. Marino, R., Dittuno, J., Donovan, W. & Maynard, F. 113. Dimar, J., Glassman, S., Raque, G., Zhang, Y. and practical application. J. Spinal Cord Med. 35,
Neurologic recovery after traumatic spinal cord injury: & Shields, C. The influence of spinal canal narrowing 211–218 (2012).
data from the model spinal cord injury systems. and timing of decompression on neurologic recovery 138. Coggrave, M. J. & Norton, C. The need for manual
Arch. Phys. Med. Rehabil. 80, 1391–1396 (1999). after spinal cord contusion in a rat model. Spine 24, evacuation and oral laxatives in the management of
92. Waters, R., Yakura, J., Adkins, R. & Sie, I. Recovery 1623–1633 (1999). neurogenic bowel dysfunction after spinal cord injury:
following complete paraplegia. Arch. Phys. Med. 114. Batchelor, P. E. et al. Meta-analysis of pre-clinical a randomized controlled trial of a stepwise protocol.
Rehabil. 73, 784–789 (1992). studies of early decompression in acute spinal cord Spinal Cord 48, 504–510 (2010).
93. Coleman, W. & Geisler, F. Injury severity as primary injury: a battle of time and pressure. PLoS ONE 8, 139. Krassioukov, A., Eng, J. J., Claxton, G.,
predictor of outcome in acute spinal cord injury: e72659 (2013). Sakakibara, B. M. & Shum, S. Neurogenic bowel
retrospective results from a large multicenter clinical 115. Wilson, J. R. et al. Early versus late surgery for management after spinal cord injury: a systematic
trial. Spine J. 4, 373–378 (2004). traumatic spinal cord injury: the results of a review of the evidence. Spinal Cord 48, 718–733
94. Kay, E., Deutsch, A. & Wuermser, L. Predicting walking prospective Canadian cohort study. Spinal Cord 50, (2010).
at discharge from inpatient rehabilitation after a 840–843 (2012). 140. Consortium for Spinal Cord Medicine Clinical Practice
traumatic spinal cord injury. Arch. Phys. Med. Rehabil. 116. Bourassa-Moreau, E., Mac-Thiong, J. M., Guidelines. Pressure ulcer prevention and treatment
88, 745–750 (2007). Feldman, D. E., Thompson, C. & Parent, S. following spinal cord injury: a clinical practice guideline
95. van Middendorp, J. J. et al. A clinical prediction rule Non‑neurological outcomes after complete traumatic for health-care professionals. J. Spinal Cord Med. 24,
for ambulation outcomes after traumatic spinal cord spinal cord injury: the impact of surgical timing. S40–S101 (2001).
injury: a longitudinal cohort study. Lancet 377, J. Neurotrauma 30, 1596–1601 (2013). 141. van Kuijk, A. A., Geurts, A. C. & van Kuppevelt, H. J.
1004–1010 (2011). 117. Grassner, L. et al. Early decompression (<8 h) Neurogenic heterotopic ossification in spinal cord injury.
A clinical prediction rule for ambulation at 1 year after traumatic cervical spinal cord injury improves Spinal Cord 40, 313–326 (2002).
post-SCI based on age, ASIA motor scores and functional outcome as assessed by spinal cord 142. Rush, P. J. The rheumatic manifestations of traumatic
sensory function in the subacute (within 15 days) independence measure after one year. J. Neurotrauma spinal cord injury. Semin. Arthritis Rheum. 19, 77–89
injury period. 33, 1658–1666 (2016). (1989).
96. Wilson, J. R. et al. A clinical prediction model for long- 118. Fehlings, M., Rabin, D., Sears, W., Cadotte, D. 143. Cardenas, D. D. & Felix, E. R. Pain after spinal
term functional outcome after traumatic spinal cord & Aarabi, B. Current practice in the timing of surgical cord injury: a review of classification, treatment
injury based on acute clinical and imaging factors. intervention in spinal cord injury. Spine 35, 166–173 approaches, and treatment assessment. PM R 1,
J. Neurotrauma 29, 2263–2271 (2012). (2010). 1077–1090 (2009).

NATURE REVIEWS | DISEASE PRIMERS VOLUME 3 | ARTICLE NUMBER 17018 | 19


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
PRIMER

144. Gomara-Toldra, N., Sliwinski, M. & Dijkers, M. P. 167. Bracken, M. B. et al. Administration of injury: a systematic review. J. Neurosurg. Spine 17,
Physical therapy after spinal cord injury: a systematic methylprednisolone for 24 or 48 hours or tirilazad 230–246 (2012).
review of treatments focused on participation. J. Spinal mesylate for 48 hours in the treatment of acute spinal 189. Tetzlaff, W. et al. A systematic review of cellular
Cord Med. 37, 371–379 (2014). cord injury. Results of the Third National Acute Spinal transplantation therapies for spinal cord injury.
145. Hwang, D. H. et al. Survival of neural stem cell grafts in Cord Injury Randomized Controlled Trial. National J. Neurotrauma 28, 1611–1682 (2011).
the lesioned spinal cord is enhanced by a combination Acute Spinal Cord Injury Study. JAMA 277, 190. Khazaei, M., Ahuja, C. S. & Fehlings, M. G. Induced
of treadmill locomotor training via insulin-like growth 1597–1604 (1997). pluripotent stem cells for traumatic spinal cord injury.
factor‑1 signaling. J. Neurosci. 34, 12788–12800 168. Kwon, B. K. et al. Cerebrospinal fluid inflammatory Front. Cell Dev. Biol. 4, 152 (2017).
(2014). cytokines and biomarkers of injury severity in acute 191. Ramon-Cueto, A., Cordero, M. I., Santos-Benito, F. F. &
146. Dobkin, B. et al. Weight-supported treadmill versus human spinal cord injury. J. Neurotrauma 27, Avila, J. Functional recovery of paraplegic rats and motor
over-ground training for walking after acute incomplete 669–682 (2010). axon regeneration in their spinal cords by olfactory
SCI. Neurology 66, 484–493 (2006). 169. Siddiqui, A. M., Khazaei, M. & Fehlings, M. G. ensheathing glia. Neuron 25, 425–435 (2000).
147. Stiens, S. A., Kirshblum, S. C., Groah, S. L., Translating mechanisms of neuroprotection, 192. Keyvan-Fouladi, N., Raisman, G. & Li, Y. Functional
McKinley, W. O. & Gittler, M. S. Spinal cord injury regeneration, and repair to treatment of spinal cord repair of the corticospinal tract by delayed
medicine. 4. Optimal participation in life after spinal injury. Prog. Brain Res. 218, 15–54 (2015). transplantation of olfactory ensheathing cells in adult
cord injury: physical, psychosocial, and economic 170. Grossman, R. G., Toups, E. G., Frankowski, R. F., rats. J. Neurosci. 23, 9428–9434 (2003).
reintegration into the environment. Arch. Phys. Med. Burau, K. D. & Howley, S. North American Clinical 193. Pascual, J. I., Gudino-Cabrera, G., Insausti, R.
Rehabil. 83, S72–S81 (2002). Trials Network for the Treatment of Spinal Cord Injury: & Nieto-Sampedro, M. Spinal implants of olfactory
148. Ho, C. H. et al. Functional electrical stimulation and goals and progress. J. Neurosurg. Spine 17 (1 Suppl.), ensheathing cells promote axon regeneration and
spinal cord injury. Phys. Med. Rehabil. Clin. N. Am. 25, 6–10 (2012). bladder activity after bilateral lumbosacral dorsal
631–654 (2014). 171. DeVivo, M. et al. International spinal cord injury core rhizotomy in the adult rat. J. Urol. 167, 1522–1526
149. Bhambhani, Y., Tuchak, C., Burnham, R., Jeon, J. data set. Spinal Cord 44, 535–540 (2006). (2002).
& Maikala, R. Quadriceps muscle deoxygenation 172. DeVivo, M. J., Go, B. K. & Jackson, A. B. Overview 194. Li, Y., Decherchi, P. & Raisman, G. Transplantation of
during functional electrical stimulation in adults with of the national spinal cord injury statistical center olfactory ensheathing cells into spinal cord lesions
spinal cord injury. Spinal Cord 38, 630–638 (2000). database. J. Spinal Cord Med. 25, 335–338 (2002). restores breathing and climbing. J. Neurosci. 23,
150. Kakebeeke, T. H. et al. Training and detraining of a 173. Ahuja, C. S. & Fehlings, M. Concise review: bridging the 727–731 (2003).
tetraplegic subject: high-volume FES cycle training. gap: novel neuroregenerative and neuroprotective 195. Boyd, J. G., Lee, J., Skihar, V., Doucette, R.
Am. J. Phys. Med. Rehabil. 87, 56–64 (2008). strategies in spinal cord injury. Stem Cells Transl Med. & Kawaja, M. D. LacZ-expressing olfactory ensheathing
151. Ragnarsson, K. T. Functional electrical stimulation after 5, 914–924 (2016). cells do not associate with myelinated axons after
spinal cord injury: current use, therapeutic effects and A review of key neuroregenerative and implantation into the compressed spinal cord.
future directions. Spinal Cord 46, 255–274 (2008). neuroprotective interventions for traumatic SCI Proc. Natl Acad. Sci. USA 101, 2162–2166 (2004).
152. Tate, D. & Forchheimer, M. Review of cross-cultural along the translational pipeline. 196. Li, J. & Lepski, G. Cell transplantation for spinal cord
issues related to quality of life after spinal cord injury. 174. Wells, J., Hurlbert, R., Fehlings, M. & Yong, V. injury: a systematic review. Biomed. Res. Int. 2013,
Top. Spinal Cord Inj. Rehabil. 20, 181–190 (2014). Neuroprotection by minocycline facilitates significant 786475 (2013).
153. Wilson, J. R., Hashimoto, R. E., Dettori, J. R. recovery from spinal cord injury in mice. Brain 126, 197. Pastrana, E. et al. Genes associated with adult axon
& Fehlings, M. G. Spinal cord injury and quality of life: 1628–1637 (2003). regeneration promoted by olfactory ensheathing cells:
a systematic review of outcome measures. Evid. Based 175. Lee, S. M. et al. Minocycline reduces cell death and a new role for matrix metalloproteinase 2. J. Neurosci.
Spine Care J. 2, 37–44 (2011). improves functional recovery after traumatic spinal 26, 5347–5359 (2006).
154. Tulsky, D. S. et al. Overview of the Spinal Cord cord injury in the rat. J. Neurotrauma 20, 1017–1027 198. Guo, J. S. et al. Cotransplant of neural stem cells
Injury–Quality of Life (SCI-QOL) measurement system. (2003). and NT‑3 gene modified Schwann cells promote the
J. Spinal Cord Med. 38, 257–269 (2015). 176. Wilson, J. R. & Fehlings, M. G. Riluzole for acute recovery of transected spinal cord injury. Spinal Cord
155. Dijkers, M. Quality of life after spinal cord injury: traumatic spinal cord injury: a promising 45, 15–24 (2007).
a meta analysis of the effects of disablement neuroprotective treatment strategy. World Neurosurg. 199. Mackay-Sim, A. et al. Autologous olfactory ensheathing
components. Spinal Cord 35, 829–840 (1997). 81, 825–829 (2013). cell transplantation in human paraplegia: a 3‑year
A meta-analysis of studies that assesses the 177. Grossman, R. G. et al. A prospective, multicenter, clinical trial. Brain 131, 2376–2386 (2008).
relationship between QOL and disability, phase I matched-comparison group trial of safety, 200. Dlouhy, B. J., Awe, O., Rao, R. C., Kirby, P. A.
impairment and handicap. pharmacokinetics, and preliminary efficacy of riluzole & Hitchon, P. W. Autograft-derived spinal cord mass
156. Clayton, K. S. & Chubon, R. A. Factors associated in patients with traumatic spinal cord injury. following olfactory mucosal cell transplantation in a
with the quality of life of long-term spinal cord injured J. Neurotrauma 31, 239–255 (2014). spinal cord injury patient: case report. J. Neurosurg.
persons. Arch. Phys. Med. Rehabil. 75, 633–638 178. Santa-Olalla, J. & Covarrubias, L. Basic fibroblast Spine 21, 618–622 (2014).
(1994). growth factor promotes epidermal growth factor 201. Chhabra, H. S. et al. Autologous olfactory [corrected]
157. Evans, R. L. et al. Quality of life after spinal cord injury: responsiveness and survival of mesencephalic neural mucosal transplant in chronic spinal cord injury:
a literature critique and meta-analysis (1983–1992). precursor cells. J. Neurobiol. 40, 14–27 (1999). an Indian pilot study. Spinal Cord 47, 887–895 (2009).
J. Am. Paraplegia Soc. 17, 60–66 (1994). 179. Teng, Y. D., Mocchetti, I., Taveira-DaSilva, A. M., 202. Tabakow, P. et al. Transplantation of autologous
158. Fuhrer, M. J., Rintala, D. H., Hart, K. A., Clearman, R. Gillis, R. A. & Wrathall, J. R. Basic fibroblast growth olfactory ensheathing cells in complete human spinal
& Young, M. E. Relationship of life satisfaction to factor increases long-term survival of spinal motor cord injury. Cell Transplant. 22, 1591–1612 (2013).
impairment, disability, and handicap among persons neurons and improves respiratory function after 203. Hill, C. E., Moon, L. D., Wood, P. M. & Bunge, M. B.
with spinal cord injury living in the community. experimental spinal cord injury. J. Neurosci. 19, Labeled Schwann cell transplantation: cell loss, host
Arch. Phys. Med. Rehabil. 73, 552–557 (1992). 7037–7047 (1999). Schwann cell replacement, and strategies to enhance
159. Gerhart, K. A., Koziol-McLain, J., Lowenstein, S. R. 180. Rabchevsky, A. G. et al. Basic fibroblast growth factor survival. Glia 53, 338–343 (2006).
& Whiteneck, G. G. Quality of life following spinal cord (bFGF) enhances functional recovery following severe 204. Li, L. et al. Effects of transplantation of olfactory
injury: knowledge and attitudes of emergency care spinal cord injury to the rat. Exp. Neurol. 164, ensheathing cells in chronic spinal cord injury:
providers. Ann. Emerg. Med. 23, 807–812 (1994). 280–291 (2000). a systematic review and meta-analysis. Eur. Spine J. 24,
160. Krause, J. S. & Crewe, N. M. Chronologic age, time 181. Batchelor, P. E. et al. Hypothermia prior to 919–930 (2015).
since injury, and time of measurement: effect on decompression: buying time for treatment of acute A meta-analysis that pools data from 1,193
adjustment after spinal cord injury. Arch. Phys. Med. spinal cord injury. J. Neurotrauma 27, 1357–1368 patients receiving OEC transplants for chronic SCI.
Rehabil. 72, 91–100 (1991). (2010). 205. Zhu, H. et al. Phase I–II clinical trial assessing safety
161. Jain, N. B., Sullivan, M., Kazis, L. E., Tun, C. G. 182. Levi, A. D. et al. Clinical outcomes using modest and efficacy of umbilical cord blood mononuclear cell
& Garshick, E. Factors associated with health-related intravascular hypothermia after acute cervical spinal transplant therapy of chronic complete spinal cord
quality of life in chronic spinal cord injury. Am. J. Phys. cord injury. Neurosurgery 66, 670–677 (2010). injury. Cell Transplant. 25, 1925–1943 (2016).
Med. Rehabil. 86, 387–396 (2007). 183. Dergham, P., Ellezam, B. & Essagian, C. Rho signalling 206. Harkema, S. et al. Effect of epidural stimulation of the
A cross-sectional study that investigates the pathway targeted to promote spinal cord repair. lumbosacral spinal cord on voluntary movement,
health-related QOL instrument and identifies J. Neurosci. 22, 6570–6577 (2002). standing, and assisted stepping after motor complete
potentially modifiable factors related to QOL in SCI. 184. Kwon, B. K., Sekhon, L. H. & Fehlings, M. G. Emerging paraplegia: a case study. Lancet 377, 1938–1947
162. Brillhart, B. & Johnson, K. Motivation and the coping repair, regeneration, and translational research advances (2011).
process of adults with disabilities: a qualitative study. for spinal cord injury. Spine 35, S263–S270 (2010). 207. Bachmann, L. C. et al. Deep brain stimulation of the
Rehabil. Nurs. 22, 249–256 (1997). 185. Fehlings, M. G. et al. A phase I/IIa clinical trial of a midbrain locomotor region improves paretic hindlimb
163. Siosteen, A., Lundqvist, C., Blomstrand, C., Sullivan, L. recombinant Rho protein antagonist in acute spinal cord function after spinal cord injury in rats. Sci. Transl Med.
& Sullivan, M. The quality of life of three functional injury. J. Neurotrauma 28, 787–796 (2011). 5, 208ra146 (2013).
spinal cord injury subgroups in a Swedish community. 186. Freund, P. et al. Anti-Nogo‑A antibody treatment 208. Bouton, C. E. et al. Restoring cortical control of
Paraplegia 28, 476–488 (1990). enhances sprouting of corticospinal axons rostral to a functional movement in a human with quadriplegia.
164. DeVivo, M. J. & Richards, J. S. Community unilateral cervical spinal cord lesion in adult macaque Nature 533, 247–250 (2016).
reintegration and quality of life following spinal cord monkey. J. Comp. Neurol. 502, 644–659 (2007). 209. Zeilig, G. et al. Safety and tolerance of the ReWalk
injury. Paraplegia 30, 108–112 (1992). 187. Zorner, B. & Schwab, M. E. Anti-Nogo on the go: exoskeleton suit for ambulation by people with
165. Saadat, S. et al. Health-related quality of life among from animal models to a clinical trial. Ann. NY Acad. complete spinal cord injury: a pilot study. J. Spinal Cord
individuals with long-standing spinal cord injury: Sci. 1198 (Suppl. 1), E22–E34 (2010). Med. 35, 96–101 (2012).
a comparative study of veterans and non-veterans. A review of anti-Nogo therapy, which provides an 210. Miller, L. E., Zimmermann, A. K. & Herbert, W. G.
BMC Public Health 10, 6 (2010). example of the bench-to-bedside path for novel Clinical effectiveness and safety of powered
166. Westgren, N. & Levi, R. Quality of life and traumatic therapeutics. exoskeleton-assisted walking in patients with spinal
spinal cord injury. Arch. Phys. Med. Rehabil. 79, 188. Harrop, J. S. et al. Evaluation of clinical experience cord injury: systematic review with meta-analysis.
1433–1439 (1998). using cell-based therapies in patients with spinal cord Med. Devices (Auckl.) 9, 455–466 (2016).

20 | ARTICLE NUMBER 17018 | VOLUME 3 www.nature.com/nrdp


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
PRIMER

211. Akhtar, A. Z., Pippin, J. J. & Sandusky, C. B. quadriplegia. Arch. Phys. Med. Rehabil. 76, 635–641 Author contributions
Animal models in spinal cord injury: a review. (1995). Introduction (M.G.F., C.S.A., J.R.W., S.N., M.R.N.K. and
Rev. Neurosci. 19, 47–60 (2008). 217. Ogura, T. et al. Sympathetic skin response in patients C.D.); Epidemiology (S.N., C.S.A., J.R.W., M.R.N.K. and
212. Basso, D. M. Behavioral testing after spinal cord with spinal cord injury. J. Orthop. Surg. (Hong Kong) C.D.); Mechanisms/pathophysiology (M.G.F., C.S.A., J.R.W.
injury: congruities, complexities, and controversies. 12, 35–39 (2004). and S.N.); Diagnosis, screening and prevention (C.S.A.,
J. Neurotrauma 21, 395–404 (2004). 218. Bradbury, E. J. & McMahon, S. B. Spinal cord repair J.R.W., S.N. and A.C.); Management (M.G.F., C.S.A.,
213. Kwon, B. K., Oxland, T. R. & Tetzlaff, W. Animal strategies: why do they work? Nat. Rev. Neurosci. 7, J.R.W., M.R.N.K., C.D. and A.C.); Quality of life (S.N.,
models used in spinal cord regeneration research. 644–653 (2006). C.S.A., J.R.W., M.R.N.K. and C.D.); Outlook (C.S.A., J.R.W.,
Spine 27, 1504–1510 (2002). 219. Takahashi, H. et al. Neuroprotective therapy using S.N., M.R.N.K. and A.C.); Overview of Primer (M.G.F.).
An overview of pertinent animal models granulocyte colony-stimulating factor for acute spinal Co‑first authors C.S.A. and J.R.W. contributed equally to
and injury paradigms to study spinal cord cord injury: a phase I/IIa clinical trial. Eur. Spine J. 21, this work.
regeneration. 2580–2587 (2012).
214. Calancie, B., Molano, M. R. & Broton, J. G. EMG for 220. Kamiya, K. et al. Neuroprotective therapy with Competing interests
assessing the recovery of voluntary movement after granulocyte colony-stimulating factor in acute M.G.F. has consulting agreements with Pfizer, Zimmer
acute spinal cord injury in man. Clin. Neurophysiol. spinal cord injury: a comparison with high-dose Biomed and InVivo therapeutics. All other authors declare
115, 1748–1759 (2004). methylprednisolone as a historical control. Eur. Spine J. no competing interests.
215. Curt, A. & Dietz, V. Nerve conduction study 24, 963–967 (2015).
in cervical spinal cord injury: significance for 221. Lo, T. P. Jr et al. Systemic hypothermia improves Publisher’s note
hand function. NeuroRehabilitation 7, 165–173 histological and functional outcome after cervical Springer Nature remains neutral with regard to jurisdictional
(1996). spinal cord contusion in rats. J. Comp. Neurol. 514, claims in published maps and institutional affiliations.
216. Jacobs, S. R., Yeaney, N. K., Herbison, G. J. 433–448 (2009).
& Ditunno, J. F. Jr. Future ambulation prognosis 222. Levi, A. D. et al. Clinical application of modest How to cite this article
as predicted by somatosensory evoked hypothermia after spinal cord injury. J. Neurotrauma Fehlings, M. G. et al. Traumatic spinal cord injury. Nat. Rev.
potentials in motor complete and incomplete 26, 407–415 (2009). Dis. Primers 3, 17018 (2017).

NATURE REVIEWS | DISEASE PRIMERS VOLUME 3 | ARTICLE NUMBER 17018 | 21


©
2
0
1
7
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.

You might also like