You are on page 1of 73

J Neurosurg Spine (Suppl) 17:157229, 2012

Translational potential of preclinical trials of neuroprotection


through pharmacotherapy for spinal cord injury
Charles H. Tator, M.D., Ph.D.,1 Robin Hashimoto, Ph.D., 2 Annie Raich, M.P.H., 2
Daniel Norvell, Ph.D., 2 Michael G. Fehlings, M.D., Ph.D.,1 James S. Harrop, M.D., 3
James Guest, M.D., Ph.D., 4 Bizhan Aarabi, M.D., F.R.C.S.C., 5
and Robert G. Grossman, M.D. 6
Division of Neurosurgery and Spinal Program, Toronto Western Hospital and University of Toronto, Ontario,
Canada; 2Spectrum Research, Inc., Tacoma, Washington; 3Department of Neurological Surgery, Thomas
Jefferson University, Philadelphia, Pennsylvania; 4Department of Neurological Surgery and the Miami
Project to Cure Paralysis, Miller School of Medicine, University of Miami, Florida; 5Department of
Neurosurgery, University of Maryland School of Medicine, Baltimore, Maryland; and 6Department of
Neurosurgery, The Methodist Hospital, Houston, Texas

There is a need to enhance the pipeline of discovery and evaluation of neuroprotective pharmacological agents
for patients with spinal cord injury (SCI). Although much effort and money has been expended on discovering effective agents for acute and subacute SCI, no agents that produce major benefit have been proven to date. The deficiencies of all aspects of the pipeline, including the basic science input and the clinical testing output, require examination
to determine remedial strategies. Where has the neuroprotective/pharmacotherapy preclinical process failed and what
needs to be done to achieve success? These are the questions raised in the present review, which has 2 objectives:
1) identification of articles that address issues related to the translational readiness of preclinical SCI pharmacological therapies; and 2) examination of the preclinical studies of 5 selected agents evaluated in animal models of SCI
(including blunt force trauma, penetrating trauma, or ischemia). The 5 agents were riluzole, glyburide, magnesium
sulfate, nimodipine, and minocycline, and these were selected because of their promise of translational readiness as
determined by the North American Clinical Trials Network Consortium.
The authors found that there are major deficiencies in the effort that has been extended to coordinate and conduct
preclinical neuroprotection/pharmacotherapy trials in the SCI field. Apart from a few notable exceptions such as the
NIH effort to replicate promising strategies, this field has been poorly coordinated. Only a small number of articles
have even attempted an overall evaluation of the neuroprotective/pharmacotherapy agents used in preclinical SCI
trials. There is no consensus about how to select the agents for translation to humans on the basis of their preclinical
performance and according to agreed-upon preclinical performance criteria.
In the absence of such a system and to select the next agent for translation, the Consortium has developed a
Treatment Strategy Selection Committee, and this committee selected the most promising 5 agents for potential translation. The results show that the preclinical work on these 5 agents has left numerous gaps in knowledge about their
preclinical performance and confirm the need for significant changes in preclinical neuroprotection/pharmacotherapy
trials in SCI. A recommendation is made for the development and validation of a preclinical scoring system involving
worldwide experts in preclinical and clinical SCI.
(http://thejns.org/doi/abs/10.3171/2012.5.AOSPINE12116)

Key Words spinal cord injury neuroprotection pharmacotherapy

advances in the medical, surgical, and rehabilitation management of human SCI, there is
no widely accepted treatment that attenuates the
complex biological processes that constitute the secondary injury. One major category of treatment is neuroprotection by pharmacotherapy offered in the acute and subespite

Abbreviations used in this paper: BBB = Basso-Beattie-Bresnahan; MABP = mean arterial blood pressure; MDA = malondialdehyde; MgSO4 = magnesium sulfate; NACTN = North American
Clinical Trials Network; PEG = polyethylene glycol; SCI = spinal
cord injury; SSEP = somatosensory evoked potential.

J Neurosurg: Spine / Volume 17 / September 2012

acute phases of injury and designed to protect spinal cord


tissue from the severely damaging pathophysiological
events that occur in the CNS after physical trauma. There
have been remarkable advances in our understanding of
the secondary injury events after CNS trauma in both the
brain and spinal cord, and there are more than 25 secondary injury processes that have been identified, offering
multiple potential therapeutic opportunities to counteract
them.6,40,58,73 In the past 30 years a huge effort has been
expended by clinicians, basic scientists, and industry to
discover effective neuroprotective agents for SCI, which
thus far have largely failed to improve recovery. There
157

C. H. Tator et al.
have already been many comprehensive reviews of neuroprotective/pharmacological agents for SCI,6,71,72 and the
present report will not duplicate these reviews, but rather
is aimed at providing a focused analysis of the field of
neuroprotection/pharmacotherapy for SCI for the purpose of ascertaining the translational readiness of a selection of key promising agents.
The present review is focused on neuroprotection
afforded by pharmacotherapy, and so omits other neuroprotective strategies such as hypothermia. The review
has 2 objectives: 1) to identify, describe, and discuss the
strengths and weaknesses of the existing preclinical grading systems or recommended criteria (that is, translational
criteria) for determining whether a given pharmacological therapy should be translated from the laboratory into
clinical trials; and 2) using the information gained in the
first objective as a guide to identify, describe, and summarize the characteristics of preclinical trials that evaluate 5 neuroprotective agents that the NACTN has deemed
to be of current interest and that we have selected because
of their actual or potential for translation as neuroprotection for SCI. These include riluzole, glyburide, MgSO4
(with and without PEG), nimodipine, and minocycline.
The preclinical studies of these agents will be discussed
and the results put into the context of the translational criteria summarized from the first objective. It is of interest
that a recent publication that scored translational readiness of 12 agents for SCI trials included 3 selected by the
current authors.40

Methods

Electronic Literature Database

A systematic search was conducted in PubMed for


literature published from 1966 through November 2011.
Details of the search may be found in Tables 1 and 2. Results were limited to articles with abstracts published in
the English language. Reference lists of key articles were
also systematically checked.
For our first objective (Table 3), articles were identified that addressed issues related to the translational readiness of SCI pharmacological therapies for clinical trials.
The primary focus was to identify articles that proposed
specific grading criteria for studies on pharmacological
treatment of SCI. Unfortunately, there was a lack of literature on this topic; therefore, the search was expanded to
include those articles with a primary focus on evaluating
criteria used to translate a pharmacological therapy from
preclinical to clinical trials (that is, translational criteria). These criteria include experimental injury models,
the timing of therapy, evidence of beneficial effects of
therapy, safety and toxicity of therapy, reproducibility/
replication and publication of study results, and miscellaneous issues. Articles were excluded by title or abstract
if it was clear that the primary focus was not relevant to
SCI translation. Other exclusions included abstracts, letters, white papers, and studies not written in English.
For the second objective (Table 4), preclinical stud-

TABLE 1: Search strategy for Key Question 1: grading systems/criteria for translating pharmacological or cell-based SCI therapy from
laboratory to clinical studies*
Search
No.
1
2

3
4

Search Term

No. of Articles

Spinal Cord Injuries [Majr] OR spinal cord injury OR Spinal Cord Injuries/therapy* [MeSH]
Biomedical Research* OR Biomedical Research/methods [MeSH] OR Biomedical Research/
trends [MeSH] OR Clinical Trials as Topic* [MeSH] OR Diffusion of Innovation* [MeSH] OR
Drug Evaluation, Preclinical/methods [MeSH] OR Drug Evaluation, Preclinical/standards
[MeSH] OR Drug Evaluation, Preclinical/trends [MeSH] OR grading system [ti/abs] OR
Guidelines as Topic [MeSH] OR Translational research [MeSH] OR Translational research/
methods [MeSH] OR Translational research/standards [MeSH] OR Translational research/
trends [MeSH]
1 AND 2
3 NOT (neoplasm OR cancer OR coronary OR comparative study OR comparative studies OR
cross-sectional studies OR cross-sectional study OR prospective studies OR prospective
study OR multicenter studies OR multicenter study OR liver OR renal OR urinary OR
disasters OR malnutrition OR wound* OR retrospective studies OR retrospective study OR
multiple sclerosis OR pilot projects OR pilot study OR adaptation, psychological OR
grief OR dogs OR canine* OR social behavior OR pressure ulcer OR muscle contraction
OR controlled study OR controlled studies OR randomized controlled trial OR randomized
controlled trials OR spinal fusion OR follow-up study OR follow-up studies OR cats)

23125
240732

563
234

246 articles reviewed at ti/abs level (234


from PubMed search + 12 articles
identified in hand-searching relevant
bibliographies)
Include at ti/abs review: 26
Include at full-text review: 4
* PubMed search date: 11/16/2011; search limits: English, only items with abstracts. Abbreviation: ti/abs = title/abstract.

158

J Neurosurg: Spine / Volume 17 / September 2012

Translational potential of pharmacotherapy for SCI


TABLE 2: Search strategy for Key Question 2: preclinical studies*
Agent

Search
No.

Search Term

No. of Articles

1
2
3

Spinal Cord Injuries[Majr] OR spinal cord injury OR Spinal Cord Injuries/therapy*[MeSH]


Riluzole OR riluzole[MeSH]
1 AND 2

23154
772
27
Include at ti/abs review: 18
Include at full-text review: 12

1
2
3

Spinal Cord Injuries[Majr] OR spinal cord injury OR Spinal Cord Injuries/therapy*[MeSH]


Glyburide OR glibenclamide OR Glyburide[MeSH]
1 AND 2

23154
7205
5
Include at ti/abs review: 3
Include at full-text review: 3

1
2
3

Spinal Cord Injuries[Majr] OR spinal cord injury OR Spinal Cord Injuries/therapy*[MeSH]


magnesium OR magnesium sulfate OR magnesium sulfate[MeSH]
1 AND 2

23154
54651
71
Include at ti/abs review: 14
Include at full-text review: 9

1
2
3

Spinal Cord Injuries[Majr] OR spinal cord injury OR Spinal Cord Injuries/therapy*[MeSH]


nimodipine OR nimodipine[MeSH]
1 AND 2

23193
2991
30
Include at ti/abs review: 18
Include at full-text review: 14

1
2
3

Spinal Cord Injuries[Majr] OR spinal cord injury OR Spinal Cord Injuries/therapy*[MeSH]


minocycline OR minocycline[MeSH]
1 AND 2

23193
3557
42
Include at ti/abs review: 19
Include at full-text review: 16

riluzole

glyburide

MgSO4

nimodipine

minocycline

* Riluzole: search date, 11/28/2011; search database, PubMed; limits, Englishonly items with abstracts. Glyburide: search date, 11/28/2011; search
database, PubMed; limits, Englishonly items with abstracts. MgSO4: search date, 11/28/2011; search database, PubMed; limits, Englishonly items
with abstracts. Nimodipine: search date, 12/5/2011; search database, PubMed; limits, Englishonly items with abstracts. Minocycline: search date,
12/5/2011; search database, PubMed; limits, Englishonly items with abstracts.

ies were identified in which the neuroprotective effects of


riluzole, glyburide, MgSO4, nimodipine, or minocycline
were evaluated in animal models of SCI (including blunt
force trauma, penetrating trauma, or ischemia). Studies
were excluded by title or abstract if they evaluated fewer
than 5 animals, if the pharmacological agent was delivered prior to SCI, or if they were not explicitly testing
some aspect of the neuroprotective effect of the agent.
Studies that clearly indicated that the pharmacological
agent was being tested in non-SCI models, in models of
infection- or tumor-based SCI, ex vivo studies, or in vitro studies were also excluded. Full-text articles of the
remaining studies were obtained and reviewed for inclusion. Furthermore, articles that did not contain a control
group (that is, SCI plus saline, vehicle, or no treatment)
were excluded. Other exclusions included preclinical trials without outcome data, studies of human subjects, unJ Neurosurg: Spine / Volume 17 / September 2012

published data, technique papers, reviews, editorials, and


studies not written in English.

Data Extraction

Each retrieved citation was assessed by 2 reviewers


working independently (R.H. and A.R.). Most articles
were excluded on the basis of information provided by
the title or abstract. Full-text versions of all citations that
appeared to be appropriate, including those that could
not be excluded unequivocally on the basis of the title
and abstract, were then assessed by the 2 reviewers. Any
disagreement between them was resolved by consensus.
The following information was extracted for Objective
1: the type of therapy addressed, the basis of the criteria, scoring, components of criteria or preclinical study
characteristics, or issues discussed. For Objective 2 the
following data were extracted from the preclinical stud159

C. H. Tator et al.
TABLE 3: Inclusion and exclusion criteria for Objective 1: translational readiness for SCI
Study Component
Study design

Intervention

Preclinical study
characteristics
of interest

Publication

Inclusion
Articles that provide grading criteria to evaluate quality of pre clinical trials for clinical translation
Articles in which primary focus is on translational research
grading criteria, components, or issues

Articles in which primary focus is not on translational research


grading criteria, components, or issues
Articles that dont specifically address SCI translational research
issues
Articles in which primary focus is on addressing issues in clinical
research
Pharmacological agents being evaluated for improving outcomes Pharmacological agents being evaluated for improving outcomes
following SCI
in other disease models
Hypothermia & other physical modalities
Bioengineered scaffolds
Type of therapy addressed
Basis of criteria
Scoring
Components of criteria or preclinical study characteristics in cluding animal/injury models, timing of therapy, evidence of
beneficial effects of therapy, safety & toxicity of therapy,
reproducibility/replication & publication of study results, &
miscellaneous issues
Articles written in English in peer-reviewed literature w/ abs
Abs, letters
available
White papers

ies: animal model, injury model, experimental groups,


timing of therapy, dosage(s) used, route of intervention,
randomization of animals to treatment groups, blinded
or independent assessment of outcomes, and reporting of
results for all animals. Data on the effectiveness of the
therapy (including pathology of the lesion, biochemical
studies, electrophysiological studies, motor function, sensory function, other neurological function, neuropathic
pain, and spasticity) and the safety of the therapy, including toxicology and adverse events, were also extracted.
Data Analysis

Data for each key question were abstracted in tables.


For the first objective, grading criteria and issues related
to translational research in SCI, such as animal/injury
model and efficacy of a therapy were summarized, and
strengths and weaknesses of the group of articles were
discussed. This information was then used to propose
components that should be considered when evaluating
whether a given pharmacological therapy is ready for
clinical translation. For the second objective, the preclinical studies were summarized and discussed in terms of
the readiness of the specific agent for clinical translation. It was hypothesized that this analysis will serve as
a prototype methodology to evaluate which characteristics best determine whether a therapy is ready for clinical
translation and to suggest additional criteria that may be
included in such an evaluation process.

Results

Objective 1: Review of Criteria for Determining the


Translational Readiness of a Pharmacological Therapy
From the Laboratory Into Clinical Trials

We identified 246 articles from the literature search

160

Exclusion

that addressed readiness of SCI pharmacological therapies for clinical translation. However, only 26 articles
were judged suitable for full-text review, 22 of which
were excluded after full-text review for the following reasons: 19 articles did not specifically address translation
research of SCI pharmacological therapies, 1 article was
an update of one of the included articles, and 2 articles
addressed only cell-based therapies. Therefore, only 4 articles met the inclusion criteria, and only 1 of the 4 (Kwon
et al.41) proposed a scoring system. Although the other 3
discussed issues related to the translation of pharmacological therapies in patients with SCI or stroke,3,13,15 they
did not include a scoring system or discuss the relative
importance of the issues. The grading system proposed
by Kwon et al.41 assigns a total score based on the translational potential of a specific therapy, where a higher score
indicates a potentially more promising experimental
treatment for a clinical trial. A summary of the findings
is presented in Table 5.
Animal/Injury Model(s). All 4 articles addressed the
issue of the animal species and injury models used in
studies of pharmacological therapies. The grading scale
in Kwon et al.41 (Table 6) assigned the highest number of
points to treatments in studies using primate and largeanimal models (for example: dog, cat, sheep) and fewer
points for small rodents. Other authors also placed importance on the use of research in larger animals,13,15 with
Anderson et al.3 proposing that more invasive or higherrisk treatments be tested in the large-animal models. Several authors stressed that treatments should be deemed
effective in several animal models13,15 before moving to
clinical trials, with a lesion in which the volume, location,
and origin were representative of the type of SCIs that
occur in humans.15 Kwon et al.s grading scale assigned
the highest points to treatments in studies using cervical
J Neurosurg: Spine / Volume 17 / September 2012

Translational potential of pharmacotherapy for SCI


TABLE 4: Inclusion and exclusion criteria for Objective 2: preclinical studies of the 5 selected agents*
Study Component

Inclusion

Exclusion

Neuroprotective agents evaluated for improving outcomes w/ Tx


Selected neuroprotective agents (riluzole, glyburide, MgSO4,
nimodipine, & minocycline) evaluated for improving outcomes prior to SCI
post-SCI
Neuroprotective agents not explicitly tested for some aspect of
their neuroprotective effect
Population
Animal models of SCI, including (but not limited to) blunt force
Animal models of ALS, MS, etc.
trauma SCI, penetrating trauma SCI, ischemic SCI
In vitro models of SCI
Ex vivo models of SCI
Characteristics of Study characteristics, including:
interest
animal model
injury model
experimental groups
timing of therapy
dosage(s) used
route of intervention
randomization of animals to Tx groups
blinded or independent assessment of outcomes
whether all animals were accounted for
Effectiveness of therapy, including:
pathology of lesion & surrounding area
biochemical studies
electrophysiology studies
motor & neurological function
neuropathic pain
spasticity
Safety of therapy, including toxicology & adverse events
Study design
Comparative preclinical trials published in peer-reviewed jourStudies w/ no control group
nals
Studies evaluating <5 animals total
Incomplete studies (no results)
Clinical trials
Unpublished data
Technique papers

Intervention

* ALS = amyotrophic lateral sclerosis; MS = multiple sclerosis; Tx = treatment.

contusion and clip compression SCI models, fewer points


to thoracic contusion and clip compression, and the lowest number of points to cervical or thoracic partial sharp
transection SCI models.

Timing of Therapy. All 4 articles discussed the timing of the proposed therapy and its therapeutic efficacy.
Kwon et al.41 assigned a higher number of points to treatments that demonstrate efficacy after a longer treatment
delay postinjury. Dobkin15 and Dietrich13 proposed that
the timing and dose of the treatment replicate that which
a human patient would receive.

Evidence of Beneficial Effects of Therapy. All 4 articles addressed the issue of judging the beneficial effects
of a therapy. Kwon et al.41 assigned points to various behavioral and nonbehavioral outcome measures, including
locomotor tests, and considered the use of dose-response
analysis for behavioral and nonbehavioral outcomes in
the grading scale. Dietrich13 asserted that the quantitative
methods of assessing outcomes after treatment should be
clinically relevant. Anderson et al.3 suggested that any
therapeutic benefits of a treatment should persist for at
least 3 months after injury to ensure that true differences
J Neurosurg: Spine / Volume 17 / September 2012

exist between treated and untreated animals. Dobkin15


advised caution in the interpretation of behavioral outcomes found in rodent SCI models because many behaviors tested in rodents cannot be extrapolated to a similar
response in humans.

Reproducibility/Replication and Publication. All articles stressed the need for study results to be peer-reviewed
by independent experts,3,13 published in peer-reviewed
journals,3,13,41 and independently replicated3,13,15,41 before
translation to clinical trials. The grading system proposed
by Kwon et al.41 assigns a higher number of points to treatments having a greater number of studies that report beneficial effects of the therapy, with negative points assigned
to treatments having studies that report negative results of
the therapy.

Safety/Toxicity. Surprisingly, only 2 articles addressed


the need for safety or toxicity monitoring in preclinical
studies. Anderson et al.3 proposed that highly invasive or
risky interventions should meet a higher standard of preclinical safety and efficacy. They also stressed that animals
should be monitored for effects such as pain, worsened autonomic dysfunction, and spasticity for a time period de161

162

Kwon et al., 2011 (grading)

Consideration of chronic/acute injuries


Therapeutic benefits should persist 3 mos

Timing/dose similar to that which human


would receive
Caution in interpretation of behavioral
outcomes in rodent SCI models
NR

Invasive/risky therapies meet high standard


of safety/efficacy
Animals should be monitored for appropriate
time period for pain, worsened autonomic
dysfunction, & spasticity
Study results peer-reviewed by independent
experts
Publication of study results in peer-reviewed
journal
Study results independently replicated

Larger animals for invasive/high-risk Txs

Anderson et al., 2005

Use of larger animals


Txs shown to be effective in several
animal models
Injury model representative of SCI in
humans

Dobkin, 2007

Dietrich, 2003

Seeks knowledge of basic mechanisms


by which a therapy works

Study results peer-reviewed by indepen dent experts


Publication of study results in peer reviewed journal
Study results independently replicated

Timing/dose similar to that which human


would receive
Consideration of chronic/acute injuries
Quantitative methods assessing out comes should be clinically relevant
Safety/toxicity to be considered at every
study phase

Use of larger animals


Txs shown to be effective in several
animal models

* Kwon et al.41 is the only article with a point system assigned to the various criteria. Abbreviations: NA = not applicable; NR = not reported or addressed in the article.
Replication of the study results in independent laboratories in the same animal and injury models.

Reproducibility/replica- Publication of study results in peerStudy results independently replicated


tion & publication
reviewed journal
Study results independently replicated
(more points for studies w/ beneficial
results vs studies w/ negative results)
Other
NA
Considers that lab environment can con- NA
found interpretation of study results
Considers effect of multiple interventions/
routinely taken medications by human
patients

Animal/injury model(s) Animal species (more points for larger


animals)
Injury paradigms (more points for cervi cal contusion & clip compression vs
thoracic contusion & clip compression)
Timing of therapy
Time window of efficacy (more points
for efficacy demonstrated at longer
delay of Tx)
Evidence of beneficial Types of behavioral/nonbehavioral out effects of therapy
come measures
Safety/toxicity
NR

Key Criteria

TABLE 5: Summary of criteria addressed in the 4 selected articles related to the translational readiness of neuroprotection/pharmacotherapy for SCI from laboratory to clinical trial*

C. H. Tator et al.

J Neurosurg: Spine / Volume 17 / September 2012

Translational potential of pharmacotherapy for SCI


TABLE 6: Preclinical grading scale, neuroprotective therapies for acute SCI*
Component
Animal species in which efficacy has
been demonstrated

Injury paradigms in which efficacy has


been demonstrated

Time window of efficacy

Demonstration of clinically meaningful


efficacy

Independent reproducibility/replication

Items
Models of traumatic SCI
primate
large animal
rat
mouse
SCI models
cervical contusion
thoracic contusion
cervical clip compression
thoracic clip compression
cervical partial transection, sharp
thoracic partial transection, sharp
Efficacy demonstrated w/ Tx
delay of 12 hrs
delay of 4 hrs & <12 hrs
delay of 1 hr & <4 hrs
given immediately or at <1 hr
given prior to injury
Thoracic SCI model: plantar wt support vs failure in controls OR consistent forelimb hindlimb coordination vs failure in controls OR improvements in nonbehavioral outcomes
Thoracic SCI model: improvement in other locomotor/motor behavioral/nonmotor behavioral
tests in study showing improvements in nonbehavioral outcomes
Cervical SCI model: improvement in motor function tests in study showing associated im provements in nonbehavioral outcomes
Cervical SCI model: improvement in nonmotor behavioral tests in study showing associated
improvements in nonbehavioral outcomes
Dose response (thoracic or cervical SCI model): single study showing improvements in
either behavioral or nonbehavioral outcomes w/ changing doses of therapy
No. of independent labs reporting on beneficial or negative results of therapy
>10 (beneficial)
510 (beneficial)
34 (beneficial)
2 (beneficial)
1 (beneficial)
1 (negative)
23 (negative)
49 (negative)
>9 (negative)

Total max score

Max
Points Score
8
6
4
2
6
3
6
3
1
1
8
6
3
2
1
4

20

20

20

20

4
4
4
4
20
12
7
3
0
3
7
12
20

20

100

* Adapted from Kwon et al., 2011.41 Abbreviations: Max = Maximum; wt = weight.

signed to detect significant adverse events. Dietrich13 noted


that safety issues, including toxicity, should be considered
at every phase of the study.

Other. Other issues were noted by authors as needing


further research or discussion. Dobkin15 suggested that
the laboratory environment experienced by rodents might
confound the interpretation of experiments and thus the
translation of a treatment from animal models to human
studies. This environment includes deprivation of social
interaction, exercise, and environmental enrichment. Dietrich13 suggested that knowledge of the basic mechanisms by which a therapy works would assist researchers
in revising treatment protocols and investigating cause-

J Neurosurg: Spine / Volume 17 / September 2012

and-effect relationships between treatments and observed


outcomes.

Strengths and Weaknesses of Existing Grading Criteria. There are various strengths and weaknesses identified in each of the 4 articles that proposed grading criteria
or issues in the translational research of pharmacological
therapies for SCI, which are summarized in Table 7; more
detailed information is available in Table 8. Although
most of the articles discussed important issues such as
animal and injury models, reproducibility and publication of study results, and evidence of beneficial effects
of the therapy, there are some weaknesses. Most of the
articles did not discuss the relative importance of the is163

C. H. Tator et al.
TABLE 7: Summary of strengths and weaknesses of studies that address translational readiness of a pharmacotherapy for SCI trials
Strengths

Weaknesses

Large range of animals & use of relevant injury models, including randomization
& blinding, & adequate numbers of animals
Appropriate timing of therapy
Evidence of beneficial effects on the basis of a range of outcome measures
Safety/toxicity included in evaluation
Reproducibility/replication & detailed publication of study results

sues raised.3,13,15 Several articles made little or no mention


of safety or toxicity,15,41 and none of the articles discussed
the importance of randomizing the treatment or having
the investigators work in a blinded fashion. The grading
system of Kwon et al.41 has several additional weaknesses:
there was no consideration of the commercial potential
of a given therapy, only scientists or spine surgeonscientists were invited to the focus group meeting that decided the weighting factors for each subscale in the grading criteria, and the focus group was composed mainly of
American and Canadian researchers.
Objective 2: Preclinical Characteristics and Outcomes for
the 5 Selected Neuroprotective Agents

The following is a detailed account of the 5 agents


selected for analysis on the basis of their promise of usefulness for translation to human SCI.

Glyburide
Background. The management of parenchymal hemorrhage has been reported to be critical for promoting
neurological recovery after SCI.22 Glyburide works primarily by blocking sulfonylurea receptor 1 (SUR1)regulated, Ca2+-activated, [ATP]i-sensitive nonspecific cation
(NCCa-ATP) channels, which helps mitigate the effects of
secondary hemorrhage and progressive hemorrhagic necrosis following SCI.57,64,65 Glyburide has been approved
by the FDA for the treatment of Type 2 diabetes at a dose
of 1.2520 mg (standard) or 0.7512 mg (micronized)
orally in 1 or 2 divided doses.
Systematic Search for Preclinical Studies. Of the 5
studies identified in our literature search, 3 that evaluated
glyburide (glibenclamide) in preclinical studies were selected to undergo full-text review, and all 3 met the inclusion criteria.57,64,65
Study Characteristics. Study characteristics are summarized in Table 9; detailed information can be found
in Table 10. All 3 studies used rats, and no other species
were used to test the effects of glyburide. The total number of animals per study ranged from 10 to 54 (1 study did
not report the number), with 38 animals per treatment
group. A blunt force weight-drop injury model was used
in all 3 studies to injure the cervical spine. All 3 studies
compared glyburide to vehicle/saline following SCI, and
Simard et al.64 additionally included a no SCI/no treat164

No consideration of randomization or blinding


No discussion of relative importance of grading issues
No consideration of commercial potential of therapy
Only scientists/spine surgeons & mostly Americans &/or Canadians
in focus groups
Little mention of lab environments effects on interpretation of study
results
Little mention of knowledge of basic mechanisms by which a therapy
works

ment group. Simard et al.65 gave a loading dose of 10 mg/


kg glyburide within 15 minutes of SCI, and Popovich et
al.57 did the same in 1 of 3 experiments; otherwise, infusion began within minutes of the injury. Glyburide was
administered continuously at a rate of 200 ng/hour via
a subcutaneous osmotic pump in all studies; vehicle/saline was given similarly. No studies evaluated the doseresponse effects of glyburide. In all 3 studies, blinded investigators evaluated outcomes. Popovich et al.57 randomized animals to treatment groups, whereas no mention of
randomization was made by the other 2 studies. Popovich
et al. accounted for all 54 animals included in the study,
but the other 2 studies did not report whether all animals
were accounted for.
Effectiveness of Therapy. The effect of glyburide on
intraspinal hemorrhaging was evaluated by 2 studies. Simard et al.64 concluded that glyburide treatment resulted
in less hemorrhaging compared with the control groups
at 24 hours, as measured by spectrophotometric analysis
of blood in the cord homogenates (p < 0.05 at 6, 12, 18,
and 24 hours), as well as by other assays. Popovich et al.57
reported that glyburide given continuously at a dose of
200 ng/hour resulted in no differences in the amounts of
visible hemorrhaging between treatment groups at up to
24 hours postinjury in one experiment. However, in a second similar experiment, and in a third in which a loading
dose was given after injury and followed with continuous
infusion of lower doses, less hemorrhaging was visible
in the glyburide group compared with the SCI/vehicle
group at 24 hours. There were slight differences between
these experiments, including details of the weight drop
injury, which may have contributed to differences in these
results. The studies performed by Simard et al. in 2007
and 2010 both suggested that glyburide treatment resulted
in less visible hemorrhaging compared with the control
groups at 24 hours. Lesion size was also shown to be significantly reduced at 1 week64 and 6 weeks65 postinjury
in animals treated with glyburide compared with vehicle
in the 2 studies in which this outcome was evaluated (p
< 0.001).
Functional improvements in glyburide-treated rats
were observed in all 3 studies. Two studies reported
improvements in spontaneous rearing (vertical exploration), which was statistically higher in animals treated
with glyburide compared with vehicle at each time point
J Neurosurg: Spine / Volume 17 / September 2012

Neuroprotective
therapies for
acute SCI

Neural repair after


stroke or SCI

Dobkin, 2007

Type of Therapy

Kwon et al.,
2011
(grading
system)

Authors & Year

J Neurosurg: Spine / Volume 17 / September 2012


NA

Perspectives/opinions of 200+
clinicians & scientists in
SCI field via questionnaire
Final criteria by consensus of
25 invited scientific experts
& spine surgeonscientists
in modified Delphi exercise

Basis of Criteria

None

0100; extent to which a


particular Tx has been
studied, w/ higher
score indicating a
greater body of peer reviewed literature

Scoring

Strengths/Weaknesses

Provides an objective measure of the transla tional potential of a specific therapy based on
a systematically collected set of literature
supporting its application in acute SCI
Animal models in which efficacy has been demonstrated
Injury models in which efficacy has been demonstrated
Time window of efficacy
Demonstration of clinically meaningful efficacy
Independent reproducibility/replication

(continued)

No consideration of commercial potential of


therapy
Full spectrum of international input somewhat
lacking
Proposed Tx can possibly achieve a higher score
in absence of some elements
Excluded safety (deemed to be a regulatory
issue for all neuroprotective agents)
Only scientists or spine surgeonscientists (PIs)
invited to focus group meeting that decided
weighting factors for each subscale
Quality of scientific preclinical SCI studies not
assessed
Needs to be modified for other therapies (cell
transplantation or biological therapies)
Animal models designed to be relevant to human No indication of relative importance of issues
disease
Little mention of safety, adverse events, or
Differences btwn rodent brain/spinal cord & hu- toxicology
man CNS
Differences btwn various rodent models (replica tion of same injury & repair model in different
labs often not performed)
Reproduce key results in >1 lab
Differences in injury induction
Dose & timing of interventions
Anatomical site & goal of interventions
Combinational therapies & multiple interventions
Lab environment (for example, animals isolation
or deprivation), rehabilitation, postlesion reorganization
Physiological, histological, & molecular markers
can serve as surrogates for behavioral out comes, & limited rodent behaviors are tested

Components or Issues

TABLE 8: Grading criteria and issues related to translation research of pharmacological therapies for SCI*

Translational potential of pharmacotherapy for SCI

165

166

Anderson et
al., 2005

Authors & Year

Pharmacological,
cell-based, &
other therapies
for SCI

Type of Therapy
Based on guidelines issued
by the ASNTR (see Ramer
et al.)

Basis of Criteria
None

Scoring

Strengths/Weaknesses

(continued)

Appropriate data should be acquired in animals No indication of relative importance of issues


prior to human studies, including:
Injury model in humans should be validated
by preclinical model in animals (for
example, contusion or ischemia)
Time frame of Tx in animals should repli cate that of acute or chronic therapies in
humans
Riskier interventions should meet higher
standard of safety/efficacy
Tx should have demonstrable & statistically
significant benefit in animal models
Study results should be peer-reviewed by
independent experts
Outcomes should be assessed at cellular,
physiological, & behavioral levels in an
appropriate model of SCI
Persistence of therapeutic benefit should
be shown for at least 3 mos postinjury
Evidence of independent replication should
be present before translation
Animal models should be monitored for
safety/adverse effects (pain, worsened
autonomic dysfunction, spasticity) in sys tems above the lesion for a time period
designed to detect significant adverse
events

Components or Issues

TABLE 8: Grading criteria and issues related to translation research of pharmacological therapies for SCI* (continued)

C. H. Tator et al.

J Neurosurg: Spine / Volume 17 / September 2012

J Neurosurg: Spine / Volume 17 / September 2012

measured, ranging from 1 day to 1 week57 or 6 weeks (p


< 0.01).65 Two studies found improvements in inclinedplane test scores in animals treated with glyburide compared with vehicle as measured between 1 and 7 days (p <
0.05), 57,64 and 1 study each reported improvements in ipsilateral paw placement64 and in BBB scores (p < 0.001)57
at 1 day following injury.
Safety of Therapy. None of the studies assessed the
safety of glyburide.
Summary of Preclinical Trials of Glyburide. Thus,
according to the criteria shown in Tables 5 and 7, there
are numerous deficiencies in the translational readiness
of this agent.

* ASNTR = American Society for Neural Transplantation and Repair; PI = principal investigator.
The method or source used to propose criteria or issues in translating pharmacological therapies from preclinical to clinical studies.

Therapy must work in several animal models, in- No indication of relative importance of compo cluding consideration of animal species, sex, nents
& large animal models (nonhuman primates)
Compelling evidence of benefit, including the de gree of benefit, a wide therapeutic window,
dosing, clinically relevant methods of assess ing outcome, & ideally an understanding of
basic mechanisms by which the therapy works
Study is clinically relevant & replicated in an in dependent lab
Major findings are published in peer-reviewed
journals
Safety issues, including toxicity, are addressed at
every testing phase
Consideration of acute, subacute, & chronic in jury settings
None
NR
Neuroprotective or
regenerative
therapies for
SCI
Dietrich, 2003

Components or Issues
Scoring
Basis of Criteria
Type of Therapy
Authors &
Year

TABLE 8: Grading criteria and issues related to translation research of pharmacological therapies for SCI* (continued)

Strengths/Weaknesses

Translational potential of pharmacotherapy for SCI

Magnesium Sulfate
Background. For the treatment of SCI, the primary
mechanism of action for MgSO4 appears to be limiting
levels of intracellular calcium by blockage of N-methyld-aspartate receptors and of voltage-gated calcium channels, subsequently reducing free radical generation, glutamate release, expression of p53-related proteins, lipid peroxidation, lactate accumulation, and cell death.24,44,46,78,79,86
Magnesium sulfate is currently indicated in humans for
the immediate control of life-threatening convulsions in
the treatment of severe toxemias (preeclampsia and eclampsia) of pregnancy, for the treatment of acute nephritis in children, and as a replacement therapy in MgSO4
deficiency, especially in acute hypomagnesemia accompanied by signs of tetany. It is also used to prevent premature contractions in pregnancy and to treat patients with
heart attack and asthma. The drug can be administered
intramuscularly or intravenously, and the dose is very
variable, ranging from 1 to 3 g daily for a maintenance
dose for adults, to 1014 g for severe preeclampsia or eclampsia.
Systematic Search for Preclinical Studies. We identified 71 articles that evaluated MgSO4, of which 14 were
eligible for full-text review. Five studies were excluded
after full-text review for the following reasons: 4 studies
administered MgSO4 prior to injury, and 1 study did not
evaluate MgSO4. Therefore, 9 articles met our inclusion
criteria.14,26,35,36,42,53,66,69,81
Study Characteristics. Study characteristics are summarized in Table 11; detailed information can be found
in Table 12. Administration of MgSO4 following SCI
was tested in rats in 8 of the studies14,26,35,36,42,66,69,81 and
in rabbits in 1 study (Ozdemir et al.).53 The total number
of animals per study ranged from 30 to 122, with 620
animals per treatment group. Most studies evaluated thoracic SCI: blunt force trauma by weight drop was used in
6 studies26,35,36,42,66,81 and clip compression in 2 (Ditor et
al.14 and Szer et al.69); 1 study provided no details on the
injury model.53 In all studies, MgSO4 was administered
systemically, 5 intraperitoneally26,35,66,69,81 and 4 intravenously.14,36,42,53 The dosages of MgSO4 used were 60 mg/
kg (2 doses nearly 6 hours apart),42 100 mg/kg,35,53 300 mg/
kg,14,69 and 600 mg/kg.26,35,36,66,69,81 Only Kaptanoglu et al.35
and Szer et al.69 evaluated more than one dose of MgSO4.
All but 1 study gave the first injection of MgSO4 within
015 minutes postinjury; Szer et al. administered the
167

C. H. Tator et al.
TABLE 9: Summary of glyburide preclinical study characteristics*
Characteristic
animal model: rats
no. of animals
no./group
injury model: blunt force (wt drop)
cervical
timing of intervention postinjury
23 min w/ cont infusion
15 min w/ cont infusion for 7 days
route of intervention: osmotic pumps placed caudal to injury
in subcutaneous pocket
IP
dosage
200 ng/hr cont infusion until planned death
initial dose of 10 g/kg; cont infusion of 200 ng/hr for 7 days
>1 dose evaluated?
yes
no
control groups
SCI (saline, DMSO)
no SCI
independent or blind assessment
yes
no
NR
animals randomized to Tx groups
yes
no
NR
% animals accounted for

Popovich et al., 2012

Simard et al., 2007

Simard et al., 2010

54
68

NR
35

10
5

X
X

X
X

X
NR

X
NR

100% (54 of 54)

* Cont = continuous; DMSO = dimethyl sulfoxide; IP = intraperitoneal.

drug at 1 hour. Wiseman et al.81 conducted 2 experiments,


one in which MgSO4 was given at 10 minutes, and another
in which animals received an injection at either 8, 12, or
24 hours. In 2 studies injections were given at 15 minutes
and again at 6 hours: in Ditor et al.14 at 300 mg/kg/injection
and Kwon et al.42 at 60 mg/kg/injection. All but 1 study53
clearly stated that animals were randomized to treatment
groups, and that assessment was done in a blinded manner;
all but 2 studies53,66 used blinded assessment of treatment
outcomes. Five studies accounted for 100% of the animals;14,26,35,42,69 1 study accounted for 100% of the animals
in the first experiment, but did not account for the animals
in the second experiment;81 3 studies did not account for all
included animals.36,53,66
Effectiveness of Therapy. Lesion size was assessed
by 2 studies. Kwon et al.42 found that 6 weeks after SCI,
intravenous administration of MgSO4 (2 doses of 60 mg/
kg, one at 15 minutes, and the other at 6 hours) reduced
lesion volumes by 33% compared with rats that received
saline alone (p = 0.03). When MgSO4 was given in PEG,
168

lesion volumes were reduced even more, by 51% compared with saline controls (p = 0.0012) (PEG alone reduced lesion volumes by 20% compared with saline
controls). The delivery of MgSO4 in PEG significantly
reduced lesion size compared with administration of
MgSO4 alone (p = 0.0386). In contrast, Wiseman et al.81
reported no differences in lesion lengths between treatment groups measured 24 hours postinjury, but noted that
the small number of specimens and the large variability
in the control group samples may have affected the results. The authors reported significantly improved white
matter sparing following MgSO4 (and methylprednisolone + MgSO4) treatment compared with saline alone: the
percentage of myelin preservation was 20.2% in the saline group compared with 32.3% (p = 0.002) for MgSO4,
30.3% (p = 0.006) for methylprednisolone, and 42.3% (p
= 0.0007) for the 2 drugs combined. In this study, rats
were treated with 600 mg/kg MgSO4 (and/or 30 mg/kg
methylprednisolone) 10 minutes after injury.
The effects of MgSO4 on neuronal apoptosis followJ Neurosurg: Spine / Volume 17 / September 2012

J Neurosurg: Spine / Volume 17 / September 2012


No SCI (controls)
SCI + vehicle (saline
+ DMSO)
SCI + glibenclamide
35 rats/group

Simard et al.,
2007

Animal model:
Long-Evans rats
N = NR
Sex: 100% F
Age: adults
Wt: 275350 g
SCI model:
Blunt force impactor; injury
at C45 level (1.3-mm
impactor head driven
by 10-g wt dropped
vertically from a 25-mm
height)

Experimental
Groups
SCI + vehicle (saline
+ DMSO; controls)
SCI + glibenclamide
68 rats/group

Animal & Injury Models

Popovich et al., Animal model:


Long-Evans rats
2012
N = 54
Sex: 100% F
Age: 10 wks
Wt: 247 8 g
SCI model:
Blunt force impactor; injury
at C-5 level (1.3-mm im pactor head driven by
10-g wt dropped verti cally from a 25-mm
height)

Authors &
Year
Reported Outcomes

Gross Visual Inspection of the Whole Spinal Cord:


Expt 1: at 6, 12, or 24 hrs postinjury revealed similar
amounts of hemorrhage at the site of impact, w/ some
visible blood extending into rostral & caudal spinal seg ments in both vehicle & glibenclamide-treated animals
Expt 2: acute delivery of glibenclamide was found to limit
intraspinal hemorrhage (vs vehicle)
Expt 3: 6 rats were treated w/ glibenclamide or vehicle (3/
group), then intraspinal hemorrhage was assessed 24
hrs later; a reduction in intraspinal hemorrhage was
found similar to Expt 2
Motor Tests:
Significant preservation of hindlimb function was evident
in glibenclamide-treated rats 1 day postinjury according
to the BBB locomotor rating scale
Similar improvements were detected on the inclined-plane
& cylinder tasks
Rearing & paw placement btwn 3 & 7 days: 22 rearing
events for glibenclamide compared w/ 3 events for
vehicle-treated rats
Average duration of rearing was ~15 sec for glibenclamide
& ~1 sec for vehicle
Safety/Adverse Events/Toxicology: NR
Route: osmotic pumps placed caudal to injury Upregulation of SUR1 in SCI:
site in a subcutaneous pocket
In controls, SCI caused a progressively expansive lesion
Timing: 23 min after SCI
w/ fragmentation of capillaries, hemorrhage that
Dosage:
doubled in vol over 12 hrs, tissue necrosis, & severe
50 mg (or 25 mg) of drug into 10 ml DMSO
neurological dysfunction
Infusion solutions made by placing 400 l (or SUR1 expression was upregulated in capillaries & neu 800 l) stock into 4.6 ml (or 4.2 ml)
rons surrounding necrotic lesions
unbuffered saline (0.9% NaCl)
Patch clamp of cultured endothelial cells exposed to hyInfusion solutions of glibenclamide & repa poxia showed that upregulation of SUR1 was associ glinide were delivered at 0.5 l/hr, yielding ated w/ expression of functional SUR1-regulated
infusion doses of 200 ng/hr
NCCa-ATP channels
Animals killed: at various times after SCI
Following SCI, block of SUR1 by glibenclamide or repag(511/group)
linide or suppression of Abcc8 was associated w/
significant sparing of white matter tracts & a 3-fold re duction in lesion vol, & resulted in marked neurobehav ioral functional (inclined-plane test & ipsilat paw place ment) improvement compared w/ controls
Safety/Adverse Events/Toxicology: NR

Route: osmotic pumps placed caudal to injury


site in a subcutaneous pocket
Timing: 23 min after SCI
Dosage:
Control: equivalent vol of DMSO diluted in
0.9% NaCl
Glibenclamide: 800 l of stock solution (5.0
mg/ml glibenclamide dissolved in DMSO)
diluted w/ 4.2 ml sterile 0.9% NaCl
Infusion rate: 0.5 l/hr, yielding 200 ng/hr of
glibenclamide
Expt 1: at OSU (injury model: Infinite Horizons
blunt impact)
Expt 2: at UM (injury model: wt drop tech nique), + used an immediate postinjury (w/
in 5 min) loading dose of glibenclamide
(10 g/kg)
Expt 3: at OSU w/ UM injury device
Animals killed: NR

Intervention Details

TABLE 10: Characteristics of SCI animal studies using glyburide (glibenclamide)*

(continued)

Main goal of article


was to replicate
Simard et al.
(2007) results

Comments

Translational potential of pharmacotherapy for SCI

169

170

* BBB = Basso-Beattie-Bresnahan motor score; Expt = experiment; inj = injection; NCCa-ATP = Ca2+-activated, [ATP]i-sensitive nonspecific cation; OSU = Ohio State University; SUR1 = sulfonylurea
receptor 1; UM = University of Maryland.

Glibenclamide Tx in rats resulted in significantly (p <0.01)


better performance in spontaneous rearing during 6 wks
of observation compared to vehicle-treated controls
Glibenclamide Tx in rats resulted in a 34 reduction in
lesion vol compared w/ vehicle-treated rats (p <0.001)
Glibenclamide Tx in rats resulted in absence of cyst forma tion & spinal cord cavitation, both of which were present
in vehicle-treated rats
Safety/Adverse Events/Toxicology: NR
Route: subcutaneous inj
Dosage & timing:
W/in 15 min of SCI, a loading dose of gliben clamide (10 g/kg) was given IP
Cont infusion of glibenclamide (200 ng/hr
subcutaneously for 7 days)
Animals killed: NR
SCI + glibenclamide
(n = 5)
SCI + saline/DMSO
(n = 5)
Animal model:
Rats
N = 10
SCI model:
Rats had a unilat cervical
injury
Simard et al.,
2010

Intervention Details
Experimental
Groups
Animal & Injury Models
Authors &
Year

TABLE 10: Characteristics of SCI animal studies using glyburide (glibenclamide)* (continued)

Reported Outcomes

Comments

C. H. Tator et al.
ing SCI were examined by Solaroglu et al.66 by measuring
caspase-3 activity. A single dose of MgSO4 (600 mg/kg)
was given immediately following weight drop injury, and
animals were killed at 24 hours. Compared with no treatment or vehicle alone, MgSO4 reduced caspase-3 activity
levels (p < 0.05) in experimental animals, as did methylprednisolone. Of note, methylprednisolone treatment
resulted in a greater reduction in caspase-3 activation
compared with MgSO4 (p < 0.05). Caspase-3 activity was
measured in 1-cm samples of spinal cord tissue homogenates taken around the injury site. The results suggest
that MgSO4 may have antiapoptotic effects in the first 24
hours following SCI.
Kaptanoglu et al.35 reported the ultrastructural findings in a 1-mm cross-section of the spinal cord obtained
at the trauma site after 2 different doses of MgSO4 (100
and 600 mg/kg) given immediately after weight drop SCI
in rats killed at 24 hours. The higher dose of MgSO4 significantly improved the general neural score, which assessed whole subcellular changes, compared with the no
treatment or saline controls (p < 0.001); in fact, the scores
from the animals that received this dose were statistically
similar to those in the no injury control group. This effect was not seen with the lower dose of MgSO4 (100 mg/
kg). Similar results were seen for the higher but not lower
dose of MgSO4 for measurements of intracytoplasmic
edema, nuclear protection, and axon myelin. Both doses
of MgSO4 improved the axonal score (a measure of axonal injury) compared with the no treatment control (p <
0.05), and the improvements were statistically similar to
the no injury control group.
Spinal cord lactate and/or MDA levels were assessed
in 2 studies. Lactate accumulation and MDA formation
occur following neural injury. Ozdemir et al.53 found that
rabbits treated with 100 mg/kg MgSO4 5 minutes after
SCI had significantly lower lactate and MDA levels than
animals treated with saline. Furthermore, these levels were
statistically similar to those seen in the no injury control
animals. Szer et al.69 reported similarly decreased MDA
levels 24 hours postinjury in rats treated with a higher dose
(600 mg/kg) but not a lower dose (300 mg/kg) of MgSO4
1 hour after SCI.
Gok et al.26 reported that infiltration of neutrophils
into the spinal cord region, as evaluated by myeloperoxidase activity of spinal cord homogenates, was significantly reduced in animals treated with MgSO4 (or methylprednisolone) compared with saline (p < 0.05). Treatment
was given immediately after trauma; the time at which
the tissue samples were collected was not reported.
Vascular permeability was assessed at 2 and 24 hours
following SCI in rats treated with 600 mg/kg MgSO4 immediately after trauma by Kaptanoglu and colleagues.36
The authors evaluated the extent of bloodspinal cord
barrier damage and the increase in microvascular permeability by using a 10-minute perfusion of Evans blue
dye. Animals treated with MgSO4 had lower Evans blue
content and thus lower vascular permeability than the
trauma group at both 2 and 24 hours, although the Evans
blue content was still higher than in the no trauma control
animals. Furthermore, Evans blue content increased with
time in the trauma group who received no MgSO4. These
J Neurosurg: Spine / Volume 17 / September 2012

animal model (no./group)


rats
rabbits
injury model
blunt force (wt drop; thoracic)
clip compression (thoracic)
timing of intervention postinjury
inj @ 0 hrs
inj @ 5 min
inj @ 10 min
inj @ 8, 12, or 24 hrs
inj @ 15 min & 6 hrs
inj @ 1 hr
route of intervention
IV; tail inj
IV; jugular vein
IV; details NR
IP
dosage
60 mg/kg
100 mg/kg
300 mg/kg
600 mg/kg
>1 dose evaluated?
yes
no
control groups
SCI (PEG)
SCI (saline)
SCI (no Tx)
no SCI (no Tx)
independent or blind assessment
yes
no
NR

Characteristic

J Neurosurg: Spine / Volume 17 / September 2012


X

X
X

32 (611)

Ditor et al.,
2007

X
X
X

X
X
X

50 (10)

Kaptanoglu et al.,
200335

35 (7)

Gok et al.,
2007

TABLE 11: Summary of MgSO4 preclinical study characteristics*

X
X

42 (6)

Kaptanoglu et
al., 200336

X
X

2 doses

50 (10)

Kwon et al.,
2009

X
X

NR

30 (10)

X
X
X

40 (8)

Ozdemir et al., Solaroglu et


2005
al., 2005

X
X

30 (10)

Szer et al.,
1999

(continued)

Expt 1
Expt 2

122 (1320)

Wiseman et al., 2009

Translational potential of pharmacotherapy for SCI

171

172

100% (30 of 30) Expt 1: 100% (70 of 70)


Expt 2: NR
* IV = intravenous.

NR
100% (50 of 50)

100% (50 of 50)

X
NR

NR

X
X
X
X
X
X

animals randomized to Tx groups


yes
X
X
no
NR
% animals accounted for
100% (32 of 32) 100% (35 of 35)

Kaptanoglu et al.,
200335
Gok et al.,
2007
Ditor et al.,
2007
Characteristic

TABLE 11: Summary of MgSO4 preclinical study characteristics* (continued)

Kaptanoglu et
al., 200336

Kwon et al.,
2009

Ozdemir et al., Solaroglu et


2005
al., 2005

Szer et al.,
1999

Wiseman et al., 2009

C. H. Tator et al.
results indicate that MgSO4 may be able to provide some
(but not complete) protection against bloodspinal cord
barrier damage.
Locomotor function was evaluated in several different ways. Four studies reported improved results on the
BBB test in animals treated with MgSO4 compared with
saline (or no treatment) as measured between 24 hours
and 6 weeks following SCI.14,36,42,81 Two of these studies
assessed the effect that the addition of PEG would have on
the results. Kwon et al.42 reported that MgSO4 in PEG was
necessary to provide significant improvement, but Ditor et
al.14 found that the addition of PEG did not have a significant effect on the results. Wiseman and colleagues81 found
that while treatment with MgSO4 improved BBB scores at
4 weeks compared with saline, there were no differences
between these groups at earlier time points. The authors
also found that rats had better BBB scores when MgSO4
was administered earlier following injury (that is, within
8 hours compared with 12 or 24 hours; p < 0.01). Three
studies reported improved function measured by the inclined-plane test in rats that received MgSO4 compared
with saline (or no treatment).26,35,36 Kaptanoglu and colleagues35 found that a dosage of 600 mg/kg (vs 100 mg/
kg) MgSO4 was necessary to elicit substantial functional
improvements as measured by the BBB scoring system,
the inclined-plane test, and a modified Tarlov scale.
Ditor and colleagues14 reported improvements in
measures of neuropathic pain (avoidance responses to
touch stimuli) in rats treated with MgSO4 (with or without
PEG) compared with saline alone, as measured 6 weeks
post-SCI. Animals had been treated at 15 minutes and 6
hours after trauma with 300 mg/kg MgSO4.
Szer and colleagues69 measured spinal SSEPs in rats
that had undergone clip compression and treatment with
MgSO4 (300 or 600 mg/kg) or saline 1 hour after injury.
All animals had significantly reduced SSEP amplitudes
30 minutes postinjury (pretreatment) compared with the
amplitudes measured prior to injury. At 4 hours after SCI,
animals treated with the higher dose of MgSO4 had significant improvements in the SSEP amplitudes of P1 and N1
(p < 0.01 for both) compared with the 30-minute postinjury
measurements. The same result was not observed in animals that received the lower dose of MgSO4 (or saline). The
authors did not offer comparisons for SSEPs measured at 4
hours between treatment groups.
Safety of Therapy. Wiseman and colleagues81 reported a higher rate of autophagia in rats treated with MgSO4
(600 mg/kg in a single injection) compared with animals
that received saline or steroid injections; however, rates
were not reported. The authors noted that autophagia began 510 days postinjury and attributed it to the return of
(unpleasant) sensation in the hindlimbs. No other studies
reported on the safety of MgSO4.
Summary of Preclinical Trials of MgSO4. Thus, significant benefit has been recorded after MgSO4 in most
preclinical studies, although there are still considerable
gaps in our knowledge of this agent in experimental SCI.
Minocycline
Background. Minocycline has been shown to target
multiple processes involved in mediating cell death and

J Neurosurg: Spine / Volume 17 / September 2012

Control (no SCI or Tx)


SCI (no Tx; trauma
group)
SCI + saline (vehicle)
SCI + MPSS
SCI + MgSO4 (magnesium)
7 rats/group

Animal model:
Wistar rats
N = 35
Sex: 100% F
Age: adult
Wt: 210250 g
SCI model:
Wt drop method;
contusion injury at
T79 spine level

Gok et al.,
2007

J Neurosurg: Spine / Volume 17 / September 2012


Route: IP
Timing: immediately after trauma
Dosage (single dose):
Control: no medication
Trauma: no medication
Vehicle: 1 ml of saline
MPSS: 30 mg/kg MPSS
Magnesium: 600 mg/kg MgSO4
Animals killed: at end of expt, all
animals were killed under deep
anesthesia

Route: IV tail vein inj


Timing: Txs given at 15 min & 6 hrs
postinjury
Dosage:
PEG: 1 g/kg, 30% w/w in sterile saline
MgSO4: 300 mg/kg
PEG + MgSO4
Saline: isovolumetric doses of saline
Animals killed: 6 wks post-SCI

SCI + PEG (n = 11)


SCI + MgSO4 (n = 5)
SCI + PEG + MgSO4
(n = 6)
SCI + saline (controls)
(n = 10)

Intervention Details

Experimental Groups

Animal model:
Wistar rats
N = 32
Sex: 100% M
Age: adult
Wt: 200250 g
SCI model:
T-4 spinal cord seg ment injured by
50-g clip compression

Animal & Injury


Models

Ditor et al.,
2007

Authors &
Year

TABLE 12: Characteristics of SCI animal studies using MgSO4*

Locomotor Function (BBB 21-point score):


PEG vs MgSO4 vs Saline vs PEG + MgSO4
42 days postinjury: 7.3 0.2 vs 7.7 0.4 vs 6.4 0.6 vs 7.6 0.2
4 of 10 saline, as opposed to only 2 of 11 PEG & 0 of 5 MgSO4
animals had <7 (extensive movement at 3 joints) BBB score
Although PEG + MgSO4 group had a significantly better locomotor
recovery compared to saline, the combination therapy added no
benefit compared w/ PEG or MgSO4 alone
Neuropathic Pain (avoidance responses to touch stimuli):
PEG vs MgSO4 vs Saline vs PEG + MgSO4
6 wks postinjury: 3.5 0.4 vs 2.8 0.9 vs 5.0 0.5 vs 3.3 0.9
All 3 Txs (PEG, MgSO4, & PEG + MgSO4) showed significantly fewer
avoidance responses compared to saline; however, the combination
PEG + MgSO4 Tx provided no added benefit compared w/ individual
Txs w/ PEG & MgSO4
Cardiovascular Function:
PEG vs MgSO4 vs Saline vs PEG + MgSO4
Change in MAP during colon distention (in mm Hg): 42.8 2.6 vs 38.3
4.2 vs 42.2 2.9 vs 42.7 3.0
None of the Txs had a significant effect on the severity of autonomic
dysreflexia
Only the resting MAP in the PEG group was significantly different
compared to the controls (p = 0.03)
Safety/Adverse Events/Toxicology: NR
MPO Activity (indicator of extent of posttraumatic neutrophil infiltration):
Statistically Significant Difference Btwn:
Control & trauma groups (p <0.05); trauma was found to raise MPO
activity
Trauma group compared w/ MgSO4 & MPSS groups (p <0.05 in
both); MgSO
4 & MPSS Txs decreased MPO activity
MPSS & MgSO4 groups (p <0.05); MPSS was more effective than
MgSO
4 in preventing the activity of MPO after SCI
No statistical difference btwn the vehicle group & the trauma group
(p >0.05)
Functional EvaluationInclined Plane (degrees):
Statistically significant difference in the means of inclined-plane scores
among groups (p <0.05)
Trauma showed statistically significant decrease in inclined-plane
values compared to control groups (p <0.05)
No significant differences btwn trauma, vehicle, & MPSS groups (p >0.05)
MgSO4 group showed significant increase in inclined-plane values
compared to trauma, vehicle, & MPSS groups (p <0.05)
Significant difference btwn control & MgSO4 groups (p <0.05)
Safety/Adverse Events/Toxicology: NR

Reported Outcomes

(continued)

Functional evaluation
performed 24 hrs
after trauma by
blinded observers

Comments

Translational potential of pharmacotherapy for SCI

173

174

Animal & Injury


Models

Animal model:
Sprague-Dawley
rats
N = 50
Sex: 100% F
Age: NR
Wt: 180230 g
SCI model:
Wt drop method;
injury at T78
spine level (50 g/
cm impact trauma
produced by a
stainless steel rod
[3 mm diameter,
weighing 5 g],
dropped vertically
through a calibrat ed tube from a
height of 10 cm)

Authors &
Year

Kaptanoglu et
al., 200335

Reported Outcomes
Ultrastructural Findings (scores range from 0 to 3; a score of 0
indicates a better outcome):
General Neural Score (whole subcellular changes)
Comparison of trauma & vehicle (p >0.05), & trauma & M100 (p >0.05)
revealed no significant difference btwn these groups
Vehicle & M100 did not show neuroprotection
Comparison of trauma & M600 (p <0.001): M600 Tx prevented tissue
injury, & results were similar to the no-Tx control
Intracytoplasmic Edema
Statistically significant difference btwn control & trauma, & btwn trauma
& M600 (p <0.001)
No difference btwn control & M600
Trauma, vehicle, & M100 groups showed similar intracytoplasmic
edema (no significant difference)
Nucleus
M100 provides limited nuclear protection compared to M600
Axon Myelin
Control & M600 showed significant difference from trauma, vehicle, &
M100 groups (p <0.05)
Clinical Findings:
Inclined-Plane Values (degrees)
No significant differences btwn trauma, vehicle, & M100 groups
(p >0.05)
M600 showed significant increase in inclined-plane values compared
to trauma, vehicle, & M100 groups (p <0.05)
Tarlov & BBB Scores:
M600 demonstrated significantly better results than trauma, vehicle, &
M100 groups (p <0.05)
Although Tarlov & BBB results of M600 were close to control group,
there was a statistically significant difference (p <0.05)
Safety/Adverse Events/Toxicology: NR

Intervention Details
Route: IP
Timing: immediately after trauma
Dosage:
Vehicle: 1-ml single dose of physio logical saline solution
M100: 100 mg/kg single dose of
MgSO
4
M600: 600 mg/kg single dose of
MgSO
4
Animals killed: rats were killed w/ de capitation under general anesthe sia immediately after spinal cord
samples were obtained (24 hrs)

Experimental Groups
Control (no SCI or Tx)
SCI (no Tx; trauma)
SCI + saline (vehicle)
SCI + MgSO4 (M100)
SCI + MgSO4 (M600)
10 rats/group

TABLE 12: Characteristics of SCI animal studies using MgSO4* (continued)

(continued)

Comments

C. H. Tator et al.

J Neurosurg: Spine / Volume 17 / September 2012

Animal & Injury


Models

Animal model:
Sprague-Dawley
rats
N = 42
Sex: 100% F
Age: adult
Wt: 220270 g
SCI model:
Wt drop method;
T79 spinous pro cesses were re moved & spinal
cord contusion
injury was per formed w/ 50 g/
cm force

Authors &
Year

Kaptanoglu et
al., 200336

Part I:
SCI (controls [C]; no
Tx)
No SCI or Tx (lami nectomy only; LI)
SCI (no Tx; trauma
[TI])
SCI + MgSO4 (RxI)
Part II:
No SCI or Tx (lami nectomy only; LII)
SCI (no Tx; trauma
[TII])
SCI + MgSO4 (RxII)
6 rats/group

Experimental Groups

J Neurosurg: Spine / Volume 17 / September 2012


Part I: animals killed
after Evans blue
application 2 hrs
posttrauma
Part II: animals killed
24 hrs posttrauma
Clinical evaluations
performed only in
Part II of the study

Evans Blue (indicator of vascular permeability & used to assess bloodspinal cord barrier damage & increased microvascular permeability) (in
mg/g wet tissue):
Control vs Laminectomy vs Trauma vs Tx (600 mg/kg MgSO4 )
2 hrs: 2.02 0.35 vs 2.12 0.27 vs 12.24 1.46 vs 6.37 0.92
24 hrs: 2.02 0.35 vs 2.22 0.27 vs 21.59 3.30 vs 8.90 1.63
Clinical Findings (24 hrs):
Control vs Laminectomy vs Trauma vs Tx (600 mg/kg MgSO4 )
Inclined-plane values (degrees):
74.17 3.76 vs 72.50 5.24 vs 46.67 4.08 vs 64.17 5.85
BBB Score:
21.00 0.00 vs 20.50 0.55 vs 8.17 1.17 vs 11.50 2.59
Tarlov Score:
5.00 0.00 vs 5.00 0.00 vs 1.83 0.98 vs 3.33 0.82
Significant difference btwn trauma & Tx groups in tissue Evans blue
content at 2 & 24 hrs (p <0.01) & clinical findings of the 24-hr groups
(inclined-plane values, p <0.01; BBB scores, p <0.05; Tarlov scores,
p <0.05)
MgSO4 Tx prevented high Evans blue content of the spinal cord &
worsening of clinical results
Tissue Evans blue content increased w/ time following SCI; this pro gressive vascular damage indicated secondary injury
Safety/Adverse Events/Toxicology: NR

Route: IV access through jugular


veins
Timing (1-cm spinal cord samples):
C: immediately after laminectomy
LI: 2 hrs after laminectomy
TI: 2 hrs posttrauma
RxI: dosage immediately after induc tion of SCI & samples obtained 2
hrs after trauma
LII, TII, & RxII samples taken after 10
min of perfusion w/ Evans blue
following clinical examination
Dosage: RxI & RxII: 600 mg/kg single
dose of MgSO4
Animals killed:
Part I: animals killed after Evans blue
application 2 hrs posttrauma
Part II: animals killed 24 hrs posttrauma

(continued)

Comments

Reported Outcomes

Intervention Details

TABLE 12: Characteristics of SCI animal studies using MgSO4* (continued)

Translational potential of pharmacotherapy for SCI

175

176

Kwon et al.,
2009

Authors &
Year

Animal model:
Sprague-Dawley
rats
N = 50
Sex: 100% F
Age: adult
Wt: 200225 g
SCI model:
Infinite Horizon Im pactor; laminecto my at T-10, im pactor delivered
contusion injury
w/ a force of 150
kdyn

Animal & Injury


Models
Intervention Details
Route: IV access through jugular
veins
Timing:
1st IV inj 15 min postinjury
2nd IV inj 6 hrs postinjury
Dosage:
MgSO4 60 mg/kg
PEG1 g/kg
Animals killed: at 6 wks postinjury,
animals were killed w/ an IP inj of
sodium pentobarbital

Experimental Groups
No SCI or Tx (sham
controls)
SCI + saline (saline
controls)
SCI + MgSO4 in saline
SCI + PEG
SCI + MgSO4 in PEG
10 rats/group

TABLE 12: Characteristics of SCI animal studies using MgSO4* (continued)


Comments

(continued)

Sought to reproduce
Results:
the results of Ditor
Saline Control vs MgSO4 vs PEG vs MgSO4 in PEG
Lesion vols (in mm3): 3.39 0.47 vs 2.28 0.19 vs 2.71 0.41 vs 1.65 et al. (2007)
0.18 (these lesion vols represent a 33%, 20%, & 51% reduction in
lesion size compared to saline controls)
BBB scale transformed to 12-point Ferguson scale (6 wks postinjury):
7.50 0.11 vs 7.78 0.32 vs 8.30 0.65 vs 8.78 0.32* (*signifi cant improvement over saline control [p = 0.0501])
No. of infusions (6 wks postinjury)
Animals treated w/ 2, 4, & 6 infusions (127 mol/kg of MgCl2 per infu sion) every 8 hrs had lesion vols (in mm3) of 3.16 0.43, 2.68
0.27, & 2.18 0.28
Dose-response effect, w/ 4 & 6 infusions providing greater reductions
in lesion vol & better locomotor recovery than 2 infusions
Dose of MgCl2the lesion vol (in mm3) for rats treated w/ the 254 mol/kg dose was smaller than that of the 127-mol/kg dose (3.16
0.43 vs 1.86 0.31); this difference trended toward, but did not
achieve, statistical significance (p = 0.068)
Shortened time btwn infusions (6 hrs rather than 8 hrs) provided im proved locomotor recovery
No major differences btwn MgCl2 & MgSO4, although there was a
slightly better early recovery in the MgCl2 group
Very early improvements in locomotor function (w/in the first few days)
were associated w/ reduced lesion vol (as measured at 42 days
postinjury), suggesting the Tx was having an early effect on reducing
secondary damage
Safety/Adverse Events/Toxicology: NR

Reported Outcomes

C. H. Tator et al.

J Neurosurg: Spine / Volume 17 / September 2012

J Neurosurg: Spine / Volume 17 / September 2012

Animal model:
NZ rabbits
N = 30
Sex: NR
Age: NR
Wt: NR
SCI model:
NR (SCT)

Animal & Injury


Models

Solaroglu et al., Animal model:


Wistar albino rats
2005
N = 40
Sex: 100% F
Age: adult
Wt: 210250 g
SCI model:
Wt drop method;
T79 spinous pro cesses removed,
laminectomy per formed, spinal
cord contusion in jury w/ 40 g/cm
force

Ozdemir et al.,
2005

Authors &
Year
Route: IV
Timing:
Tx 5 min after SCT
Lactate & MDA levels tested 60 min
after SCT
Dosage: Group III100 mg/kg of
MgSO
4
Animals killed: NR

Route: IP inj
Timing: immediately after SCI
Dosage:
MPSSsingle dose of 30 mg/kg
Magnesiumsingle dose of 600 mg/
kg MgSO4
Vehicle1 ml of vehicle solution
(saline)
Animals killed: at 24 hrs

No SCI or Tx (controls)
SCI (no Tx; trauma)
SCI + MPSS
SCI + MgSO4 (magnesium)
SCI + saline (vehicle)
8 rats/group

Intervention Details

No SCI or Tx (sham;
Group I)
SCI (no Tx; Group II)
SCI + MgSO4 (Group
III)
10 rabbits/group

Experimental Groups

TABLE 12: Characteristics of SCI animal studies using MgSO4* (continued)

Results:
Group I vs Group II vs Group III
HR
Before SCT: 244 6.6 vs 242 8.7 vs 243 9.1
60 min after SCT: 247 7.7 vs 268 8.1 vs 257 8.3
MAP (in mm Hg)
Before SCT: 74 2.7 vs 73 2.9 vs 72 2.7
60 min after SCT: 75 2.5 vs 68 2.3 vs 67 2.6
PaO2 (in mm Hg)
Before SCT: 96.39 2.1 vs 97.12 2.3 vs 98.22 1.7
60 min after SCT: 97.33 1.8 vs 95.16 2.5 vs 97.66 2.2
Carbon dioxide (PaCO2; in mm Hg)
Before SCT: 28.40 1.6 vs 26.35 1.7 vs 29.15 1.5
60 min after SCT: 27.45 1.9 vs 27.32 2.1 vs 28.15 1.7
Significant differences in MAP & HR values btwn Group I & the other
groups 60 min after SCT (p <0.05)
Significant increase in Group II (p <0.05) in lactate & MDA levels when
compared w/ Groups I & III
No significant elevation of lactate level in Group III, compared w/ Group I
MgSO4 Tx inhibited the increase in tissue lactate & MDA levels
Safety/Adverse Events/Toxicology: NR
Caspase-3 Activity:
Statistically significant difference btwn the trauma & MgSO4 groups
(p <0.05); MgSO4 Tx decreased caspase-3 activity
The MPSS group was significantly different from the trauma group
(p <0.05); MPSS also prevented an increase in caspase-3 activity
values
Statistically significant differences btwn the MPSS & MgSO4 groups
(p <0.05); MPSS was more effective than MgSO4 in preventing
activity of caspase-3 after SCI
Difference btwn the vehicle & trauma groups was not statistically
significant (p >0.05)
Safety/Adverse Events/Toxicology: NR

Reported Outcomes

(continued)

Fig. 2 in article pro vides more data


for the distribution
of spared spinal
cord matter

Comments

Translational potential of pharmacotherapy for SCI

177

178
SCI + physiological
saline
SCI + MgSO4 (300
mg/kg)
SCI + MgSO4 (600
mg/kg)
10 rats/group

Experimental Groups
Route: IP inj
Timing:
Tx 1 hr post-SCI
Post-Tx SSEP recordings 3 hrs after
inj
Dosage:
MgSO4: 300 mg/kg
MgSO4: 600 mg/kg
Animals killed: all animals were killed
by intracardiac inj of KCl 24 hrs
after trauma

Intervention Details

Reported Outcomes

Comments

* Values represent mean standard deviation unless otherwise indicated. Abbreviations: Admin = administration; HR = heart rate; MAP = mean arterial pressure; MPO = myeloperoxidase; MPSS =
methylprednisolone sodium succinate; NYU = New York University; NZ = New Zealand; SCT = spinal cord trauma; SSEP = somatosensory evoked potential.

Szer et al.,
1999

Animal model:
Albino rats
N = 30
Sex: 100% M
Age: adult
Wt: 210310 g
SCI model:
Extradural clip com pression of cord
at T-9 w/ aneu rysm clip exerting
force of 50 g for
30 sec, causing
paraplegia

Animal & Injury


Models

SSEPs:
Amplitudes of SSEPs were decreased significantly at 30 min after
injury in all 30 animals (p <0.001)
Significant difference btwn postinjury & preinjury amplitudes of P1
(p <0.01) & N1 (p <0.01) for all groups
Reduction in N1 & P1 amplitudes did not recover in the saline-treated
(p >0.05) & low-dose MgSO4 treated (p >0.05) groups
In the high-dose magnesiumtreated groups there was significant
improvement in the amplitudes of P1 (p <0.01) & N1 (p <0.01) when
compared w/ postinjury measurements
Biochemistry Studies:
The difference btwn saline-treated & low-dose magnesiumtreated
groups was not statistically significant (p >0.05)
The MDA content was found to be lower in the high-dose magnesium
treated group than in the saline-treated & low-dose magnesium
treated groups (p <0.01)
Safety/Adverse Events/Toxicology: NR
Expt 1:
Route: IP inj
Analysis of BBB Scores:
Wiseman et al., Animal model:
Sprague-Dawley
SCI + saline (control) Timing:
Expt 1
2009
rats
(n = 13)
All injs occurred w/in 10 min of injury The mean scores were significantly better for the MgSO4 group (6.9
N = 122
SCI + MgSO4 (n = 20) for Expt 1
3.9) than for the control group (4.2 2.0) after 4 wks (p <0.01), but
Sex: 100% F
SCI + MPSS (n = 17) Injs given at 10 min (saline group
there were no significant differences at earlier time points among
Age: adult
SCI + MgSO4 + MPSS only) or at 8, 12, or 24 hrs post-SCI any of the groups (p >0.1)
Wt: 250350 g
(n = 20)
(Tx group) for Expt 2
Expt 2
SCI model:
Expt 2:
Motor scoring performed postop Day The rats in which MgSO4 was administered w/in 8 hrs (BBB score 13.8
NYU impactor mod- SCI + saline (control) 1 & then again each wk for 4 wks
3.7) had significantly better function than rats in the control (8.6
el; laminectomies (n = 13)
Dosage:
5.1), 12-hr (8.1 3.1), or 24-hr (7.9 2.8) groups (p <0.01)
from T910
SCI + MgSO4 (600
Expt 1
The difference in motor improvement started at 2 wks for the rats that
Expt 1: moderate-to- mg/kg) at 8 hrs
Control: 1 ml of normal saline
received MgSO4 w/in 8 hrs of injury, compared w/ the late-admin or
severe injury;
MgSO4: 600 mg/kg
post-SCI (n = 13)
control groups (p <0.01)
10-g wt dropped SCI + MgSO4 (600
MPSS: 30 mg/kg
Histological Findings:
from 25 mm
mg/kg) at 12 hrs
Expt 1
Expt 2
Expt 2: less severe post-SCI (n = 13)
Control: 1 ml of saline
All Tx groups had a significantly greater vol of white matter spared after
injury caused by SCI + MgSO4 (600
MgSO4: 600 mg/kg in 1-ml vol
the injury (MgSO4 32.3%, MPSS 30.3%, & MgSO4 + MPSS 42.3%)
10-g wt dropped mg/kg) at 24 hrs
Animals killed: all animals were killed than did the control group (20.2%) (p <0.05)
from 12.5 mm
post-SCI (n = 13)
by intracardiac inj of KCl 24 hrs
No differences in mean lesion length (2.82 mm MgSO4, 3.30 mm
after trauma
control) could be found btwn the groups; however, the small no.
of specimens & the variance in the control group (2.65.2 mm) were
potential limitations
Safety/Adverse Events/Toxicology:
Animals receiving MgSO4 had a higher complication rate of autophagia
vs saline-treated animals; autophagia began 510 days post-SCI

Authors &
Year

TABLE 12: Characteristics of SCI animal studies using MgSO4* (continued)

C. H. Tator et al.

J Neurosurg: Spine / Volume 17 / September 2012

Translational potential of pharmacotherapy for SCI


the development of secondary injury in SCI. Reported
mechanisms of action include the following: inhibition of
caspase-1 and caspase-3, which are involved in interleukin-1 generation and cell apoptosis, respectively;10 inhibition of the overproduction of nitric oxide and reduction
of the activation of microglia and macrophages;2,31,70,83
mitigation of the effects of matrix metalloproteinases,
which cause various kinds of damage to the CNS;82 and
attenuation of glutamate excitotoxicity.76 Minocycline is a
tetracycline-class drug used for the treatment of bacterial
infections including acne at a maintenance dose of 50
100 mg taken orally once a day. Minocycline may also be
used to treat rheumatoid arthritis.
Systematic Search for Preclinical Studies. Forty-two
articles were identified that evaluated minocycline in animal models of SCI. Nineteen were eligible for full-text
review, 3 of which were excluded because in 2 studies
minocycline was administered prior to injury, and 1 was a
technique paper. Therefore, 16 articles were identified that
met our inclusion criteria.4,21,29,31,43,45,47,55,62,67,70,75,77,80,84,85
Study Characteristics. The study characteristics are
summarized in Table 13; detailed information is available in Table 14. The majority (13) evaluated minocycline in rats, 21,29,31,43,45,47,55,62,67,70,75,77,84 whereas the remaining 3 studies used mice.4,80,85 The number of rodents
per study ranged from 8 to 124. The SCI was induced
in the thoracic region in 14 studies (by weight drop in
9,4,21,29,31,45,70,75,77,84 transection in 3,47,67,85 balloon compression in 1,62 or clip compression in 180) and in the cervical
region (by weight drop) in 2 studies.43,55 The drug was
administered systemically in all cases: intraperitoneal
injection (14), 21,29,43,45,47,55,62,67,70,75,77,80,84,85 intravenous injection (1),4 or intrathecal catheter (1).31 There was a great
deal of variability in dosage and timing of drug delivery
among the studies. Doses of minocycline ranged from
10 to 180 mg/kg; nearly all studies delivered multiple
doses of the drug beginning 02 hours postinjury and
then again every 1224 hours for 16 days. Two studies began minocycline treatment more than 30 days after SCI: Hains et al.31 gave the first injection at 31 days
and continued twice a day for 3 days, and Arnold et al.4
gave the first dose at 6 weeks, followed by daily injections for 2 weeks. Teng et al.75 evaluated 3 different doses
(18 mg/kg, 90 mg/kg, and 180 mg/kg), but the remaining
studies used only 1 dose. Animals were randomized to
treatment groups in 9 of the 16 studies.29,31,43,45,47,62,67,75,84
All but 3 studies62,80,85 evaluated outcomes in a blinded
manner. Eleven studies did not account for all of the animals.4,21,31,45,47,67,70,75,77,80,84
Effectiveness of Therapy. Four studies showed that
minocycline treatment reduced lesion size compared with
saline/vehicle or tetracycline control treatments measured between 7 and 38 days (p < 0.05),21,45,67,80 whereas
2 studies found no difference in cavity area or lesion size
between treatment groups as measured at 28 days.55,75 Saganov and colleagues62 found that both short- and longterm minocycline treatment (1 day and 5 days, respectively) increased white and gray matter sparing rostral but not
caudal to the injury site versus saline treatment 28 days
after injury, whereas Teng and colleagues75 reported sigJ Neurosurg: Spine / Volume 17 / September 2012

nificant white matter sparing in both directions 4 hours


after injury in animals treated with minocycline versus
vehicle (p < 0.03). Two studies reported no difference in
white matter sparing between treatment groups measured
at 17 weeks;4,43 similarly, no difference in gray matter
sparing was found by 1 study at 7 weeks.43
Teng and colleagues75 reported significant protection of
oligodendrocytes as well as the neurons in the ventral horn
in minocycline-treated compared with vehicle-treated rats.
Similarly, these authors found that there were fewer reactive astroglia in animals treated with minocycline versus
vehicle alone. Four studies found that activation of microglia was significantly reduced in minocycline versus saline/
vehicle-treated or untreated animals in tissue examined
between 1 and 5 weeks following injury,29,31,47,70 whereas 1
study saw a reduction that did not reach statistical significance.67 Wells et al.80 reported increases in the preservation
of red nucleus neurons in the rubrospinal tract as well as in
axonal integrity in minocycline-treated animals measured
4 weeks after injury, although these results did not appear to
reach statistical significance. Zang and Cheema85 reported
similar increases at 6 weeks in the number of myelinated
axons surrounding the lesion in minocycline-treated mice
compared with mice treated with vehicle alone. Similarly,
6 studies reported that minocycline treatment resulted in
significantly decreased apoptosis of neurons or oligodendrocytes at and surrounding the lesion site as measured 3
days to 1 week post-SCI, compared with control treatments
(p < 0.05).21,29,45,67,75,84 Two studies reported a decrease in
inflammatory markers between 6 hours and 4 weeks following SCI in minocycline-treated versus saline/vehicletreated or untreated animals,21,45 whereas 1 study reported
no difference between groups at 1 week.4
Minocycline produced variable functional outcomes
following SCI. Ten studies evaluated motor improvement
by using the BBB score; 7 of them reported improved
outcomes with minocycline versus control treatment at
238 days postinjury.21,45,70,75,77,80,84 The remaining 3 studies reported no difference between treatment groups at 5
weeks,31,55,62 and Saganov et al.62 found significant worsening of BBB scores with long-term minocycline treatment (that is, 5 days) compared with placebo measured 14
and 21 days following SCI. The worsening was not seen
with short-term minocycline treatment (1 day). Improved
functional outcomes were also reported following minocycline versus saline/vehicle or no treatment, as measured
by the following: inclined-plane test at 15 weeks, 29,80,84
downward inclined-plane test between 1 and 4 weeks,75
Basso Mouse Scale score (hindlimb motor function
score) at 1 week,4 motor function score at 1 week, 29 and
grid walk test at 5 weeks.84 No differences were found
between treatment groups in the following measures of
functional improvement: upward inclined-plane test between 1 and 4 weeks;75 horizontal ladder test at 6 weeks;43
cylinder rearing test at 6 weeks;43 rotarod balancing at 5
and 10 weeks;85 and bar grab, bar walk, and platform hang
at 5 and 10 weeks.85 Stirling and colleagues67 reported
significant improvements in the coordination of forelimb
and hindlimb movements in minocycline- versus controltreated animals at 7 and 14 days after injury; Yune and
colleagues84 reported similar results.
179

180

animal model (no./group)


mice

18 (6)

Characteristic

rats
injury model
blunt force (wt drop) (cervical)
blunt force (wt drop) (thoracic)
transection (thoracic)
balloon compression (thoracic)
clip compression (thoracic)
timing of intervention postinjury
@ 0 hrs & every 12 hrs there after (length NR)
@ 0 hrs & every 12 hrs for 3
days
@ 0 & 9 hrs & then daily for 3
days
@ 5 min & every 12 hrs for 5
days
@ 0.5, 1, & 24 hrs
@ 0.5, 12, 24, & 36 hrs
@ 0.5, 12, 24, 36, & 48 hrs
@ 1 hr
@ 1 hr & every 12 hrs there after for either 1 day or 5
days
@ 1 hr & every 12 hrs there after for 5 days
@ 1 hr; 2 & 3 days
@ 1 hr; 2, 3, 4, 5, & 6 days
@ 2 hrs, 3/wk (length NR)
@ 31 days (2/day for 3 days)
@ 6 wks; daily for 2 wks (until
8 wks postinjury)

Arnold
& Hagg,
2011

Ha
et al.,
2008

NR

Hains &
Marchand
Waxman, Lee et al., Lee et al.,
et al.,
2006
2010
2003
2009

31 (26) 32 (8) 52 (1018) 43 (910) 95 (313)

Festoff
et al.,
2006

TABLE 13: Summary of minocycline preclinical study characteristics*

NR

42 (817)

NR

8 (4)

Saganov Stirling
Teng Ueno
et al.,
et al.,
Tan et et al., et al.,
2008
2004 al., 2009 2004 2011

43 (910) 35 (1112)

Pinzon
et al.,
2008

124 (10
43)

Wells
et al.,
2003

NR

(continued)

60 (10)

Yune Zang &


et al., Cheema,
2007
2003

C. H. Tator et al.

J Neurosurg: Spine / Volume 17 / September 2012

Characteristic

J Neurosurg: Spine / Volume 17 / September 2012

* IT = intrathecal; NR = not reported.

% animals accounted for

route of intervention
IV inj (tail vein)
IP
IP + subcutaneous
IT catheter
dosage
10 mg/kg
18 mg/kg
30 mg/kg
40 mg/kg
50 mg/kg
50 mg/kg 1st & 2nd dose,
then 25 mg/kg thereafter
90 mg/kg
90 mg/kg 1st dose, then 45
mg/kg thereafter
180 mg/kg
100 mg/dose
200 mg/day
>1 dose evaluated?
yes
no
control groups
SCI (saline/vehicle)
SCI (no Tx)
no SCI
independent or blind assessment
yes
no
NR
animals randomized to Tx groups
yes
no
NR
X

88% (46
of 52)

100% (43 68% (65


of 43)
of 95)

NR

Hains &
Marchand
Waxman, Lee et al., Lee et al.,
et al.,
2006
2010
2003
2009

65% (20 100%


of 31) (32 of
32)

NR

Ha
et al.,
2008

Festoff
et al.,
2006

Arnold
& Hagg,
2011

TABLE 13: Summary of minocycline preclinical study characteristics* (continued)

100% (45
of 45)

Pinzon
et al.,
2008

100% (35
of 35)

NR

NR

NR

NR

Saganov Stirling
Teng Ueno
et al.,
et al.,
Tan et et al., et al.,
2008
2004 al., 2009 2004 2011

NR

Wells
et al.,
2003

NR

100% (60
of 60)

Yune Zang &


et al., Cheema,
2007
2003

Translational potential of pharmacotherapy for SCI

181

182
SCI + minocycline
SCI + tetracycline
SCI + vehicle (sterile
saline)
Prior to SCI, rats under went adaptation & train ing for 1 wk on the BBB
locomotor rating

Animal model:
Sprague-Dawley rats
N = 31
Sex: 100% F
Age: adult
Wt: 300325 g
SCI model:
Wt drop method; mod erate injury at T-9
spine level (10 g
2.5 cm; 25 g/cm
force)

Route: IP inj
Dosage & timing:
Motor function & histology
Minocycline 30 mg/kg at 0.5, 1, & 24
hrs post-SCI (n = 4)
Minocycline 90 mg/kg at 0.5 (n = 4), 1
(n = 4), or 24 (n = 2) hrs post-SCI
Tetracycline 30 mg/kg 1 hr post-SCI
(n = 6)
Caspase substrate cleavage
Minocycline 90 mg/kg at 1 hr post-SCI
(n = 3)
Tetracycline 30 mg/kg at 1 hr post-SCI
(n = 3)
qRT-PCR
Minocycline 90 mg/kg at 1 hr post-SCI
(n = 4)
Tetracycline 30 mg/kg at 1 hr post-SCI
(n = 3)
Animals killed:
28 days (motor function & histology)
3 days (caspase substrate cleavage)
1 day (qRT-PCR)

Route: IV inj through the tail veins


Timing: 6 wks post-SCI (chronic phase)
Dosage (Txs lasted 2 wks):
Vehicle: 0.1 ml saline daily
Minocycline: 200 g in saline daily
CD25: 100 g in saline every 3rd day
Animals killed: all, 1 wk after Txs were
terminated (to see if there is a
potential washout effect)

SCI + vehicle (saline)


SCI + minocycline
SCI + rat monoclonal anti
mouse CD25 antibody
6/group
(mice were assigned to
their Tx groups to
achieve similar displace ment & 6-wk BMS
scores, & then treated)

Arnold & Hagg, Animal model:


C57BL/6 mice
2011
N = 18
Sex: 100% F
Age: 8 wks
Wt: 1825 g
SCI model:
Wt drop method;
moderate injury at
T-9 spine level
(impact of 50 kdyn)

Festoff et al.,
2006

Intervention Details

Experimental Groups

Authors & Year Animal & Injury Models

TABLE 14: Characteristics of SCI animal studies using minocycline*


Comments

J Neurosurg: Spine / Volume 17 / September 2012

(continued)

Independent/blinded
BMS Score (hindlimb motor function score):
Vehicle vs minocycline vs CD25
assessment of
outcomes: yes
1 wk: 6.8 vs 7.2 0.5 vs 6.3
2 wks: 5.8 vs 7.7 0.4 vs 7.1 0.5
1 wk post-Tx (washout phase): 6.1 vs 7.1 0.3 vs 6.5 0.5
At the maximal BMS levels (w/ 2 wks of Tx), the minocycline &
CD25 antibodytreated mice had significantly higher values
than the vehicle-treated mice (p <0.01 & 0.05, respectively)
Histology:
White Matter Sparing: Vehicle vs Minocycline vs CD25
29 2% vs 27 2% vs 29 3%
No difference btwn Tx groups
Inflammatory Markers
CD68- & CD45-positive areas were not significantly different
among the 3 groups (only data in fig.)
Only minocycline-treated mice had a significant & substantial
correlation (inverse) btwn the total CD68 (R 2 = 0.884; p =
0.017) or CD45 (R 2 = 0.872; p = 0.02) area & 9-wk BMS
scores; amount of epicenter white matter did not correlate w/
BMS scores at 59 wks postinjury (data not provided)
BBB Score:
Multiple Doses of Minocycline: SCI + Minocycline (30 mg/kg at
0.5, 1, & 24 hrs post-SCI) vs SCI + Tetracycline
Day 2: 4.25 1.2 vs 1.83 0.54 (p <0.05)
Days 728: from 10.8 1.3 to 14.6 0.62 vs from 4.94 0.17 to
8.33 0.66 (p <0.001)
Note: a plateau of functional recovery was reached at 21 days
post-SCI in both minocycline & tetracycline groups
Timing of Minocycline: SCI + Minocycline (90 mg/kg at 0.5, 1, or
24 hrs post-SCI) vs SCI + Tetracycline
Single-dose Tx w/ minocyclineindependent of when adminis tered after SCIshowed significant improvement in function al recovery from 7 to 28 days compared w/ tetracycline
(p <0.05 on Day 7; p <0.001 from Day 10 to 28)
Of interest for human SCI, w/in the first 24 hrs after SCI, the time
of admin of minocycline (0.5, 1, or 24 hrs) did not significantly
influence functional recovery

Reported Outcomes

C. H. Tator et al.

Festoff et al.,
2006
(continued)

Authors & Year Animal & Injury Models

Experimental Groups

Intervention Details

TABLE 14: Characteristics of SCI animal studies using minocycline* (continued)

Histology:
Neuronal Profiles/Tissue Sparing
Minocycline Tx resulted in significant sparing of tissue & reduc tion in tissue damage & cavitation at the lesion site compared
w/ tetracycline & saline controls
much greater preservation of the gray matter, even at the
epicenter
segments caudal to the site of the injury appeared relatively
normal
Spinal cord cross-sections from the minocycline group showed
a dramatic reduction in necrosis, gliosis, & cavitation at T-9
epicenter compared w/ those from the tetracycline group,
which were almost entirely destroyed
much smaller macrophage-containing cavities w/in the
posterior columns
much less chromatolysis rostral to the level of injury
Cavity Size
Significant tissue sparing (p <0.02) was seen in rats treated w/
minocycline (93 6%) compared to tetracycline (82 4%)
Furthermore, a pairwise multiple comparison procedure found
significant (p <0.04) improvement for minocycline compared
w/ tetracycline (control) Tx
Apoptosis & Caspase-3
Minocycline significantly reduced apoptosis in ventral spinal
cord motor neurons in both lesioned & adjacent segments (w/
all minocycline admins compared to tetracycline)
Minocycline-treated rat spinal cords displayed greatly reduced
caspase-3 activation, w/ a greatly increased no. of preserved
motor neurons compared w/ tetracycline-treated rats, which
showed significant caspase-3 activation w/ numerous apop totic neurons
Caspase-3 mRNA was reduced 53% (p <0.04) by minocycline;
no reduction was seen in the tetracycline group; this novel
finding represents the first quantification by real-time tech niques (qRT-PCR) of caspase-3 mRNA after minocycline Tx
There was a 74% reduction in caspase-3-mediated cleavage
overall in minocycline-treated rats compared to tetracycline treated controls (p <0.05); surprisingly, the amount of
caspase-3 cleavage product was less at the epicenter than in
both rostral & caudal segments of the lesioned spinal cord

Reported Outcomes

(continued)

Comments

Translational potential of pharmacotherapy for SCI

J Neurosurg: Spine / Volume 17 / September 2012

183

184

Ha et al., 2008

Festoff et al.,
2006
(continued)

Animal model:
Sprague-Dawley rats
N = 32
Sex: 100% M
Age: adult
Wt: 300350 g
SCI model:
Wt drop method; mod erate injury at T-9
spine level (10 g
2.5 cm; 25 g/cm
force)

Authors & Year Animal & Injury Models

SCI only (controls)


SCI + methylprednisolone
(MP)
SCI + minocycline (MC)
SCI + pregabalin (GP)
8/group

Experimental Groups

Route: IP inj
Dosage & timing:
MP30 mg/kg at 30 min, 12 hrs, & 24
hrs post-SCI
MC30 mg/kg at 30 min, 12 hrs, 24
hrs, 36 hrs, & 48 hrs post-SCI
GP30 mg/kg at 30 min & every 12
hrs for the 1st 2 days post-SCI
Animals killed: all, 1 wk post-SCI

Intervention Details

TABLE 14: Characteristics of SCI animal studies using minocycline* (continued)

Antineuroinflammatory Effects:
Injury-induced microglial activation was substantially reduced in
minocycline-treated rats compared w/ tetracycline-treated
rats, in which microglial profiles in both dorsal & ventral
gray matter, & to a lesser extent in white matter, were strik ingly increased compared to normal
TNF- mRNA levels assessed by real-time qRT-PCR were
significantly reduced in minocycline-treated rats compared w/
tetracycline-treated rats at 24 hrs after SCI (2.8 0.34 vs 3.4
0.2; p <0.05)
Motor Function Score (Gale et al.):
Control vs MP vs MC vs GP
1 wk: 1.0 vs 2.42 0.5 vs 2.62 0.5 vs 3.87 0.8
Mean scores of all 4 groups differed from each other (p = 0.01)
GP score was superior to the MP & MC groups (p <0.05)
Inclined-Plane Test ():
Control vs MP vs MC vs GP
1 wk: 22.5 2.6 vs 30.6 3.2 vs 32.5 1.7 vs 37.2 5.5
Mean scores of all 4 groups differed from each other (p = 0.01)
GP score was superior to the MP & MC groups (p <0.05)
TUNEL-Positive Cells (neuron & glial cell death):
Control vs MP vs MC vs GP
Mean nos. of TUNEL-positive cells: 63.5 7.4 vs 53.6 4.0 vs
44.2 3.9 vs 36.5 3.6
p <0.05 for control group vs other 3 groups
p <0.05 for GP group vs MP & MC groups
Anti-OX-42 Positive Cells (activation of microglia):
Control vs MP vs MC vs GP
Mean nos.: 29.8 3.9 vs 22.7 4.1 vs 21.0 3.9 vs 17.8 4.3
p <0.01 for control group vs other 3 groups
p <0.05 for GP group vs MP & MC groups

Reported Outcomes

(continued)

GP was main focus


of study; no com parison made
btwn MC & MP or
MC & controls

Comments

C. H. Tator et al.

J Neurosurg: Spine / Volume 17 / September 2012

Hains & Wax man, 2006

Animal model:
Sprague-Dawley rats
N = 52
Sex: 100% M
Age: adult
Wt: 200225 g
SCI model:
Wt drop method; mod erate injury at T-9
spine level (10 g
2.5 cm; 25 g/cm
force)

Authors & Year Animal & Injury Models

Intervention Details
Route: IT catheterization (on Day 28)
Timing: Day 31 post-SCI
Dosage (2 daily for 3 days):
Minocycline 100 g/5 l (in artificial
CSF)
Vehicle (artificial CSF)
Animals killed:
Day 33 (for histology; 6 rats per group
[sham, SCI + vehicle, SCI + minocycline])
Day 34 (electrophysiological exams; 4
rats per the 3 groups)

Experimental Groups
SCI + vehicle (n = 18)
SCI + minocycline (n = 18)
Sham surgery (n = 10;
laminectomy & place ment into vertebral clips
of the impactor)

TABLE 14: Characteristics of SCI animal studies using minocycline* (continued)

Behavioral Testing:
BBB Scores (locomotor): SCI + Vehicle vs SCI + Minocycline
Day 33 (3 days postinjection): 10.1 2.0 vs 11.9 2.6
unchanged from Day 30; SCI group mean 10.4 2.1
Day 35: no significant differences/changes
Mechanical Paw Withdrawal Thresholds
After SCI, group mean: 3.9 1.5 gsignificant decrease
compared w/ intact rats (p <0.05)
Minocycline resulted in immediate increase: range 7.9 1.5 g to
14.2 2.0 g
Cessation of minocycline = return to predrug levels: 3.9 1.8 g
Thermal Paw Withdrawal Latencies
After SCI, group mean: 5.8 2.1 secsignificant decrease
compared w/ intact rats (p <0.05)
Minocycline resulted in immediate increase: 11.1 1.7 sec
Cessation of minocycline = return to predrug levels: 5.6 0.9
secintegral analysis revealed that minocycline resulted in
a significantly (p <0.05) more robust modulation of mechani cal nociception compared w/ thermal nociception
Immunocytochemistry:
GFAP (astroglia activation): Intact vs SCI + Vehicle vs SCI +
Minocycline
Proportional area of GFAP signal: 24% vs 33% vs 32%
Significantly (p <0.05) elevated levels of astroglial activation in
SCI + vehicle & SCI + minocycline groups, compared w/
intact animals
P-p38 (microglia activation): Intact vs SCI + Vehicle vs SCI +
Minocycline
Number of P-p38+ cells: 15 vs 90 vs 30
Significantly (p <0.05) higher in the SCI groups when compared
w/ intact rats
SCI + minocycline resulted in a significant reduction in no. of
positive cells compared w/ SCI + vehicle (p <0.05).
OX-42 Antibodies (microglia activation): Intact vs SCI + Vehicle
vs SCI + Minocycline
Resting/activated states (population process length): 0.9/0.1 vs
0.1/0.9 vs 0.7/0.3
SCI groups demonstrated a significant shift from resting to
activated forms compared w/ intact rats (p <0.01)
SCI + minocycline resulted in significant reduction in activated
forms compared w/ SCI + vehicle (p <0.01)

Reported Outcomes

J Neurosurg: Spine / Volume 17 / September 2012

185

(continued)

OX-42 = shift from


resting to activated
forms of microglia
determined by
counting the no. of
cells w/ processes
longer/shorter than
the soma diameter

P-p38 = not a specific


marker of, but has
been associated
w/, microglial
activation

GFAP = marker for


normal as well as
reactive astroglia

All behavioral testing


was performed by
a blinded observer

Comments

Translational potential of pharmacotherapy for SCI

Lee et al., 2010 Animal model:


Sprague-Dawley rats
N = 43
Sex: NR
Age: NR
Wt: 300550 g
SCI model:
Cervical C-5 unilat con tusion model (Infinite
Horizon impactor);
following C-5 hemilaminectomy

Authors & Year Animal & Injury Models


Minocycline (n = 9)
Simvastatin
For 7 days (n = 10)
For 42 days (n = 10)
SCI + saline (control,
n = 10)

Experimental Groups
Route: IP & subcutaneous inj
Timing: 1 hr post-SCI
Dosage & timing:
Minocycline: IP inj of 90 mg/kg/day for
3 days, followed by saline for 39
days, & subcutaneous inj of simvas tatin vehicle for 42 days
Simvastatin (7 days): IP inj of saline for
42 days & subcutaneous inj of
simvastatin: 20 mg/kg/day for 3 days
& 5 mg/kg/day for 4 days, followed
by simvastatin vehicle for 35 days
Simvastatin (42 days): IP inj of saline
for 42 days & subcutaneous inj of
simvastatin: 20 mg/kg/day for 3 days
& 5 mg/kg/day for 39 days
Control: IP inj of saline (0.7 ml/day) &
subcutaneous inj of simvastatin
vehicle (0.35 ml/day) for 42 days
Animals killed: all animals killed at 7
wks post-SCI (1 wk after antero grade tracing)

Intervention Details

TABLE 14: Characteristics of SCI animal studies using minocycline* (continued)

Horizontal Ladder Test:


% ipsilat forelimb error, 6 wks post-SCI
Control: 15.49 2.85%
Minocycline: 14.85 1.96%
Simvastatin 7 days: 19.24 2.71%
Simvastatin 42 days: 17.46 2.79%
p = NS for all group comparisons
Cylinder Rearing Test:
% ipsilat forelimb usage, 6 wks post-SCI
Control: 7.14 3.76%
Minocycline: 12.45 5.43%
Simvastatin 7 days: 7.95 2.49%
Simvastatin 42 days: 12.93 5.53%
p = NS for all group comparisons
Modified Montoya Staircase (food pellet retrieval):
Wk 2: simvastatin 7 days group significantly worse than control
& minocycline groups; p = 0.002
Wk 6: simvastatin 42 days group significantly worse than control
& minocycline groups; p = 0.002
Grooming Test:
Grooming score at 6 wks post-SCI
Simvastatin 42 days: 2.78 0.22
Control: 1.90 0.18
Minocycline: 2.00 0.22
Simvastatin 7 days: 2.00 0.32
p = 0.003 for simvastatin 42 days vs control, minocycline, &
simvastatin 7 days groups
Sensory Testing for Mechanical Allodynia:
At 6 wks, no difference in force for ipsilat forepaw withdrawal
was seen btwn the 4 groups
No suggestion of mechanical allodynia & hypersensitivity was
observed
White & Gray Matter Sparing (7 wks post-SCI):
Significantly greater white matter sparing at c400 and c800 (seri al cross-sections) was seen in simvastatin 7 days vs minocy cline; 30% vs 15% & 35% vs 25%, respectively; p <0.0001
No difference seen btwn groups in gray matter sparing
Cumulative extent of white or gray matter sparing did not differ
significantly btwn the 4 groups
CST & RST Sprouting (7 wks post-SCI):
No statistically significant differences btwn the 4 groups
Simvastatin & Minocycline Bioactivity:
Plasma minocycline levels 2 hrs after the 3rd dose were, on
average, 40.34 6.70 mg/L
At 3 days & 2 wks, both simvastatin w/ or w/o injury had lower
high-density lipoprotein/low-density lipoprotein ratio change
compared to sham & injury-only group (p <0.001 and p = 0.003)

Reported Outcomes

186
(continued)

4 animals excluded
due to peak injury
forces that ex ceeded 165 kdyn

Comments

C. H. Tator et al.

J Neurosurg: Spine / Volume 17 / September 2012

Reported Outcomes
BBB Score:
Scores were significantly higher in minocycline-treated rats 24
38 days after SCI compared to those in vehicle-treated rats
At 38 days post-SCI: minocycline 18 0.7 & vehicle 15 0.5
(p <0.01)
Tissue Necrosis:
Lesion size increased progressively in minocycline- & vehicle treated spinal cords; however, 14 days postinjury there was
a marked decrease in lesion size in minocycline-treated cords
compared to that of vehicle
Lesion size was significantly reduced at 28 (p <0.05) & 38 days
(p <0.01) post-SCI by Tx w/ minocycline compared to that of
vehicle
38 days: 1.6 0.5 mm2 vs 3.5 0.8 mm2
Apoptotic Cell Death:
Minocycline Tx significantly reduced the no. of TUNEL-positive
cells 24 hrs post-SCI compared to vehicle: 84 29 & 185
30 (p <0.001)
DNA gel electrophoresis revealed a marked decrease in DNA
laddering following minocycline Tx
Caspase-3 Activity:
Minocycline Tx significantly reduced specific caspase-3 activity
4 hrs post-SCI compared to that of vehicle control (p <0.01):
7 pg/min/mg vs 13 pg/min/mg
Minocycline-treated rats still showed significant increase com pared to sham-treated rats (p <0.05): 7 pg/min/mg vs 5 pg/
min/mg
IL-10 & TNF- Expression:
By 6 hrs after SCI, minocycline Tx significantly increased the
mRNA level of IL-10, a potent antiinflammatory cytokine,
compared to that of vehicle control (p <0.05)
By contrast, minocycline Tx significantly decreased the mRNA
level of TNF-, a proinflammatory cytokine, compared to that
of vehicle control (p <0.05)
Minocycline had no significant effect on the mRNA levels of
IL-1, TGF, & IL-6 compared to those of vehicle control

Intervention Details
Route: IP inj
Timing: 0 hrs post-SCI
Dosage & timing:
Minocycline: 90 mg/kg at 0 hrs post SCI, then 45 mg/kg every 12 hrs
Vehicle: sterile saline at same times as
minocycline group
Animals killed:
Days 14, 21, 28, & 38 post-SCI (lesion
area analysis)
24 hrs post-SCI (TUNEL staining,
caspase-3 activity)
0, 1, 6, 12, & 24 hrs post-SCI (cytokine
expression)

Experimental Groups
SCI + minocycline
SCI + vehicle (sterile
saline)
Sham group (T910 lami nectomy w/o SCI)
Normal group
Lesion area analysis: n = 3
for vehicle & minocy cline tested at 14, 21,
28, & 38 days; total 24
TUNEL staining: n = 3 for
vehicle & minocycline;
total 6
Caspase-3 activity: n = 3
for sham, vehicle, &
minocycline; total 9
Cytokine expression: n = 3
for normal, sham, vehi cle, & minocycline;
total 30
Behavioral testing: n = 13
for vehicle & minocy cline; total 26

Authors & Year Animal & Injury Models

Lee et al., 2003 Animal model:


Sprague-Dawley rats
N = 95
Sex: 100% M
Age: NR
Wt: 230250 g
SCI model:
Wt drop method; mod erate injury at T-9
spine level (10 g
12.5 mm)

TABLE 14: Characteristics of SCI animal studies using minocycline* (continued)

(continued)

Comments

(continued)

Translational potential of pharmacotherapy for SCI

J Neurosurg: Spine / Volume 17 / September 2012

187

188

Marchand et
al., 2009

Animal model:
Wistar rats
N = NR
Sex: 100% M
Age: adult
Wt: 220250 g
SCI model:
Hemisection at T-13

Authors & Year Animal & Injury Models


SCI + saline
SCI + minocycline
Sham + saline
Sham + minocycline
SCI + etanercept (immedi ate Tx)
SCI + etanercept (delayed
Tx)

Experimental Groups
Route:
Minocycline or sham: IP inj
Etanercept: IT (minipump & subdural
catheter)
Dosage & timing:
Minocycline: 40 mg/kg 2/day for 2
days starting 30 min post-SCI
Sham + saline: 40 mg/kg 2/day for 2
days starting 30 min post-SCI
Etanercept: 50 g/day for 7 days (350
g total) starting 30 min post-SCI
(immediate Tx)
Etanercept: 50 g/day for 7 days (350
g total) starting 14 days post-SCI
(delayed Tx)
Animals killed:
Days 7 & 14 (n = 4 each from SCI &
sham groups for immunohistochemi cal analysis)
Day 14 or 28 (immediate etanercept
group)
Day 21 (delayed etanercept group)

Intervention Details

TABLE 14: Characteristics of SCI animal studies using minocycline* (continued)

Only reported out comes for minocy cline vs sham (not


for immediate
etanercept vs de layed etanercept
Tx)

Mechanical Allodynia (mechanical threshold):


SCI + Minocycline vs SCI + Saline vs Sham + Minocycline vs
Sham + Saline
Baseline
Ipsilat paw: 30 g vs 31 g vs 29 g vs 32 g
Contralat paw: 29 g vs 32 g vs 30 g vs 26 g
Day 7
Ipsilat paw: 31 g vs 21 g vs 31 g vs 33 g
Contralat paw: 31 g vs 24 g vs 30 g vs 31 g
Day 14
Ipsilat paw: 26 g vs 19 g vs 31 g vs 27 g
Contralat paw: 27 g vs 20 g vs 25 g vs 27 g
SCI + minocycline significantly (p <0.05) decreased the develop ment of mechanical allodynia in both ispilat & contralat paws
compared to SCI + saline for at least 14 days post-SCI
Thermal Hyperalgesia (withdrawal threshold):
SCI + Minocycline vs SCI + Saline vs Sham + Minocycline vs
Sham + Saline
Baseline
Ipsilat paw: 10 sec vs 11 sec vs 13 sec vs 10 sec
Contralat paw: 11 sec vs 10 sec vs 11 sec vs 11 sec
Day 7
Ipsilat paw: 11 sec vs 8 sec vs 12 sec vs 10 sec
Contralat paw: 14 sec vs 9 sec vs 12 sec vs 10 sec
Day 14
Ipsilat paw: 12 sec vs 8 sec vs 12 sec vs 10 sec
Contralat paw: 11 sec vs 8 sec vs 10 sec vs 10 sec
SCI + minocycline significantly (p <0.05) decreased the devel opment of thermal hyperalgesia in both ispilat & contralat
paws compared to SCI + saline for at least 14 days post-SCI
OX-42 Expression:
Day 7: SCI + minocycline showed significantly less (p <0.05)
expression bilat; by 46.6 1.8% & 34.8 3.9% for ipsilat &
contralat sides compared to SCI + saline
Day 14: SCI + minocycline showed significantly less (p <0.05)
expression bilat; by 32.8 1.4% & 33.1 3.1% for ipsilat &
contralat sides compared to SCI + saline
Expression in the SCI + minocycline group was still significantly
higher than in the sham group (p <0.05)
c-FosPositive Cells:
Day 7: SCI + minocycline showed significantly fewer positive
cells compared to SCI + saline; 44.6 16.3%
Day 14: SCI + minocycline showed significantly fewer positive
cells compared to SCI + saline; 36 14.7%

(continued)

Comments

Reported Outcomes

C. H. Tator et al.

J Neurosurg: Spine / Volume 17 / September 2012

Same as Lee (2003), w/


the following differences:
N = 45
Wt: NR

Animal model:
Wistar rats
N = 35
Sex: 100% M
Age: adult
Wt: 300330 g
SCI model:
Balloon compression

Pinzon et al.,
2008

Saganov et
al., 2008

Authors & Year Animal & Injury Models


Route: IP & IV injs
Timing: 0 hrs post-SCI
Dosage & timing:
Minocycline: 90 mg/kg IP at 0 hrs post SCI, then 45 mg/kg at 12 & 24 hrs
Minocycline: 90 mg/kg IV via jugular
at 0 hrs post-SCI, then 45 mg/kg at
12 & 24 hrs
Vehicle: saline IP at same times as
minocycline IP group
Vehicle: saline IV at same times as
minocycline IV group
Animals killed: all animals at 6 wks
post-SCI

SCI + minocycline (IP)


SCI + minocycline (IV)
SCI + vehicle (saline IP)
SCI + vehicle (saline IV)
Histology: n = 45
Behavioral testing: n = 45

Reported Outcomes

BBB Score:
2-Way Repeated-Measures ANOVA:
group (F3, 41 = 0.734, p = NS)
group time (F21, 269 = 0.757, p = NS)
time (F7, 269 = 550.37, p <0.001)
No significant difference was seen at any point in time among
the 4 groups
The highest average score at the end of the study was for the
minocycline group (12.1 0.18), & the lowest score was for
the saline control group (11.8 0.2)
Subscore Analysis:
group (F3, 41 = 1.51, p = NS)
group time (F21, 269 = 0.862, p = NS)
time (F7, 269 = 550.37, p <0.001)
Histology (data are from 20-mm segmental cord sections):
Spared tissue area did not differ significantly by group (F3, 39 =
1.04, p = NS)
The greatest amount of spared tissue was obtained in Group 3
(control IP): 35.35 2.17 mm2
The total cavity area was also not significantly different btwn
groups (F3, 39 = 0.33, p = NS)
Group 4 (control IV) had the smallest area of cavitation (1.19
0.36 mm2)
SCI + saline (n = 11)
Route: IP inj
BBB Score (locomotor recovery):
SCI + minocycline (n = 24; Dosage & timing:
Placebo vs SCI + Minocycline Short-Term vs SCI + Minocycline
n = 12 in both the short- SCI + minocycline: 90 mg/kg (3 g/ml in Long-Term
& long-term dose
normal saline) at 1 hr post-SCI, then Baseline: 21 for all groups
groups)
45 mg/kg every 12 hrs as follows:
Day 1: 0 for all groups
Group 1 (short-term): 1 day (total dose Day 7: 5 vs 5 vs 6
180 mg/kg)
Day 14: 11 vs 10 vs 8
Group 2 (long-term): 5 days (total dose Day 21: 13 vs 13 vs 10
450 mg/kg)
Day 28: 14 vs 14 vs 13
Animals killed: all, on Day 28 post-SCI No significant difference btwn placebo & short-term minocycline
Tx at any time point
Significant worsening seen w/ long-term minocycline Tx vs
placebo on Days 14 & 21 (p <0.05)
Rostrocaudal Distribution of Spared Spinal Cord Matter at Day
28 post-SCI:
Both long- & short-term minocycline Tx increased sparing of
gray & white matter, compared w/ placebo, which was
significant (p <0.05) in the rostral direction beginning 2 mm
from lesion epicenter
Neither Tx reduced gray & white matter damage caudal to lesion
epicenter

Intervention Details

Experimental Groups

TABLE 14: Characteristics of SCI animal studies using minocycline* (continued)

J Neurosurg: Spine / Volume 17 / September 2012


(continued)

Authors replicated
experimental con ditions described
by Lee et al.
(2003), using the
same injury model,
injury severity, &
behavioral out come measures
to corroborate the
use of minocycline
as a neuroprotec tive agent

Comments

Translational potential of pharmacotherapy for SCI

189

190

Stirling et al.,
2004

Animal model:
Wistar rats
N = NR
Sex: NR
Age: adult
Wt: NR
SCI model:
C78 dorsal column
transection

Authors & Year Animal & Injury Models


SCI + saline
SCI + minocycline

Experimental Groups
Route: IP inj
Timing: 30 min post-SCI
Dosage:
Minocycline: 50 mg/kg (in 1.0 ml of
saline) 2/day for 2 days
Saline: dosage NR, 2/day for 2 days
Animals killed: Days 7 & 14 post-SCI

Intervention Details

TABLE 14: Characteristics of SCI animal studies using minocycline* (continued)

Active Caspase-3Mediated Oligodendrocyte Death (mean no.


of apoptotic profiles):
SCI + Minocycline vs SCI + Saline
Day 7:
Proximal AST segment: 32.7 2.7 vs 56.1 3.9 (p <0.001)
Distal AST segment: 47.9 4.0 vs 78.7 3.5 (p <0.001)
Day 14:
Proximal AST segment: 19.8 3.3 vs 37.6 3.8 (p <0.01)
Distal AST segment: 36.8 4.1 vs 58.2 5.0 (p <0.01)
Results suggest that minocycline Tx continued for 48 hrs postin jury is sufficient to reduce secondary cell death by ~39%
(distal) & 42% (proximal) at 7 days, & by 37% (distal) & 47%
(proximal) at 14 days postinjury
Mean no. of Apoptotic Oligodendrocytes:
SCI + Minocycline vs SCI + Saline
Day 7:
Proximal AST segment: 3.9 0.5 vs 6.2 0.6 (p <0.01)
Distal AST segment: 4.8 0.8 vs 7.9 0.8 (p <0.01)
Day 14:
Proximal AST segment: 4.6 0.7 vs 7.4 0.9 (p <0.05)
Distal AST segment: not significantly different btwn groups
(data NR)
ED1 Density (ED1-positive microglia/macrophage) w/in the
Degenerating Distal Segments of the AST & CST:
SCI + Minocycline vs SCI + Saline
AST100 = 1.6 0.5% vs 6.0 1.1% (p <0.01)
AST300 = 2.0 0.3% vs 4.7 0.7% (p <0.01)
CST = 5.1 2.1% vs 11.8 1.4% (p <0.05)
Trend toward reduction of ED1 density w/in the proximal seg ment of the AST closest to CST & w/in proximal CST in SCI +
minocycline group, but this failed to reach significance
ED1 Density w/in Proximal Stump of CST (immediately adjacent
to lesion):
SCI + Minocycline vs SCI + Saline
8.2 1.6% vs 19.4 3.8% (p <0.01)
Proximal CST Dieback (assessed by measuring the distance
btwn the closest, intact, anterogradely traced CST fibers &
the center of the lesion site):
SCI + Minocycline vs SCI + Saline
Day 7: 598.4 48.6 m vs 877.1 47.7 m
Day 14: 751.2 58.0 m vs 1120.2 80.5 m
On average, minocycline Tx reduced the distance of the leading
CST axons from the injury site by ~32% & 33% at 7 & 14
days, respectively

Reported Outcomes

J Neurosurg: Spine / Volume 17 / September 2012

(continued)

Distal AST = rostral


to lesion site
Distal CST = caudal
to lesion site
Proximal AST = cau dal to lesion site
Proximal CST = ros tral to lesion site
AST100 = measured
from 100 m
dorsal to CST
AST300 = measured
from 300 m
dorsal to CST
Proximal CST die back = degenera tion of proximal
axon stumps that
occurs during the
first few wks af ter SCI injury; may
have implications
for regeneration &
sprouting after
injury
Lesion area = region
demarcated by
activated astrocytes

Comments

C. H. Tator et al.

Tan et al., 2009 Animal model:


Sprague-Dawley rats
N = 42
Sex: 100% M
Age: adult
Wt: 200225 g
SCI model:
Wt drop method; mod erate injury at T-9
spine level (10 g
2.5 cm; 25 g/cm
force)

Stirling et al.,
2004
(continued)

Authors & Year Animal & Injury Models

SCI + minocycline (n = 17)


SCI + vehicle (saline)
(n = 17)
Sham surgery (n = 8)

Experimental Groups

Reported Outcomes

Lesion Size:
Minocycline significantly reduced lesion area at both 7 & 14
days postinjury compared w/ saline (p <0.01 & p <0.001);
data provided in figure only
Footprint Analysis/Functional Outcome (assessed for perturba tions in limb coordination, angle of rotation, toe spread):
SCI + Minocycline vs SCI + Saline
forelimbhindlimb coordination was significantly improved in
minocycline- vs saline-treated rats at both 7 & 14 days
postinjury (p <0.05); data provided in figure only
injury-induced hindlimb angle of rotation was reduced in minocy cline- vs saline-treated rats at both 7 & 14 days postinjury
(p <0.05); data provided in figure only
injury-induced hindlimb toe spread was attenuated in minocy cline- vs saline-treated rats at Day 7; 0.07 0.02 vs 0.13
0.01 (p <0.01), but no differences btwn groups at Day 14
Microglial Activation (Cd11b/c field area [arbitrary units]):
Route: IP inj
SCI + Minocycline vs SCI + Vehicle
Timing: 5 mins post-SCI
Baseline: 135 vs 140
Dosage:
1 wk: 150 vs 180
Minocycline: 90 mg/kg (in 0.3 ml sa2 wks: 150 vs 200 (p <0.05)
line), then 45 mg/kg (in 0.3 ml sa3 wks: 159 vs 210 (p <0.05)
line) 2/day for additional 5 days
4 wks: 157.6 12.2 vs 216.6 24.7 (p = 0.001)
Saline: 0.3 ml (0.9% normal saline),
Dorsal Horn Unit Recordings:
then 2/day for additional 5 days
Sham vs SCI Groups
Animals killed:
Weekly for 4 wks beginning Wk 1 post- Phasic brush: 7.4 4.4 Hz vs 43.0 15.3 Hz
SCI (immunohistochemistry: intact, Pressure: 6.5 5.0 Hz vs 32.4 9.2 Hz
Pinch: 7.9 5.3 Hz vs 38.8 22.9 Hz
n = 3; SCI + vehicle, n = 12; SCI +
0.39 g von Frey filament: 4.8 4.7 Hz vs 16.5 5.8 Hz
minocycline, n = 12)
Day 28 post-SCI (behavioral testing; 5 1.01 g von Frey filament: 6.5 5.7 Hz vs 22.8 9.1 Hz
20.8 g von Frey filament: 8.6 7.1 Hz vs 31.5 15.4 Hz
animals/group)
SCI displayed increased peripherally evoked unit responses for
all stimuli; p <0.001 for all
SCI + Minocycline vs SCI + Vehicle
Phasic brush: 9.9 6.1 Hz vs 43.0 15.3 Hz
Pressure: 9.6 5.0 Hz vs 32.4 9.2 Hz
Pinch: 11.8 5.3 Hz vs 38.8 22.9 Hz
0.39 g von Frey filament: 4.3 2.6 Hz vs 16.5 5.8 Hz
1.01 g von Frey filament: 5.6 3.3 Hz vs 22.8 9.1 Hz
20.8 g von Frey filament: 12.0 4.6 Hz vs 31.5 15.4 Hz
Early minocycline Tx significantly reduced evoked responses to
all peripheral stimuli recorded 30 days after SCI (p <0.001
for all)
No significant differences were observed btwn sham & SCI +
minocycline

Intervention Details

TABLE 14: Characteristics of SCI animal studies using minocycline* (continued)

(continued)

Comments

Translational potential of pharmacotherapy for SCI

J Neurosurg: Spine / Volume 17 / September 2012

191

192

Teng et al.,
2004

Tan et al., 2009


(continued)

Animal model:
Sprague-Dawley rats
N = NR
Sex: 100% F
Age: NR
Wt: 280330 g
SCI model:
Wt drop method; mod erate injury at T-10
spine level (10 g
2.5 cm; 25 g/cm
force)

Authors & Year Animal & Injury Models

SCI + minocycline
SCI + vehicle
Sham group

Experimental Groups

Reported Outcomes

Behavioral Testing (Day 28/4 wks post-SCI):


SCI + Minocycline vs SCI + Vehicle
BBB locomotor rating scale: 11.2 0.8 vs 9.6 0.5 (p = 0.002)
Mechanical paw-withdrawal thresholds: 9.7 3.4 g vs 3.4 1.4
g (p = 0.003)
Thermal paw-withdrawal latencies: 7.5 0.7 sec vs 5.6 1.0
sec (p = 0.009)
Route: IP inj
Dose-Response Effect of Minocycline on Cytochrome c Release
Timing: 1 hr post-SCI
(Western blots):
Dosage:
Minocycline at 90 & 180 mg/kg showed significant blocking
Dose-response expt
effects, essentially reducing cytochrome c release to the
Minocycline: 18, 90, & 180 mg/kg
negligible pre-SCI level
Behavioral & histological expts
Mechanical Paw Withdrawal Reflex:
Minocycline: 90 mg/kg (in 10 ml normal Greater return to a normal withdrawal reflex in response to an ex saline) then 45 mg/kg (in 5 ml nor tension stimulus was noted in SCI + minocycline vs SCI + vehi mal saline) every 12 hrs for 5 days
cle rats (70% vs 25% of hind limbs; p <0.010) at 4 wks post-SCI
Vehicle: saline at corresponding time
Toe-Spread Reflex:
points
A trend toward greater recovery to normal of the toe-spread
Animals killed:
reflex was noted in the SCI + minocycline vs SCI + vehicle
4 hrs post-SCI (tissue collection for
rats (65% vs 35% of hind limbs; p <0.370) at 4 wks post-SCI
dose-response study)
Inclined-Plane Testing:
4 wks (after behavioral tests)
SCI + Minocycline vs SCI + Vehicle
Upward (forelimb strength):
1 wk: 58 vs 60
2 wks: 66 vs 64
3 wks: 66 vs 64
4 wks: 67 vs 66
2 groups did not differ significantly at any time point measured
Downward (coordinated hindlimb motor function):
1 wk: 52 vs 47
2 wks: 55 vs 49
3 wks: 55 vs 50
4 wks: 56 vs 51
SCI + minocycline showed a statistically significant improvement
compared w/ SCI + vehicle (p <0.040) beginning on Day 7
post-SCI & persisting through Day 28
BBB Score:
SCI + Minocycline vs SCI + Vehicle
1 wk: 9 vs 7
2 wks: 12 vs 10
3 wks: 14 vs 11 (p <0.05)
4 wks: 17 vs 13 (p <0.05)
Recovery of capacity to use the hind limbs in open-field locomo tion significantly improved in an overall statistical comparison
(p <0.033)

Intervention Details

TABLE 14: Characteristics of SCI animal studies using minocycline* (continued)

(continued)

Comments

C. H. Tator et al.

J Neurosurg: Spine / Volume 17 / September 2012

Teng et al.,
2004
(continued)

Authors & Year Animal & Injury Models

Experimental Groups

Intervention Details

TABLE 14: Characteristics of SCI animal studies using minocycline* (continued)

Histopathology:
Lesion Vol
No significant differences btwn SCI + minocycline- & SCI +
vehicle-treated groups in:
overall cross-section profiles of lesion epicenters
lesion area size in tissue at other loci rostral & caudal to
epicenter
longitudinal lesion lengths (9.01 0.47 mm vs 9.12 0.59
mm)
White Matter Sparing
Significantly greater in SCI + minocycline vs SCI + vehicle group
in:
Average area of residual total white matter (p = 0.014)
At 2, 3, & 4 mm rostral to the lesion epicenter (p <0.03)
At 3 & 4 mm caudal to the lesion epicenter (p <0.03)
Linear regression analysis indicated a significant positive corre lation btwn the area of preserved white matter 4 mm caudal
to the injury epicenter & overall hindlimb function, as
measured by the BBB scale 4 wks after SCI (10 rats/group;
p <0.036)
Ventral Horn Neuron Protection
SCI + minocycline significantly protected ventral horn neurons
vs SCI + vehicle in the overall analysis (p <0.03), & 3 & 4 mm
rostral to the epicenter, as well as 2, 3, & 4 mm caudal to the
epicenter (p <0.05)
Reactive Astrocytes (GFAP-positive glia):
SCI + minocycline showed a significantly reduced no. of reactive
astroglia vs SCI + vehicle: 18.0 2.3 vs 26.1 1.9 astroglia
per 10 m of tissue (p = 0.017)
Protection of Oligodendrocytes (CNPase):
Systemic minocycline admin protected oligodendrocytes: a sig nificantly greater luminosity was detected in the SCI + mino cycline vs SCI + vehicle group (73.8 3.5 luminosity units vs
26.1 1.9 luminosity units, respectively; p <0.001)

Reported Outcomes

(continued)

Comments

Translational potential of pharmacotherapy for SCI

J Neurosurg: Spine / Volume 17 / September 2012

193

194

Animal model:
CD-1 mice
N = 124
Sex: 100% M
Age: 3 mos
Wt: NR
SCI model:
Extradural compres- sion at T3/4 using a
modified aneurysm
clip (closing force of
8 g); clip left in for 1
min & then removed

Wells et al.,
2003

Route: IP inj
Timing: 0 min post-SCI
Dosage: minocycline 90 mg/kg imme diately post-SCI, then 45 mg/kg 9
hrs later; the following day, 45 mg/
kg 2 /day up to 3 days post-SCI
Animals killed: none; blood samples
were taken daily from the tail vein of
rats on Days 14 post-SCI to test
pNF-H levels

Intervention Details

Reported Outcomes

BBB Score (hindlimb motor function):


SCI + Minocycline vs SCI + Vehicle
Day 28: 14.5 0.6 vs 12.4 0.4 (p <0.05)
pNF-H Levels (biomarkers of tissue damage/neurodegeneration):
SCI + Minocycline vs SCI + Vehicle
Day 3: 0.088 0.018 g/g vs 0.112 0.011 g/g (p = 0.27)
The same trend was observed at Day 4. The reduction in the
plasma pNF-H levels at 3 & 4 days post-SCI may indicate the
protective effects of minocycline against axonal damage in
the treated group
pNF-H Levels & Locomotor Function Recovery:
A negative correlation (rs = 0.78; p <0.05) was observed btwn
the pNF-H levels at 3 days post-SCI & BBB scores at 28 days
post-SCI, indicating that rats w/ lower pNF-H levels are more
likely to achieve higher hindlimb motor scores
Expt 1 (mice in both temRoute: IP inj
Expt 1:
perature environments): Timing: 1 hr post-SCI
Nontemperature-Controlled Environment
SCI + vehicle (saline)
Dosage:
Mortality:
(n = 41)
Expt 1
SCI + Minocycline vs SCI + Vehicle
SCI + minocycline (n = 43) Minocycline: 50 mg/kg 1 hr post-SCI,
Over 3 wks post-SCI: 20% (5 of 25) vs 61.5% (16 of 26); p =
Expt 2 (behavioral recov- then 50 mg/kg 24 hrs later; then 25 0.0039
ery only; temperature mg/kg every 24 hrs for the next 5
Temperature-Controlled Environment
controlled environment days
BBB Score:
only):
Vehicle: normal saline (dose NR) at
Repeated-measures analysis revealed a significant Tx effect
SCI + minocycline (n = 10) same inj schedule as minocycline
(F = 18.283, p = 0.0002) & a day effect (F = 22.404,
SCI + vehicle (normal sa- Expt 2
p <0.0001), as well as a significant Tx by day interaction (F =
line; n = 10)
Minocycline: normal saline & saline pH 2.345, p = 0.0251), where SCI + minocycline mice exhibited
SCI + vehicle (pH 5.0 sa- 5.0 as in Expt 1 above
significant recovery in hindlimb function compared w/ SCI +
line to mimic pH of the
vehicle
Methylprednisolone: 30 mg/kg intra minocycline solution;
muscularly 1 hr post-SCI, then every Average score on Day 3 post-SCI: minocycline 5 vs vehicle 2
n = 10)
6 hrs for 24 hrs (total of 5 injs/
(p <0.005)
SCI + MPSS (n = 10)
SCI + minocycline mice continue to show significant improvemouse)
n = 51 in the nontempera ments compared w/ SCI + vehicle mice at Days 7, 10, 14, 17,
Animals killed: Day 28 post-SCI
ture-controlled environ 21, & 24 (p <0.005)
ment
Average score at 4 wks post-SCI: minocycline 10 vs vehicle 4.5
n = 73 in the temperature (p <0.005)
controlled environment
>50% of the mice that received minocycline had scores >10,
indicating consistent stepping patterns as well as displaying
some evidence of forelimb-hindlimb coordination

SCI + minocycline
Animal model:
SCI + vehicle (saline)
Sprague-Dawley rats
4/group
N=8
Sex: NR
Age: adult; 1012 wks
old
Wt: 280320 g
SCI model:
Wt drop method; mod erate injury at T-10
level (10 g 2.5 cm;
25 g/cm force)

Experimental Groups

Ueno et al.,
2011

Authors & Year Animal & Injury Models

TABLE 14: Characteristics of SCI animal studies using minocycline* (continued)

(continued)

Because of the high


mortality rate of
mice in nontemperature-con trolled environ ment, subsequent
expts used ani mals that were
housed in a warm
room maintained
at 27C constantly
throughout the sur vival period (re ferred to as a temperature-con trolled environment)
BBB scores at other
time points (Days
724) would need
to be estimated
from Fig. 2-A in
article

Comments

C. H. Tator et al.

J Neurosurg: Spine / Volume 17 / September 2012

Wells et al.,
2003
(continued)

Authors & Year Animal & Injury Models


Expt 2 (continued):
Mice in the nontempera ture-controlled environ ment were used for mor tality studies; mice in the
temperature-controlled
environment were sub jected to behavioral
testing (all mice), while a
subset were used for flu orogold & other histolog ical analyses

Experimental Groups

Intervention Details

TABLE 14: Characteristics of SCI animal studies using minocycline* (continued)

Expt 1 (continued):
Temperature-Controlled Environment
Inclined-Plane Test (hindlimb strength):
Protective effect seen w/ minocycline Tx (F = 5.878, p = 0.0249)
SCI + minocycline mice showed significantly higher scores than
SCI + vehicle mice at Wks 3 & 4 postinjury
Day 7: 37 vs 32 (p = NS)
Day 14: 50 vs 41 (p = NS)
Day 21: 55 vs 40 (p <0.05)
Day 28: 58 vs 47 (p <0.05)
There was a significant correlation btwn BBB score & inclined plane score over all test days (r = 0.70, p <0.0001)
Retrograde Labeling of Red Nucleus Neurons:
There was a trend toward greater preservation of the RST in SCI
+ minocycline vs SCI + vehicle mice
There was a significant correlation btwn BBB score at Day 28 &
neuronal soma counts in the red nucleus (r = 0.84, p <0.0001)
Preservation of Axonal Integrity (Bielschowsky silver stain):
Qualitatively more intact axons in SCI + minocycline (n = 6)
compared w/ SCI + vehicle mice (n = 5)
Global Neuroprotective Effect on Spinal Cord Tissue (including
nonaxonal elements):
H&E-stained tissue revealed a significant reduction in lesion
size in SCI + minocycline compared w/ SCI + vehicle mice
(1.76 vs 2.80 mm2, respectively; p <0.001)
Expt 2:
Minocycline Tx resulted in significant recovery of hindlimb func tion; MPSS inj did not lead to behavioral improvement com pared w/ saline controls
Repeated-measures analysis revealed a significant Tx effect
(F = 6.020, p = 0.003) & a day effect (F = 57.362, p <0.001),
as well as a significant Tx by day interaction (F = 2.169, p =
0.004)
Multiple-comparison post hoc analysis using the Tukey test
uncovered significant differences btwn SCI + minocycline
mice & mice from all other Tx groups (p <0.01, p <0.05, &
p <0.05 for vehicle; vehicle [pH 5.0]; & MPSS, respectively)
The pH 5.0 vehicle group did not differ from saline controls

Reported Outcomes

J Neurosurg: Spine / Volume 17 / September 2012


(continued)

In Expt 2, they con trolled for the


possibility that the
pH (5.0) of the
minocycline solu tion might have
generated a stress
response, by in cluding a vehicle
group in which
the pH of the sa line solution was
lowered to 5.0 w/
HCl

Comments

Translational potential of pharmacotherapy for SCI

195

196

Yune et al.,
2007

Animal model:
Sprague-Dawley rats
N = NR
Sex: 100% M
Age: adult
Wt: 250300 g
SCI model:
Wt drop method; mod erate injury at T-10
spine level (10 g
2.5 cm; 25 g/cm
force)

SCI + minocycline
SCI + vehicle (saline)
SCI + MPSS
Sham (T-10 laminectomy
w/o wt drop injury)
Pro-NGF expression:
n = 6 for vehicle & minocycline
Phosphorylated p38MAPK
expression: n = 6 for
vehicle & minocycline
Phosphorylated
MAPKAPK-2 expres sion: n = 6 for vehicle &
minocycline
p75NTR expression:
n = 3 for sham, vehicle, &
minocycline
Rho expression: n = 3 for
vehicle, minocycline, &
SB203580
Caspase-3 staining:
n = 3 for vehicle & minocycline
Behavioral testing:
n = 20 for vehicle, minocy cline, & MPSS

Authors & Year Animal & Injury Models Experimental Groups


Route: IP inj
Timing: 0 hrs post-SCI
Dosage:
Molecular expts
Minocycline: 90 mg/kg immediately
post-SCI, then 45 mg/kg every 12
hrs for 3 days
Vehicle: normal saline (equivolumetric)
on same injection schedule as
minocycline
Caspase-3 staining (oligodendrocyte
death) & behavioral expts
Minocycline: 90 mg/kg either immedi ately or 2 hrs post-SCI, then 45 mg/
kg every 12 hrs for 3 days
Vehicle: normal saline (equivolumetric)
on same inj schedule as minocycline
MPSS: 30 mg/kg either immediately or
2 hrs post-SCI, then every 12 hrs
for 3 days
For some expts, SB203580 (1 & 5 g)
was injected directly into the spinal
cord at the lesion epicenter 2 hrs
after SCI (n = 3 for sham, vehicle, or
SB203580)
Animals killed: NR

Intervention Details

TABLE 14: Characteristics of SCI animal studies using minocycline* (continued)

Pro-NGF is the death


ligand in apoptosis
of oligodendro cytes after SCI

Pro-NGF Expression:
Minocycline significantly inhibited pro-NGF expression when
compared w/ that in vehicle controls
Relative levels Day 5 post-SCI: minocycline 3 vs vehicle 10;
p <0.001
p38MAPK-Dependent Pro-NGF Expression in Microglia:
Minocycline Tx significantly inhibited the phosphorylation of both
p38MAPK & MAPKAPK-2 compared w/ vehicle at 3 & 5 days
post-SCI (p <0.001)
An in vitro model of LPS-induced microglial activation indicates
that pro-NGF expression is mediated by activation of
p38MAPK & that minocycline or SB203580 inhibits
p38MAPK-dependent pro-NGF expression in microglia
Minocycline (1 & 5 nM) Tx significantly inhibited the LPS-induced
p38MAPK activation 30 min after LPS Tx compared w/
cultures treated w/ LPS only (p <0.001)
Minocycline (1 & 5 nM) or SB203580 (1 & 5 m), an inhibitor
of p38MAPK Tx, significantly inhibited the phosphorylation of
MAPKAPK-2 30 min after LPS Tx compared w/ cultures
treated w/ LPS only (p <0.001)
Microglia-Derived Pro-NGF Induces Apoptosis of Oligodendro cytes:
In vitro results indicate that pro-NGF produced by activated
microglia induces the p75NTR-mediated apoptotic cell death
of oligodendrocytes & that minocycline or SB203580 inhibits
LPS-induced pro-NGF production in microglia; p <0.001
compared w/ LPS
p75NTR Expression & RhoA (downstream molecule of the
p75NTR signaling pathway) Activation:
Minocycline Tx significantly inhibited p75NTR mRNA & protein
expression compared w/ vehicle-treated controls at 5 days
post-SCI (p <0.001)
Minocycline Tx significantly inhibited RhoA activation compared
w/ vehicle-treated controls at 3 (p <0.05) & 5 (p <0.001) days
post-SCI
Apoptosis of Oligodendrocytes:
Rats were treated 2/day for 3 days w/ minocycline, beginning 2
hrs postinjury
Minocycline Tx significantly decreased the no. of activated cas pase-3positive oligodendrocytes at 5 days post-SCI compared
w/ that observed in the vehicle-treated controls (p <0.001)
Minocycline Tx markedly reduced the extent of both myelin
& axon loss at Day 38 post-SCI compared w/ vehicle-treated
controls (p <0.05)

(continued)

p75NTR & RhoA


activation leads to
apoptosis of
neurons & oligo dendrocytes after
SCI

p38MAPK mediates
inflammatory re sponses in
microglia
MAPKAPK-2 is a
downstream mole cule of p38MAPK
LPS is known to pro mote the activation
of BV2 cells that
exhibit phenotypic
& functional prop erties of activated
microglial cells
in vivo

Comments

Reported Outcomes

C. H. Tator et al.

J Neurosurg: Spine / Volume 17 / September 2012

J Neurosurg: Spine / Volume 17 / September 2012

Animal model:
C57BL/6J mice
N = 60
Sex: 50% M
Age: 6 wks
Wt: NR
SCI model:
Transection (using iri dectomy scissors)
of the bulbospinal
tract ipsilaterally &
dorsal & ventral
CSTs bilaterally at
T-12

SCI + minocycline
SCI + vehicle
SCI + LIF
6 groups of 10 mice (5 M &
5 F/group)

Experimental Groups

Route: IP inj
Timing:
Minocycline & vehicle groups: 2 hrs
post-SCI
LIF groups: 2, 8, 24 hrs, & 1 wk postSCI
Dosage:
Minocycline: 10 mg/kg 3/wk
Vehicle: 1% albumin in 0.1 M mouse
tonicity phosphate-buffered saline
3/wk
LIF: 25 g/kg 3/wk
Animals killed: 6 wks post-SCI

Intervention Details
Functional Recovery:
BBB Scores
Minocycline Tx either immediately or 2 hrs after SCI significantly
increased the hindlimb locomotor function (higher BBB
scores) 2134 days post-SCI compared w/ vehicle-treated
controls (p <0.05 to 0.01)
Inclined-Plane Test
Minocycline Tx either immediately or 2 hrs after SCI significantly
increased the hindlimb locomotor function (higher angles of
inclined plane) 14 wks post-SCI (p <0.05 & p <0.01, respec tively) compared w/ vehicle-treated controls
Grid Walk Test
The grid error % (the % of footfalls per steps) of the minocycline
group (administered either immediately or after a 2-hr delay)
was significantly lower than that observed in the vehicle treated group 5 wks postinjury (p <0.05)
Footprint Recordings
Footprints from the minocycline-treated rats showed fairly consis tent forelimbhindlimb coordination & very few toe drags; these
findings were comparable w/ those observed in normal animals
In contrast, the footprints obtained from vehicle- or methylpred nisolone-treated animals showed inconsistent coordination &
extensive drags
Rotarod (balancing posture):
The 2-, 8-, & 24-hr delayed LIF groups had significantly
(p <0.001) improved outcomes compared to vehicle &
minocycline groups
Bar Grab, Bar Walk, & Platform Hang:
In all 3 behavioral tests, the recovery of locomotor function was
statistically significant (p <0.05) in the 2-, 8-, & 24-hr delayed
LIF compared to the 1-wk delayed LIF, vehicle, & minocycline
groups
Histology:
In all 4 compartments tested (dorsal CSTs & dorsal, lateral, &
ventral white matter) the no. of myelinated axons was higher
in the 2-, 8-, & 24-hr delayed LIF groups compared to the
vehicle group (p <0.05 to 0.01)
A significantly (p <0.01) higher no. of the CST axons below the
lesion at L-1 were seen in the 2-, 8-, & 24-hr delayed LIF
groups compared to minocycline & vehicle groups

Reported Outcomes

No data given for


comparison of
minocycline & ve hicle groups, but
outcomes on all
behavioral testing
appear to be simi lar btwn the 2
groups, as shown
by Fig. 3

Comments

* Values represent mean standard deviation unless otherwise indicated. Abbreviations: AST = ascending sensory tract; BMS = Basso Mouse Scale; CST = corticospinal tract; HDL = high-density
lipoprotein; IL = interleukin; LDL = low-density lipoprotein; LIF = leukemia inhibitory factor; LPS = lipopolysaccharide; MP = methylprednisolone; NGF = nerve growth factor; NS = not significant; pNF-H =
plasma neurofilamentheavy; p75NTR = p75 neurotrophin receptor; RST = rubrospinal tract; qRT-PCR = quantitative reverse transcriptasepolymerase chain reaction; TGF = transforming growth factor ;
TNF- = tumor necrosis factor; TUNEL = terminal deoxynucleotidyl transferasemediated deoxyuridine triphosphate nick-end labeling.
Estimated from figure(s) in article.
Gale K, Kerasidis H, Wrathall JR: Spinal cord contusion in the rat: behavioral analysis of functional neurologic impairment. Exp Neurol 88:123134, 1985.

Zang &
Cheema,
2003

Yune et al.,
2007
(continued)

Authors & Year Animal & Injury Models

TABLE 14: Characteristics of SCI animal studies using minocycline* (continued)

Translational potential of pharmacotherapy for SCI

197

C. H. Tator et al.
Hains and Waxman31 found that minocycline treatment resulted in an immediate increase in mechanical and
thermal paw withdrawal thresholds that was not observed
in vehicle-treated animals; cessation of minocycline
treatment resulted in a return of thresholds to predrug
levels. Other studies reported similar improvements with
minocycline treatment at 14 weeks following injury in
mechanical allodynia47,70,75 and thermal hyperalgesia.47,70
In contrast, Lee and colleagues43 reported no difference
at 6 weeks in the mechanical force needed for forepaw
withdrawal between treatment groups.
Safety of Therapy. Wells and colleagues80 reported
significantly reduced mortality rates in mice who received minocycline compared with those treated with vehicle only and evaluated 3 weeks after SCI (5 of 25 vs 16
of 26, respectively; p = 0.0039). None of the other studies
assessed the safety of minocycline.
Summary of Preclinical Trials of Minocycline. There
is a remarkable variability in the preclinical results reported for minocycline, with many gaps in information
required to assess translational readiness.

Nimodipine
Background. Excessive entry of calcium ions into
cells plays a key role in the pathogenesis of posttraumatic
ischemia and ischemic cell death.11,19 Nimodipine primarily functions in the CNS as a calcium antagonist, also referred to as a slow channel blocker,20 helping to mitigate
the effects of secondary injury following SCI. Nimodipine has also been shown to increase posttraumatic spinal
cord blood flow due to a selective vasodilatory effect on
spinal cord vessels.1,27 Nimodipine has been approved for
oral administration only to treat symptoms from ruptured
intracranial aneurysms and subarachnoid hemorrhage, at
a dose of 60 mg (two 30-mg capsules) every 4 hours for
21 consecutive days.
Systematic Search for Preclinical Studies. We identified 30 articles evaluating nimodipine in animal models
of SCI. Eighteen were judged suitable for full-text review;
4 of which were then excluded for the following reasons:
2 studies did not include a control group, and in 2 studies
nimodipine was administered prior to injury. Therefore,
14 articles were identified that met our inclusion criteria.9,18,20,23,25,28,30,33,34,37,56,5961
Study Characteristics. Study characteristics are summarized in Table 15; detailed information can be found in
Table 16. Nine of the 14 studies used a range of 15 to 44
rats,9,20,28,33,34,37,5961 2 used 32 or 80 rabbits,18,25 2 used 10 or
22 cats,23,30 and 1 used 13 baboons.56 Most of the studies
induced SCI in the thoracic spine by one of the following
methods: clip compression (9 studies),9,20,25,28,34,37,5961 balloon compression (1), 56 plate compression (1),33 or weight
drop (1).30 Ford and Malm23 injured the lumbar spinal
cord by weight drop, and Faden and colleagues18 used
aortic occlusion to induce ischemic SCI. Systemic injection was used by all studies; intravenous,18,23,25,28,30,33,59,60
intraperitoneal,9,20,37,56,61 or intrathecal34 infusions were
used. There was a wide range of doses and duration of infusion. The first dose was given up to 2 hours after injury,
and continuous infusions lasted for 35 minutes to 8 hours.
198

Two studies gave 3 additional daily doses for 4 or 7 days


after the period of continuous infusion, 23,61 and 3 studies
evaluated multiple doses of nimodipine.18,20,59 The majority of studies did not evaluate multiple doses of nimodipine. All but 3 studies18,25,37 randomized animals to treatment groups, and outcomes were assessed in a blinded or
independent manner by 8 studies18,20,23,28,30,5961 of the 14
total. Four of the studies18,25,28,34 did not account for all of
the included animals.
Effectiveness of Therapy. The effect of nimodipine on necrosis and hemorrhage of the spinal cord was
evaluated by a number of studies, and in general no effect was found. Ceylan and colleagues9 reported similar
degrees of necrosis and hemorrhaging in both the white
and gray matter in rats treated with nimodipine or saline
as measured 5 hours after injury; Faden18 reported similar results for necrosis in rabbits killed 48 hours postSCI. Three additional studies reported similar degrees of
hemorrhage in the white and gray matter between treatment groups.20,59,60 Guha and colleagues28 reported similar amounts of hemorrhage between treatment groups in
the areas closest to the lesion, but found that there was
more hemorrhage in nimodipine-treated animals at areas
farther from the injury site compared with those that received saline alone, although this difference was not statistically significant.
Nimodipine was found to have no effect on the MABP,
which declines after SCI. After treatment, MABP was in
general statistically similar in the nimodipine and saline
treatment groups, as reported in 7 studies.9,20,23,25,33,59,61 In
contrast, Guha and colleagues28 reported that treatment
with nimodipine and adrenaline (or adrenaline alone)
resulted in a significant increase in MABP. Adrenaline
was required to counteract the hypotensive effect of nimodipine alone. Imamura and Tator34 also found that nimodipine treatment resulted in significantly lower MABP
levels at 60 minutes compared with those seen in the
placebo group, although the effect was transient. Spinal
cord blood flow was reported to be similar between nimodipine and saline or placebo groups after injury in 5
studies.28,33,34,59,60 In contrast, Pointillart and colleagues56
found that spinal cord blood flow was significantly increased in nimodipine-treated animals by 90 minutes after injury compared with that in the saline-treated group;
this improvement lasted until the baboons were killed at
8 days postinjury. Fehlings and colleagues20 reported increases in spinal cord blood flow in some regions of the
cord following nimodipine with dextran treatment (but
not nimodipine alone). In a study of 13 baboons, Pointillart and colleagues56 reported that nimodipine produced
significant improvements in lesion length in both the
white and gray matter as measured 8 days after injury.
The SSEPs were similar between treatment groups at up
to 5 hours after injury as reported by 4 studies,9,34,59,60 and
at 8 days after injury as reported by 2 studies.56,61 Fehlings and colleagues20 reported a significant improvement
in SSEPs in rats treated with nimodipine and dextran-40
(but not with nimodipine or dextran alone) as measured
in the first few hours after injury. In contrast, Haghighi
and colleagues30 reported a deleterious effect on spinal
evoked potential activity in a small study of 10 cats.
J Neurosurg: Spine / Volume 17 / September 2012

animal model (no./group)


rats
rabbits
cats
baboons
injury model
blunt force (wt drop) (lumbar)
blunt force (wt drop) (thoracic)
clip compression (thoracic)
balloon compression
aortic occlusion
plate compression (thoracic)
timing of intervention (postinjury)
@ 0 min (single dose)
@ 0 min for 2 hrs
@ 0 min for 2 hrs, then higher dose
for 8 days cont infusion
@ 5-min loading dose, then cont
infusion of a lower dose for 2 hrs
@ 15 min for 60- or 120-min cont
infusion
@ 15 min for 180-min cont infusion
@ 15 min for 8 hrs
@ 15 min for 8 hrs, then 3/day for
7 days
@ 30 min for 24 hrs, then oral
admin for 4 days (3/day)
@ 1 hr (single dose)
@ 1 hr for 30-min cont infusion
@ 1 hr for 60-min cont infusion
@ 1 hr for 240-min cont infusion
@ 2 hrs for 35-min cont infusion
@ 2 hrs for 60-min cont infusion

Characteristic

J Neurosurg: Spine / Volume 17 / September 2012


X

30 (10)

Ceylan
et al.,
1992

32 (715)

Faden
et al.,
1984

30 (5)

Fehlings
et al.,
1989

TABLE 15: Summary of nimodipine preclinical study characteristics

22 (11)

Ford &
Malm,
1985

X (T12L1)

80 (16)

Gambardella et
al., 1995

15 (5)

Guha
et al.,
1989

10 (5)

Haghighi
et al.,
1988

30 (10)

Holtz
et al.,
1989

25 (5)

Imamura
& Tator,
1998

X (C7T1)

40 (10)

Kaynar
et al.,
1998

13 (5)

25 (5)

Pointillart Ross &


et al.,
Tator,
1993
1991

30 (10)

Ross &
Tator,
1993

(continued)

X
X

44 (1013)

Ross
et al.,
1993

Translational potential of pharmacotherapy for SCI

199

200

route of intervention
IV
IP
IT
dosage
0.005 mg/kg
0.01 mg/kg
0.02 mg/kg
0.02 mg/kg for 8-hr cont infusion,
then 20 mg/kg 3/day for 7 days
0.02 mg/kg/hr for 2 hrs, then 0.04
mg/kg for 8 days cont infusion
0.025 mg/kg
0.05 mg/kg
1 g/kg/min for 24 hrs, then oral
doses of 5 mg 3/day for 4 days
loading dose of 10 g/kg, then 1
g/kg/min 120-min infusion
1 g/kg/min 60 min
1.5 g/kg/min 60 min
1.5 g/kg/min 120 min
1.5 g/kg/min 240 min
2 g/kg/min 120 min
>1 dose evaluated?
yes
no
control groups
SCI (saline)
SCI (no Tx)
SCI (PEG + ethanol)
no SCI
independent or blind assessment
yes
no
NR

Characteristic

X
X

Fehlings
et al.,
1989

Faden
et al.,
1984

Ceylan
et al.,
1992

Ford &
Malm,
1985

TABLE 15: Summary of nimodipine preclinical study characteristics (continued)

Gambardella et
al., 1995

Guha
et al.,
1989

Haghighi
et al.,
1988

Holtz
et al.,
1989

Imamura
& Tator,
1998

Kaynar
et al.,
1998

X
X

X
X

Pointillart Ross &


et al.,
Tator,
1993
1991

X (per hr)

Ross &
Tator,
1993

Ross
et al.,
1993

C. H. Tator et al.

J Neurosurg: Spine / Volume 17 / September 2012

100% (44
of 44)
100%
(30 of
30)

NR

100% (40 100% (13


of 40)
of 13)

100%
(25 of
25)

X
X
X

100%
(30 of
30)
100% (10
of 10)

NR

X
X

Holtz
et al.,
1989

72% (23
of 32)
100%
(30 of
30)

X
X

animals randomized to Tx groups


yes
no
NR
% animals accounted for

Characteristic

100% (30 100% (22


of 30)
of 22)

X
NR
X
X

Ford &
Malm,
1985
Fehlings
et al.,
1989
Faden
et al.,
1984
Ceylan
et al.,
1992

TABLE 15: Summary of nimodipine preclinical study characteristics (continued)

Gambardella et
al., 1995

Guha
et al.,
1989

Haghighi
et al.,
1988

Imamura
& Tator,
1998

Kaynar
et al.,
1998

Pointillart Ross &


et al.,
Tator,
1993
1991

Ross &
Tator,
1993

Ross
et al.,
1993

Translational potential of pharmacotherapy for SCI

J Neurosurg: Spine / Volume 17 / September 2012

In general, there were no differences in measures of


neurological and motor function. Faden and colleagues18
reported no differences between treatment groups in neurological scores, which were not defined, at 24 and 48
hours after injury. Ford and Malm23 found that motor recovery scores at 6 weeks were similar between groups:
all but 2 animals in the nimodipine group were unable to
walk at 6 weeks; however, this difference did not reach
statistical significance (0 of 11 vs 2 of 11; p < 0.10). Ross
et al.61 reported similar inclined-plane scores between nimodipine-treated animals and those that received placebo
alone at all times up to 7 weeks; similar results were reported for a composite outcome score. In contrast, Gambardella and colleagues25 reported improvements in the
Tarlov scale scores in rabbits that received nimodipine
and adrenaline compared with saline at 24 and 48 hours
after injury; however, the improvements with nimodipine
alone were minimal. The authors did not assess statistical
significance.
Safety of Therapy. None of the studies assessed the
safety of nimodipine, other than the hypotensive effects
described above.
Summary of Preclinical Trials of Nimodipine. Although a large body of preclinical data exists on nimodipine, there are several gaps in knowledge about its translational readiness in the categories of its effectiveness,
safety, dosage, and route of administration.

Riluzole
Background. Riluzole works by blocking voltageactivated sodium and calcium ion channels, inhibiting
presynaptic glutamate release, and activating potassium
ion channels.16,17 Potassium serves to stabilize the resting
membrane potential, thereby modulating cell excitability
in the central and peripheral nervous system.8 Riluzole
has shown effectiveness for the treatment of patients with
amyotrophic lateral sclerosis at a recommended dose of
50 mg every 12 hours.
Systematic Search for Preclinical Studies. Twentyseven articles were identified for riluzole, 18 of which
were selected to undergo full-text review. Six studies were
excluded after full-text review for the following reasons:
riluzole was administered to spinal cords maintained in
vitro in 1 study, it was administered prior to SCI in 4, and
1 study was a review. Therefore, 12 studies met our inclusion criteria.5,7,32,38,4852,54,63,68
Study Characteristics. Study characteristics are summarized in Table 17; detailed information can be found
in Table 18. All studies examined the effects of riluzole
in SCI in rats; 2090 animals were used per study. All
regions of the spinal cord were injured in at least 1 study:
thoracic SCI was induced by clip (2 studies)32,63 or by
balloon compression (1),68 lumbar injury by weight drop
(4)5,4850 or avulsion (3),7,51,52 sacral injury by transection
(1),38 and cervical injury by avulsion (1).54 The dosage
of riluzole ranged from 0.8 to 10 mg/kg, and was given immediately or up to 2 hours following injury. Many
of the studies gave repeated doses of riluzole for 316
days. The route of drug delivery included intraperitoneal, 5,7,32,38,4952,54,63 intrathecal,32 and intravenous injec201

C. H. Tator et al.
tions,68 as well as delivery via intracerebroventricular
cannula32 or microdialysis fiber.48 Half the studies randomized animals to the treatment groups, 5,32,49,50,63,68 and
in half blind or independent assessment of treatment outcomes were performed.5,7,38,50,63,68 Finally, half the studies
accounted for 100% of the treated animals.5,51,52,54,63,68
Effectiveness of Therapy. A wide variety of histopathological analyses was undertaken to assess the effects
of riluzole on spinal cord tissue following SCI. Treatment
with riluzole significantly improved lesion size compared
with that observed in animals treated with SCI and saline
or vehicle at 6 or 7 weeks, as reported by 2 studies5,63 (p
< 0.05). One of these studies also reported significantly
greater normalized residual tissue area and white and gray
matter volumes as well as cell diameters in riluzole-treated
animals at 7 weeks post-SCI (p < 0.05).63 Five studies reported significantly greater numbers of neurons retained
in rats treated with riluzole compared with saline/vehicletreated controls or no treatment,5,7,51,52,63 but 1 study reported no difference in the number of cholinergic neurons between treatment groups.54 Schwartz and Fehlings63 found
that the improvements in neuron counts between the riluzole and control groups were localized to the red nucleus (p
= 0.02). Ngrdi et al.51 reported improvements in the number of motoneurons, reinnervating neurons, and cholinergic cells in the spinal cords of injured rats that received
riluzole compared with no treatment, regardless of when
treatment was started (5, 10, 14, or 16 days) after injury,
although larger effects were observed when treatment began at 5 or 10 days compared with 14 or 16 days postinjury.
Furthermore, the authors found that the number of retrogradely labeled neurons decreased sharply when animals
were treated with riluzole at 14 or 16 days (compared with
initial treatment at 010 days).
Riluzole also improved the following indicators of oxidative stress compared with saline/vehicle or no treatment:
lipid peroxidation levels,5 glucose uptake,50 and mitochondrial function.50 Mu and colleagues50 noted significant improvements in glutamate uptake in the riluzole compared
with the vehicle/saline groups (p < 0.01), whereas McAdoo
et al.48 reported no effect on glutamate release following
SCI. In addition, Mu and colleagues50 reported that riluzole alone had no effect on reactive oxygen species levels
in synaptosomes compared with vehicle/saline treatment.
Improved outcomes in motor function were observed
in riluzole-treated compared with vehicle/saline-treated rats. In general, the available evidence suggests that
functional improvements with riluzole treatment may
not become apparent until at least 1 month after injury.
Two studies reported significant improvement in the BBB
score at 13 months.7,63 Mu and colleagues49 reported that
only when riluzole was combined with methylprednisolone was a significant improvement in BBB scores observed; this effect was not seen in animals that received
either drug alone, and was not observed until 4 weeks
after trauma. Schwartz and Fehlings63 similarly found
that significant BBB improvements did not occur in the
riluzole group until 4 weeks after SCI, and Hama and
Sagen32 reported no effect at 2 hours after trauma. Ates
and colleagues5 found that while riluzole-treated animals
had significant functional improvements as measured
202

by motor function and inclined-plane scores at 6 weeks,


there were no differences between the riluzole and control groups at 1 or 3 weeks.
Bergerot et al.7 reported significantly improved time
on the accelerating rotarod test in riluzole-treated versus
untreated animals at 3 months after injury. Ngrdi and
colleagues51 found that riluzole treatment resulted in recovery from paralysis beginning 34 weeks after injury;
by 3 months animals were able to walk almost normally,
with no dragging of hindlimbs, and had the ability to flex
their ankle joints. This effect was seen in animals that
received their first dose of riluzole up to 14 days after
injury, whereas untreated controls had no functional recovery. Riluzole treatment had no or transient effects on
improving responses to antinociceptive simuli (heat, von
Frey filament test, light touch, pinch, and stretch) compared with vehicle/saline-treated or untreated animals,
as evaluated by 2 studies.32,38 Pintr and colleagues54
reported significant improvements in wrist contraction
and dorsiflexion in riluzole-treated animals beginning at
8 and 9 weeks, respectively, compared with control animals (p 0.01 and p 0.05, respectively). Stutzmann and
colleagues68 reported similarly near-normal behaviors in
70% of riluzole- and 0% of vehicle-treated animals, and
these effects were observed as early as 1 week after injury. Similarly, these authors reported improvements in
SSEP amplitude, duration, and latency in rats treated with
riluzole but not in those that received vehicle.
Safety of Therapy. None of the studies evaluated the
safety of riluzole.
Summary of Preclinical Trials of Riluzole. This agent
has undergone much preclinical testing in SCI models, but
there are still major gaps in knowledge that could impair
translation.

Discussion

A large effort to find effective neuroprotective agents


for patients with acute SCI has occurred since the 1980s,
beginning with the 3 studies of methylprednisolone and
related agents in the National Acute Spinal Cord Injury
Study. Many patients with SCI have been entered into several well-designed clinical trials, and industry, government,
and clinical scientists have all made major contributions.
Indeed, worldwide there have been at least 9 randomized
prospective controlled trials of neuroprotective pharmacological agents in acute SCI.71 With some exceptions such
as GM-1 ganglioside, these trials occurred after the agents
had undergone a considerable amount of testing in animal
models. Unfortunately, the field still lacks effective neuroprotective/pharmacotherapy agents for improving neurological recovery after acute SCI. In the meantime, in addition to the agents already tested in patients, a very large
number of new agents has shown positive results in one
or more preclinical trials. However, as stated above it has
been very difficult for our NACTN Consortium to select
agents for trial. Clearly, against this background of essentially negative results in human SCI trials, a new method of
evaluating and selecting agents for translation is required.
The 2 objectives of this report were to define current sysJ Neurosurg: Spine / Volume 17 / September 2012

Translational potential of pharmacotherapy for SCI


tems of evaluation, and then to examine in detail the preclinical data and translational readiness of 5 promising
agents based on their preclinical track record.
It was because of this negative background that the
Christopher and Dana Reeve Foundation developed, in
2004, a consortium of 9 centers to plan and conduct clinical trials in traumatic SCI, and this consortium has just
completed its first Phase I study of riluzole. In 2009, the
NACTN developed its Treatment Strategy Selection Committee. The Committee developed its approach to therapy
selection on the basis of 4 broad categories of types of
strategies for human SCI: 1) neuroprotection/pharmacotherapy; 2) neuroregeneration through cell-based therapy
and transplantation; 3) bioengineered platforms; and 4)
physical modalities.
The present review of 5 translationally relevant neuroprotective/pharmacotherapy agents is based on the selection of these agents by the NACTN Treatment Strategy
Selection Committee. These 5 agents were selected because they were judged to be of major interest on the basis
of the amount and quality of preclinical study that had
been performed and the evidence provided of positive
preclinical results predictive of success if translated to
humans with SCI. The basis for this selection process was
somewhat arbitrary, but the intent was to facilitate selection of agents for translation. The Committee then commissioned this detailed review of these 5 pharmacological agents to confirm or reject its a priori selection. It is
recognized as well that the present review is a necessary
step by the Committee to fulfill its mission of developing
an improved system of agent selection for translation.
Unfortunately, in the SCI field, the process for selecting the most favorable preclinically tested agents
for translation has not been well developed or evidence
based, although there have been attempts to improve
the selection through the development of various guidelines. For example, in 2006, Tator71 provided the following guidelines for the assessment of preclinical studies
for purposes of translation: the agents should have been
examined in more than one laboratory, in more than one
species, and in more than one injury model in which the
mechanism of injury is relevant to human SCI. There
are so many primary mechanisms of acute human SCI,
including compression, impact, distraction, laceration,
missile, and others, that it was thought that multiple preclinical models are required to provide confidence from
preclinical trials that an agent will be useful in patients
with SCI.72 The combination of impact and persisting
compression produced by the clip compression model in
rats simulates the most common type of injuries in humans, which are burst fractures and fracture-dislocations.
In contrast, other impact models with brief dwell times
following impact may not reproduce human injuries as
well.74 It is obvious that there is no unanimity of opinion
on this matter, and therefore it is likely that preclinical
investigators will continue to use a variety of injury methods, which complicates comparison of results, but reflects
the diversity of human SCI.
One recent effort to develop a method of evaluating
preclinical studies for translatability was based on a survey conducted among North American experts in SCI,
J Neurosurg: Spine / Volume 17 / September 2012

including several of the present authors, followed by a


modified Delphi consensus-building process among a
small segment of those experts.39 This culminated in the
development of a 100-point preclinical grading scale of
the so-called translational potential that is arbitrary and
of unproven value. Extra points were assigned for cervical injury models over thoracic injury models, which is
hard to justify because both are important sites in human SCI. Other features seem more reasonable, such as
the awarding of more points for rat than mouse studies,
because the pathology of SCI in mice differs markedly
from that in the rat, which resembles human injury so
closely.12 More points were awarded for clinically relevant
models (as is reasonable) as discussed above, and largeanimal studies received more points than rodent studies,
which also seems reasonable. More points were given for
clinically meaningful results, with an emphasis on behavioral outcome measures, and more points for replication
studies showing therapeutic benefit, the value of which
is obvious. However, there are important omissions from
the grading scale, such as safety, drug toxicity, and route
of administration, because only systemically administered agents were included. It is of interest to note that 13
agents were scored, 3 of which are included in the present
review of 5 agents. The scores for MgSO4, minocycline,
and riluzole were 45, 48, and 33, respectively; minocycline was the highest-ranking pharmacological agent of
the 13. It would have been helpful if this previous exercise
had included all the agents that have been translated over
the past 30 years, especially methylprednisolone, naloxone, and GM-1 ganglioside.

Recommendations

We would recommend the following strategy for


developing and validating an improved scoring system
that could be used in the future to assess the translational
readiness of neuroprotection with pharmacotherapy for
human SCI.
Phase I: Develop a Prototype Scoring System

Continue efforts to identify systematically the


existing scoring and grading systems, and to summarize the key criteria of these systems. Identify the
strengths and weaknesses of these systems.

Use this information to develop systematic strategy for identifying additional criteria that may be used
for developing the new system. Identify and summarize additional key criteria from the systematic search,
including their strengths and weaknesses.
Propose a prototype version of the scoring system
to be developed through a formal process and validation.

Apply the prototype system to all agents that have


already been translated and to the most promising of
the nontranslated agents.

Communicate throughout this phase with preclinical and clinical experts worldwide.
203

C. H. Tator et al.
Phase II: Formal Approach to Developing a new Scoring
System

Yoder, and Claire Ely at Spectrum Research, Inc., Tacoma, Washington.

Develop a survey to evaluate the criteria for a


future system based on the prototype from Phase I.

References

Disseminate the results from Phase I and survey


preclinical and clinical experts worldwide.
Collate feedback in a report for review.

Use modified Delphi consensus-building process


and/or focus groups of those worldwide experts to
finalize the scoring system.
Phase III: Validate the new Scoring System

Develop criteria for evaluating the effectiveness


of the scoring system.

Apply scoring system to all preclinical neuroprotective/pharmacotherapy agents that have already been
translated to human SCI and to the most promising of
the current nontranslated agents.
Measure effectiveness through consultation with
worldwide expert group.
Report the new scoring system.

Conclusions

Efforts are currently underway to translate riluzole,


minocycline, magnesium-PEG, and glyburide to clinical
use for SCI. Although these therapies show considerable
promise, gaps in the preclinical literature remainparticularly with regard to issues related to effective time windows, optimal dosing via dose-response studies, toxicity,
and therapeutic tissue levels of the agent. In addition, objective standards are required to assess the preclinical literature related to SCI therapeutic agents to facilitate clinical translation. There is a need for a new scoring system
to evaluate the preclinical results of experimental neuroprotection/pharmacotherapy studies in SCI to determine
translational readiness, and the steps have been outlined
for the development of an optimal scoring system.
Disclosure
Support was received from AOSpine and the Christopher and
Dana Reeve Foundation. The authors report no conflict of interest
concerning the materials or methods used in this study or the findings specified in this paper.
Author contributions to the study and manuscript preparation
include the following. Conception and design: Tator, Hashimoto,
Norvell, Fehlings, Harrop, Guest, Aarabi, Grossman. Acquisition
of data: Hashimoto, Raich. Analysis and interpretation of data:
all authors. Drafting the article: all authors. Critically revising the
article: all authors. Reviewed submitted version of manuscript: all
authors. Approved the final version of the manuscript on behalf
of all authors: Tator. Administrative/technical/material support: all
authors. Study supervision: Tator, Norvell, Fehlings, Harrop, Guest,
Aarabi, Grossman.
Acknowledgments
The authors acknowledge assistance from Erika Brodt, Emily

204

1. Allen GS, Ahn HS, Preziosi TJ, Battye R, Boone SC, Boone
SC, et al: Cerebral arterial spasma controlled trial of nimodipine in patients with subarachnoid hemorrhage. N Engl
J Med 308:619624, 1983
2. Amin AR, Attur MG, Thakker GD, Patel PD, Vyas PR, Patel
RN, et al: A novel mechanism of action of tetracyclines: effects on nitric oxide synthases. Proc Natl Acad Sci U S A
93:1401414019, 1996
3. Anderson DK, Beattie M, Blesch A, Bresnahan J, Bunge M,
Dietrich D, et al: Recommended guidelines for studies of human subjects with spinal cord injury. Spinal Cord 43:453
458, 2005
4. Arnold SA, Hagg T: Anti-inflammatory treatments during the
chronic phase of spinal cord injury improve locomotor function in adult mice. J Neurotrauma 28:19952002, 2011
5. Ates O, Cayli SR, Gurses I, Turkoz Y, Tarim O, Cakir CO,
et al: Comparative neuroprotective effect of sodium channel
blockers after experimental spinal cord injury. J Clin Neurosci 14:658665, 2007
6.Baptiste DC, Fehlings MG: Pharmacological approaches to
repair the injured spinal cord. J Neurotrauma 23:318334,
2006
7.Bergerot A, Shortland PJ, Anand P, Hunt SP, Carlstedt T:
Co-treatment with riluzole and GDNF is necessary for functional recovery after ventral root avulsion injury. Exp Neurol
187:359366, 2004
8.Cadaveira-Mosquera A, Ribeiro SJ, Reboreda A, Prez M,
Lamas JA: Activation of TREK currents by the neuroprotective agent riluzole in mouse sympathetic neurons. J Neurosci
31:13751385, 2011
9. Ceylan S, Ilbay K, Baykal S, Ceylan S, Sener U, Ozmenolu
M, et al: Treatment of acute spinal cord injuries: comparison
of thyrotropin-releasing hormone and nimodipine. Res Exp
Med (Berl) 192:2333, 1992
10. Chen M, Ona VO, Li M, Ferrante RJ, Fink KB, Zhu S, et al:
Minocycline inhibits caspase-1 and caspase-3 expression and
delays mortality in a transgenic mouse model of Huntington
disease. Nat Med 6:797801, 2000
11. Cheung JY, Bonventre JV, Malis CD, Leaf A: Calcium and
ischemic injury. N Engl J Med 314:16701676, 1986
12. De Girolami U, Frosch MP, Tator CH: Regional neuropathology: diseases of the spinal cord and vertebral column, in Graham DI, Lantos P (eds): Greenfields Neuropathology, ed 7.
London: Arnold Publishers, 2002, Vol 1, pp 10631101
13. Dietrich WD: Confirming an experimental therapy prior to
transfer to humans: what is the ideal? J Rehabil Res Dev 40
(4 Suppl 1):6369, 2003
14. Ditor DS, John SM, Roy J, Marx JC, Kittmer C, Weaver LC:
Effects of polyethylene glycol and magnesium sulfate administration on clinically relevant neurological outcomes after
spinal cord injury in the rat. J Neurosci Res 85:14581467,
2007
15. Dobkin BH: Curiosity and cure: translational research strategies for neural repair-mediated rehabilitation. Dev Neurobiol
67:11331147, 2007
16. Doble A: The pharmacology and mechanism of action of riluzole. Neurology 47 (6 Suppl 4):S233S241, 1996
17. Duprat F, Lesage F, Patel AJ, Fink M, Romey G, Lazdunski
M: The neuroprotective agent riluzole activates the two P domain K(+) channels TREK-1 and TRAAK. Mol Pharmacol
57:906912, 2000
18. Faden AI, Jacobs TP, Smith MT: Evaluation of the calcium
channel antagonist nimodipine in experimental spinal cord
ischemia. J Neurosurg 60:796799, 1984

J Neurosurg: Spine / Volume 17 / September 2012

Translational potential of pharmacotherapy for SCI


19. Farber JL: The role of calcium in cell death. Life Sci 29:1289
1295, 1981
20. Fehlings MG, Tator CH, Linden RD: The effect of nimodipine
and dextran on axonal function and blood flow following experimental spinal cord injury. J Neurosurg 71:403416, 1989
21. Festoff BW, Ameenuddin S, Arnold PM, Wong A, Santacruz
KS, Citron BA: Minocycline neuroprotects, reduces microgliosis, and inhibits caspase protease expression early after
spinal cord injury. J Neurochem 97:13141326, 2006
22.Flanders AE, Spettell CM, Tartaglino LM, Friedman DP,
Herbison GJ: Forecasting motor recovery after cervical spinal
cord injury: value of MR imaging. Radiology 201:649655,
1996
23. Ford RW, Malm DN: Failure of nimodipine to reverse acute
experimental spinal cord injury. Cent Nerv Syst Trauma 2:
917, 1985
24.Fu ES, Tummala RP: Neuroprotection in brain and spinal
cord trauma. Curr Opin Anaesthesiol 18:181187, 2005
25. Gambardella G, Collufio D, Caruso GN, Abbate F, German
G, Tomasello F: Experimental incomplete spinal cord injury:
treatment with a combination of nimodipine and adrenaline. J
Neurosurg Sci 39:6774, 1995
26. Gok B, Okutan O, Beskonakli E, Kilinc K: Effects of magnesium sulphate following spinal cord injury in rats. Chin J
Physiol 50:9397, 2007
27. Guha A, Tator CH, Piper I: Effect of a calcium channel blocker on posttraumatic spinal cord blood flow. J Neurosurg 66:
423430, 1987
28. Guha A, Tator CH, Smith CR, Piper I: Improvement in posttraumatic spinal cord blood flow with a combination of a calcium channel blocker and a vasopressor. J Trauma 29:1440
1447, 1989
29. Ha KY, Kim YH, Rhyu KW, Kwon SE: Pregabalin as a neuroprotector after spinal cord injury in rats. Eur Spine J 17:864
872, 2008
30. Haghighi SS, Chehrazi BB, Wagner FC Jr: Effect of nimodipine-associated hypotension on recovery from acute spinal
cord injury in cats. Surg Neurol 29:293297, 1988
31. Hains BC, Waxman SG: Activated microglia contribute to the
maintenance of chronic pain after spinal cord injury. J Neurosci 26:43084317, 2006
32. Hama A, Sagen J: Antinociceptive effect of riluzole in rats
with neuropathic spinal cord injury pain. J Neurotrauma 28:
127134, 2011
33. Holtz A, Nystrm B, Gerdin B: Spinal cord injury in rats: inability of nimodipine or anti-neutrophil serum to improve spinal cord blood flow or neurologic status. Acta Neurol Scand
79:460467, 1989
34. Imamura H, Tator CH: Effect of intrathecal nimodipine on
spinal cord blood flow and evoked potentials in the normal or
injured cord. Spinal Cord 36:497506, 1998
35.Kaptanoglu E, Beskonakli E, Okutan O, Selcuk Surucu H,
Taskin Y: Effect of magnesium sulphate in experimental spinal cord injury: evaluation with ultrastructural findings and
early clinical results. J Clin Neurosci 10:329334, 2003
36. Kaptanoglu E, Beskonakli E, Solaroglu I, Kilinc A, Taskin Y:
Magnesium sulfate treatment in experimental spinal cord injury: emphasis on vascular changes and early clinical results.
Neurosurg Rev 26:283287, 2003
37. Kaynar MY, Erdinler P, Tadayyon E, Belce A, Gmstas K,
Ciplak N: Effect of nimodipine and N-acetylcysteine on lipid
peroxidation after experimental spinal cord injury. Neurosurg Rev 21:260264, 1998
38. Kitzman PH: Effectiveness of riluzole in suppressing spasticity in the spinal cord injured rat. Neurosci Lett 455:150153,
2009
39. Kwon BK, Hillyer J, Tetzlaff W: Translational research in spinal cord injury: a survey of opinion from the SCI community.
J Neurotrauma 27:2133, 2010

J Neurosurg: Spine / Volume 17 / September 2012

40. Kwon BK, Okon E, Hillyer J, Mann C, Baptiste D, Weaver LC,


et al: A systematic review of non-invasive pharmacologic neuroprotective treatments for acute spinal cord injury. J Neurotrauma 28:15451588, 2011
41. Kwon BK, Okon EB, Tsai E, Beattie MS, Bresnahan JC, Magnuson DK, et al: A grading system to evaluate objectively the
strength of pre-clinical data of acute neuroprotective therapies
for clinical translation in spinal cord injury. J Neurotrauma
28:15251543, 2011
42. Kwon BK, Roy J, Lee JH, Okon E, Zhang H, Marx JC, et al:
Magnesium chloride in a polyethylene glycol formulation as a
neuroprotective therapy for acute spinal cord injury: preclinical refinement and optimization. J Neurotrauma 26:1379
1393, 2009
43. Lee JH, Tigchelaar S, Liu J, Stammers AM, Streijger F, Tetzlaff W, et al: Lack of neuroprotective effects of simvastatin
and minocycline in a model of cervical spinal cord injury.
Exp Neurol 225:219230, 2010
44. Lee JS, Han YM, Yoo DS, Choi SJ, Choi BH, Kim JH, et al:
A molecular basis for the efficacy of magnesium treatment
following traumatic brain injury in rats. J Neurotrauma 21:
549561, 2004
45. Lee SM, Yune TY, Kim SJ, Park DW, Lee YK, Kim YC, et
al: Minocycline reduces cell death and improves functional
recovery after traumatic spinal cord injury in the rat. J Neurotrauma 20:10171027, 2003
46. Mami AG, Ballesteros J, Mishra OP, Delivoria-Papadopoulos
M: Effects of magnesium sulfate administration during hypoxia on Ca(2+) influx and IP(3) receptor modification in
cerebral cortical neuronal nuclei of newborn piglets. Neurochem Res 31:6370, 2006
47. Marchand F, Tsantoulas C, Singh D, Grist J, Clark AK, Bradbury EJ, et al: Effects of Etanercept and Minocycline in a rat
model of spinal cord injury. Eur J Pain 13:673681, 2009
48. McAdoo DJ, Hughes MG, Nie L, Shah B, Clifton C, Fullwood
S, et al: The effect of glutamate receptor blockers on glutamate release following spinal cord injury. Lack of evidence
for an ongoing feedback cascade of damage > glutamate
release > damage > glutamate release > etc. Brain Res
1038:9299, 2005
49.Mu X, Azbill RD, Springer JE: Riluzole and methylprednisolone combined treatment improves functional recovery
in traumatic spinal cord injury. J Neurotrauma 17:773780,
2000
50. Mu X, Azbill RD, Springer JE: Riluzole improves measures of
oxidative stress following traumatic spinal cord injury. Brain
Res 870:6672, 2000
51.Ngrdi A, Szab A, Pintr S, Vrbov G: Delayed riluzole
treatment is able to rescue injured rat spinal motoneurons.
Neuroscience 144:431438, 2007
52. Ngrdi A, Vrbov G: The effect of riluzole treatment in rats
on the survival of injured adult and grafted embryonic motoneurons. Eur J Neurosci 13:113118, 2001
53. Ozdemir M, Cengiz SL, Grbilek M, On TC, Ustn ME:
Effects of magnesium sulfate on spinal cord tissue lactate and
malondialdehyde levels after spinal cord trauma. Magnes Res
18:170174, 2005
54. Pintr S, Gloviczki B, Szab A, Mrton G, Ngrdi A: Increased survival and reinnervation of cervical motoneurons
by riluzole after avulsion of the C7 ventral root. J Neurotrauma 27:22732282, 2010
55. Pinzon A, Marcillo A, Quintana A, Stamler S, Bunge MB,
Bramlett HM, et al: A re-assessment of minocycline as a neuroprotective agent in a rat spinal cord contusion model. Brain
Res 1243:146151, 2008
56. Pointillart V, Gense D, Gross C, Bidab AM, Gin AM, Rivel
J, et al: Effects of nimodipine on posttraumatic spinal cord
ischemia in baboons. J Neurotrauma 10:201213, 1993
57. Popovich PG, Lemeshow S, Gensel JC, Tovar CA: Indepen-

205

C. H. Tator et al.
dent evaluation of the effects of glibenclamide on reducing
progressive hemorrhagic necrosis after cervical spinal cord
injury. Exp Neurol 233:615622, 2012
58. Ramer LM, Ramer MS, Steeves JD: Setting the stage for functional repair of spinal cord injuries: a cast of thousands. Spinal Cord 43:134161, 2005
59. Ross IB, Tator CH: Further studies of nimodipine in experimental spinal cord injury in the rat. J Neurotrauma 8:229
238, 1991
60. Ross IB, Tator CH: Spinal cord blood flow and evoked potential responses after treatment with nimodipine or methylprednisolone in spinal cord-injured rats. Neurosurgery
33:470477, 1993
61. Ross IB, Tator CH, Theriault E: Effect of nimodipine or methylprednisolone on recovery from acute experimental spinal
cord injury in rats. Surg Neurol 40:461470, 1993
62. Saganov K, Orendcov J, Czkov D, Vanick I: Limited
minocycline neuroprotection after balloon-compression spinal cord injury in the rat. Neurosci Lett 433:246249, 2008
63. Schwartz G, Fehlings MG: Evaluation of the neuroprotective
effects of sodium channel blockers after spinal cord injury:
improved behavioral and neuroanatomical recovery with riluzole. J Neurosurg 94 (2 Suppl):245256, 2001
64. Simard JM, Tsymbalyuk O, Ivanov A, Ivanova S, Bhatta S,
Geng Z, et al: Endothelial sulfonylurea receptor 1-regulated
NC Ca-ATP channels mediate progressive hemorrhagic necrosis following spinal cord injury. J Clin Invest 117:2105
2113, 2007
65. Simard JM, Woo SK, Norenberg MD, Tosun C, Chen Z, Ivanova S, et al: Brief suppression of Abcc8 prevents autodestruction of spinal cord after trauma. Sci Transl Med 2:28ra29,
2010
66. Solaroglu I, Kaptanoglu E, Okutan O, Beskonakli E, Attar A,
Kilinc K: Magnesium sulfate treatment decreases caspase-3
activity after experimental spinal cord injury in rats. Surg
Neurol 64 (Suppl 2):S17S21, 2005
67. Stirling DP, Khodarahmi K, Liu J, McPhail LT, McBride CB,
Steeves JD, et al: Minocycline treatment reduces delayed oligodendrocyte death, attenuates axonal dieback, and improves
functional outcome after spinal cord injury. J Neurosci
24:21822190, 2004
68. Stutzmann JM, Pratt J, Boraud T, Gross C: The effect of riluzole on post-traumatic spinal cord injury in the rat. Neuroreport 7:387392, 1996
69. Szer T, Coskun E, Islekel H, Tahta K: Neuroprotective effect
of magnesium on lipid peroxidation and axonal function after
experimental spinal cord injury. Spinal Cord 37:480484,
1999
70. Tan AM, Zhao P, Waxman SG, Hains BC: Early microglial
inhibition preemptively mitigates chronic pain development
after experimental spinal cord injury. J Rehabil Res Dev
46:123133, 2009
71.Tator CH: Review of treatment trials in human spinal cord
injury: issues, difficulties, and recommendations. Neurosurgery 59:957987, 2006
72. Tator CH, Fehlings M: Clinical trials in spinal cord injury, in
Biller J, Bogousslavsky J (eds): Clinical Trials in Neurologic
Practice, Blue Books of Practical Neurology, ed 2. Boston:
Butterworth-Heinemann, 2001, pp 99120
73. Tator CH, Fehlings MG: Review of the secondary injury the-

206

ory of acute spinal cord trauma with emphasis on vascular


mechanisms. J Neurosurg 75:1526, 1991
74. Tator CH, Poon P: Acute clip impact-compression model, in
Chen J, Xu XM, Xu ZC, et al (eds): Animal Models of Acute
Neurological Injuries. Totowa, NJ: Humana Press, 2009, pp
449460
75. Teng YD, Choi H, Onario RC, Zhu S, Desilets FC, Lan S, et al:
Minocycline inhibits contusion-triggered mitochondrial cytochrome c release and mitigates functional deficits after spinal
cord injury. Proc Natl Acad Sci U S A 101:30713076, 2004
76. Tikka T, Fiebich BL, Goldsteins G, Keinanen R, Koistinaho
J: Minocycline, a tetracycline derivative, is neuroprotective
against excitotoxicity by inhibiting activation and proliferation of microglia. J Neurosci 21:25802588, 2001
77. Ueno T, Ohori Y, Ito J, Hoshikawa S, Yamamoto S, Nakamura
K, et al: Hyperphosphorylated neurofilament NF-H as a biomarker of the efficacy of minocycline therapy for spinal cord
injury. Spinal Cord 49:333336, 2011
78. Ustn ME, Grbilek M, Ak A, Vatansev H, Duman A: Effects
of magnesium sulfate on tissue lactate and malondialdehyde
levels in experimental head trauma. Intensive Care Med 27:
264268, 2001
79. Vink R, Cernak I: Regulation of intracellular free magnesium
in central nervous system injury. Front Biosci 5:D656D665,
2000
80. Wells JE, Hurlbert RJ, Fehlings MG, Yong VW: Neuroprotection by minocycline facilitates significant recovery from
spinal cord injury in mice. Brain 126:16281637, 2003
81. Wiseman DB, Dailey AT, Lundin D, Zhou J, Lipson A, Falicov
A, et al: Magnesium efficacy in a rat spinal cord injury model.
Laboratory investigation. J Neurosurg Spine 10:308314,
2009
82. Yong VW, Power C, Forsyth P, Edwards DR: Metalloproteinases in biology and pathology of the nervous system. Nat Rev
Neurosci 2:502511, 2001
83. Yrjnheikki J, Tikka T, Keinnen R, Goldsteins G, Chan PH,
Koistinaho J: A tetracycline derivative, minocycline, reduces
inflammation and protects against focal cerebral ischemia
with a wide therapeutic window. Proc Natl Acad Sci U S A
96:1349613500, 1999
84. Yune TY, Lee JY, Jung GY, Kim SJ, Jiang MH, Kim YC, et
al: Minocycline alleviates death of oligodendrocytes by inhibiting pro-nerve growth factor production in microglia after
spinal cord injury. J Neurosci 27:77517761, 2007
85. Zang DW, Cheema SS: Leukemia inhibitory factor promotes
recovery of locomotor function following spinal cord injury in
the mouse. J Neurotrauma 20:12151222, 2003
86. Zhang L, Rzigalinski BA, Ellis EF, Satin LS: Reduction of
voltage-dependent Mg2+ blockade of NMDA current in mechanically injured neurons. Science 274:19211923, 1996
Manuscript submitted January 30, 2012.
Accepted May 3, 2012.
Please include this information when citing this paper: DOI:
10.3171/2012.5.AOSPINE12116.
Address correspondence to: Charles H. Tator, M.D., Ph.D.,
Toronto Western Hospital, 399 Bathurst Street, Suite 4W-433,
Toronto, Ontario M5T 2S8, Canada. email: charles.tator@uhn.on.ca.

J Neurosurg: Spine / Volume 17 / September 2012

Ceylan et al.,
1992

Animal model:
Wistar rats
N = 30
Sex: NR
Age: NR
Wt: 180250 g
SCI model:
Extradural clip compression injury
of cord at T-9 w/ a
modified aneurysm
clip exerting a force
of 50 g, causing
complete paraplegia

SCI + saline (control)


SCI + nimodipine +
dextran-40
SCI + TRH +
dextran-40
10 rats/group

Authors & Year Animal & Injury Models Experimental Groups

Intervention Details
Route: IP inj
Dosage & timing:
Control: received only saline
Nimodipine: 0.02 mg/kg nimo dipine & dextran 40 (3 ml)
IV 1 hr after trauma
TRH: 2 mg/kg of TRH &
dextran-40 (3 ml), then 1
mg/kg TRH IV for 4 hrs at
1-hr intervals
Animals killed: at completion of
experimental protocol (5 hrs
postinjury), animals were
killed by an IV infusion of
Somlethal

TABLE 16: Characteristics of SCI animal studies using nimodipine*

Physiological Parameters:
Mean preinjury parameters including MABP, hematocrit, HR, pH, PO2, PCO2,
Na+, and K+ were not statistically different (p >0.05) among the groups
After SCI, MABP (t: 6.986), HR (t: 7.373), & hematocrit (t: 4.29) in all groups
& PO2 (t: 5.06), PCO2 (t: 11.588), Na+ (t: 3.993), & K+ (t: 2.301) in the TRH
& dextran-treated group were significantly decreased or increased
(dt: 2.23, p <0.05)
At 1 hr after TRH & dextran infusion, the MABP & HR were significantly
increased (p <0.05; t: 12.96, t: 6.429)
SSEPs:
No return of SSEPs in controls
In the nimodipine & dextran-treated group, 8 of 10 animals had no significant
return of SSEP; the remaining 2 demonstrated minimal return of SSEP
In the TRH-treated group, 8 of 10 animals had return of SSEPs (p <0.01);
3 of these animals had return of SSEPs to a nearly normal pattern; 5 had
return of SSEPs w/ delayed latencies & decreased amplitudes; the re maining 2 demonstrated no significant return of SSEPs
Histopathological Scores:
Control vs Nimodipine vs TRH
Grade 0: NR vs NR vs NR
Grade 1: 2 vs 2 vs 2
Grade 2: 3 vs 4 vs 5
Grade 3: 5 vs 4 vs 3
Scores in all groups did not differ significantly (p >0.05), although scores for
TRH-treated animals were higher than those in the nimodipine or control
groups
There was serious hemorrhage in the central & peripheral gray matter in the
control group
In the nimodipine & TRH groups, petechial hemorrhages were identified in
the peripheral gray & white matter
TRH Tx significantly improved SSEPs & MABPs, whereas nimodipine Tx had
no effect on these variables (p <0.01)

Reported Outcomes

(continued)

Comments

Translational potential of pharmacotherapy for SCI

J Neurosurg: Spine / Volume 17 / September 2012

207

208
Route: IV
Timing & dosage:
Nim 1: 1 g/kg/min, beginning
15 min after termination of
aortic occlusion, w/ infusion
continued for 1 hr
Nim 2: 2 g/kg/min, beginning
15 min before occlusion, w/
infusion continued at this
rate for 2 hrs
Control: saline beginning 15
min before occlusion &
continuing for 2 hrs
Animals killed: 48 hrs after
occlusion, the animals were
killed by admin of sodium
pentobarbital
Route: IP infusion
Timing: postinfusion SCBF &
EP recordings were made
immediately after the infu sion & at 1 & 2 hrs after the
cessation of drug delivery
Drug delivery: 1 hr post-SCI;
infusion continued for 1 hr
Dosage:
0.02 mg/kg nimodipine
0.05 mg/kg nimodipine
Animals killed: NR

SCI + nimodipine 1
(Nim 1) (n = 10)
SCI + nimodipine 2
(Nim 2) (n = 7)
SCI + saline (control)
(n = 15)

SCI + saline (placebo


& saline)
SCI + dextran-40
(placebo &
dextran-40)
SCI + nimodipine 0.02
mg/kg + saline
SCI + nimodipine 0.02
mg/kg + dextran40
SCI + nimodipine 0.05
mg/kg + saline
SCI + nimodipine 0.05
mg/kg + dextran40
5 rats/group

Animal model:
NZ albino rabbits
N = 32
Sex: 100% M
Age: NR
Wt: 2.0 0.25 kg
SCI model:
Aorta occlusion for
20 min

Animal model:
Wistar rats
N = 30
Sex: NR
Age: adult
Wt: 341 22 g
SCI model:
Laminectomy from C-6
to T-2, & each rat
received a 1-min ex tradural clip com pression injury of
cord at T-1 w/ a
modified aneurysm
clip exerting a force
of 53 g, causing
complete paraplegia

Fehlings et al.,
1989

Intervention Details

Faden et al.,
1984

Authors & Year Animal & Injury Models Experimental Groups

TABLE 16: Characteristics of SCI animal studies using nimodipine* (continued)

Neurological Scores:
No significant differences btwn the control group & Nim 1 (p >0.05)
Although neurological scores among all groups appeared to be virtually
identical at 2 hrs postischemia, scores at 24 & 48 hrs postischemia
appeared, if anything, to be somewhat worse in the nimodipine-treated
groups
Histopathological Scores:
No significant differences among the groups (p >0.05)
On gross inspection, the spinal cords displayed myelomalacia to varying
degrees w/in the lower spinal cord from T-12 to the conus medullaris
Microscopic examination also revealed varying degrees of cavitary necrosis
Pathological changes in all cases involved gray matter, but not white matter:
the anterior horn was predominantly affected, w/ relative sparing of the
intermediate gray matter & sparing of the posterior gray horn
Neurological scores correlated well w/ the histopathological scores (Spear mans rho = 0.994, p <0.01)
Nimodipine at doses shown to be effective in improving cerebral blood flow &
in dilating central blood vessels failed to improve either the histopathologi cal changes or the functional deficit caused by temporary aortic occlusion
Physiological Parameters:
Mean preinjury scores including the MABP, hematocrit, HR, pH, PO2, &
PCO2 were not significantly different (p >0.05) among the groups
pH, PO2, & PCO2 remained constant throughout the expt
After SCI there was a significant decrease in the MABP, similar in all groups;
however, the MABP was significantly increased following infusion in
Groups 1, 2, & 4
At 1 hr after drug infusion, the MABP was again similar among the 6 Tx
groups
Although the postinjury & postinfusion hematocrit was similar among the
groups, at 1 hr postinfusion the mean hematocrit of the 3 groups that re ceived dextran alone or in combination w/ nimodipine was significantly
lower (p <0.02) than that of the other 3 groups
SCBF:
Preinjury SCBF at T-1 was similar among the 6 Tx groups
SCBF at T-1 was influenced by SCI (decreased) & drug infusion (p <0.0001)
SCBF in Group 4 was significantly greater than that of the other 5 groups
By 2 hrs postinfusion, SCBF at T-1 was again similar in all groups
SCBF at C-6 was significantly increased in Groups 2, 4, & 6; no significant
difference in the postinfusion SCBF among these groups
SCBF at T-10 did not change significantly in any group at any point during
the course of the expt

Reported Outcomes

(continued)

Only reported results


in Nim 1 group
because Nim 2
group had nimo dipine Tx prior to
occlusion

Comments

C. H. Tator et al.

J Neurosurg: Spine / Volume 17 / September 2012

Ford & Malm,


1985

Fehlings et al.,
1989
(continued)

SCI, no Tx (control)
Animal model:
SCI + nimodipine
Cats
11 cats/group
N = 22
Sex: 100% M
Age: adult
Wt: 3.06.3 kg
SCI model:
Modified Allen tech nique: a 10-g iron wt
was dropped
through a brass
tube onto L-1,
causing complete
paraplegia

Authors & Year Animal & Injury Models Experimental Groups

Route: IV
Timing:
30 min after SCI, Tx was
started in the Tx group &
continued for 24 hrs
Followed by oral admin of
5-mg capsules every 8 hrs
for 12 doses
Dosage: nimodipine 1 g/kg/
min
Animals killed: 6 wks postinjury

Intervention Details

TABLE 16: Characteristics of SCI animal studies using nimodipine* (continued)

Electrophysiological Analysis:
SCI resulted in a significant reduction in the amplitude of each peak in all Tx
groups
Improvement in SSEPs shown w/ Group 4
Histological Assessment & Quantitation of Cord Hemorrhages:
The total cord vol was not significantly different among groups
Neither the total vol of hemorrhage at the injury site nor the % hemorrhage
by vol varied significantly among Tx groups, indicating that pharmacologi cal improvement of posttraumatic SCBF did not exacerbate the degree of
hematomyelia
An increase in posttraumatic SCBF can significantly improve the function of
injured spinal cord axons, & strongly implicates posttraumatic ischemia in
pathogenesis of acute SCI
Physiological Parameters:
Control vs Tx
Wt (kg): 4.4 1.1 vs 4.5 0.4
PaO2 (mm Hg): 155 18 vs 143 21
PaCO2 (mm Hg): 30 2 vs 30 2
MAP (mm Hg): 90 14 vs 89 12
Peak increase in MAP (% of preinjury MAP): 39 18 vs 41 30
Esophageal temperature (C): 37.5 0.9 vs 37.6 0.6
Time anvil under cord: 3.7 2.8 vs 3.1 0.9 sec
No significant difference btwn groups for any parameter (p >0.10)
30 min after nimodipine infusion, the average MAP was 15 19% lower than
the immediate pre-Tx value (significant difference; p <0.05)
Average MAP in control increased 2 13% over a comparable period (differ ence not significant; p >0.75)
Final Motor Recovery Scores (6 wks post-SCI):
Control vs Tx
Grade 0 (unable to walk): 11 vs 9
Grade 1 (walks a few steps only): 0 vs 2
Grade 2 (unlimited weak gait): 0 vs 0
Grade 3 (walks w/ slight weakness): 0 vs 0
Grade 4 (jumps up 3 ft): 0 vs 0
Grade 5 (normal): 0 vs 0
No control animal regained the ability to walk even a few steps during the
6-wk assessment period
2 of the treated cats recovered sufficiently to elevate their hindlimbs & take a
few steps (failed to reach statistical significance; G-statistic, 0.05 <
p <0.10)
The gray matter in all cords was completely destroyed at the site of impact, &
all cords had central cavitation
Nimodipine Tx did not significantly increase white matter preservation
(p >0.10)

Reported Outcomes

(continued)

Comments

Translational potential of pharmacotherapy for SCI

J Neurosurg: Spine / Volume 17 / September 2012

209

210

Gambardella et Animal model:


Albino NZ rabbits
al., 1995
N = 80
Sex: either sex
Age: NR
Weight: 11.3 kg
SCI model:
Laminectomy per formed at T12L1,
modified Sujita vas cular clip used to ex pose the spinal cord
so a 60.564.2%
reduction in diameter
was caused (T12L1)

No SCI or Tx (control,
Group I)
SCI + saline (Group
II)2 subgroups
(8 each):
IIa: spinal cord com pression induced
for 2 min
IIb: induced for 5 min
SCI + nimodipine +
adrenaline (Group
III)2 subgroups
(n = 8 each; same
as Group II):
IIIa: 2-min compression
IIIb: 5-min compression
SCI + nimodipine
alone (Group IV)
2 subgroups (n = 8
each; same as II):
IVa: 2-min compression
IVb: 5-min compression
SCI + adrenaline
alone (Group V)
2 subgroups (n = 8
each, same as II):
Va: 2-min compression
Vb: 5-min compression
16 rabbits/group

Authors & Year Animal & Injury Models Experimental Groups

Reported Outcomes
Arterial BP:
Spinal cord compression decreased arterial BP significantly, to 81 3.5 mm
Hg in all animals
Normal saline did not restore BP to control levels
Tx w/ nimodipine alone decreased BP further, by 2040 mm Hg
Tx w/ adrenaline only or in combination w/ nimodipine maintained BP w/in
physiological limits
Motor Responses:
Degrees (Tarlov scale) After Spinal Compression1 hr vs 24 hrs vs 48 hrs
Group I: 0 vs 0 vs 0
Group IIa: 5 vs 4 vs 43
Group IIb: 5 vs 54 vs 54
Group IIIa: 5 vs 43 vs 41
Group IIIb: 5 vs 42 vs 42
Group IVa: 5 vs 5 vs 54
Group IVb: 5 vs 5 vs 5
Group Va: 5 vs 54 vs 43
Group Vb: 5 vs 54 vs 54
In all animals, spinal cord compression for 2 & 5 min induced a 5th-degree
motor deficit
Tx w/ saline slightly improved the motor deficit
Tx w/ nimodipine & adrenaline (Group III) showed a trend in the improvement
of motor deficit
Rabbits treated w/ adrenaline alone showed a better recovery in comparison
w/ rabbits treated w/ saline alone
Morphometric Studies:
The mean no. of myelinated axons in this unit area (NA/UA) was 90 in Group
I (control group)
The axonal number was significantly (p <0.050.001) reduced in all groups
subjected to SCI compared to that of control group

Intervention Details
Route: IV
Timing & dosage: Tx started
immediately postinjury
Group I: no Tx
Group II: 40 drops/min for 2
hrs (saline)
Group III: 1.5 g/kg/min IV for
2 hrs (nimodipine); arterial
BP was maintained near
control levels by infusion of
adrenaline (1:100,000 solu tion in saline; 40 drops/min)
Group IV: 1.5 g/kg/min
nimodipine solution alone
administered for 2 hrs
Group V: 1:100,000 solution of
adrenaline in saline admin istered for 2 hrs
Animals killed: NR

TABLE 16: Characteristics of SCI animal studies using nimodipine* (continued)

(continued)

Comments

C. H. Tator et al.

J Neurosurg: Spine / Volume 17 / September 2012

Animal model:
Wistar rats
N = 15
Sex: 100% M
Age: NR
Wt: 400500 g
SCI model:
Clip compression injury
model used to de liver a 53-g injury to
T-1 spinal segment
for 1 min; blunt den tal probe used below
exiting C-8 nerve
roots

SCI + saline
Animal model:
SCI + nimodipine
Cats
5 cats/group
N = 10
Sex: NR
Age: adult
Wt: 34 kg
SCI model:
A 100 g/cm impact in jury (20-g weight,
dropped 5 cm) was
delivered at the T-13
vertebral level by us ing a modification of
Allen trauma device

Guha et al.,
1989

Haghighi et al.,
1988

SCI + saline
SCI + adrenaline
SCI + adrenaline +
nimodipine
5 rats/group

Authors & Year Animal & Injury Models Experimental Groups


Route: IV through femoral
veins
Timing:
Measurements taken 30 min
preinjury, at injury, & 30 min
postinjury
Tx given for 1 hr, starting 2 hrs
postinjury
Dosage:
Adrenaline: 2.5 ml/hr of
1/100,000 dilution
Nimodipine: 1.5 g/kg/min
Vol of infusion of all 3 Tx
groups was approximately
equivalent (2.5 ml) over the
1-hr Tx period
Animals killed: after comple tion of last flow, the animals
were killed w/ a bolus of
potassium chloride
Route: IV
Timing:
Tx given 5 min posttrauma
SEPs were recorded immedi ately posttrauma, & subse quent recordings were
made every 5 min up to
2 hrs
Dosage:
Nimodipine: 10-g/kg bolus
given 5 min after trauma,
followed by a 1-g/kg/min
infusion for 2 hrs given
through an infusion pump
Control group received equal
vols of saline solution
Animals killed: at the termina tion of each expt, the cat
was injected w/ a lethal dose
of sodium pentobarbital &
potassium chloride solutions

Intervention Details

TABLE 16: Characteristics of SCI animal studies using nimodipine* (continued)

J Neurosurg: Spine / Volume 17 / September 2012


MAP Activity:
Following the wt drop on the spinal cord, there was a transient surge of MAP
in all animals
The nimodipine-treated cats showed a markedly prolonged systemic hypo tension after the start of nimodipine infusion; this hypotension was signifi cant (p <0.05) at 30, 60, & 90 min
The maximum drop in MAP occurred w/in the first 30 min of onset of infusion
The electrocardiograms remained unchanged in all animals; however,
transient episodes of cardiac arrhythmias were noticed in 2 cats at the
later part of the nimodipine Tx
SEP Activity:
The average time course for the return of the cephalad SEPs was 50 15
min in the saline group
In the nimodipine-treated cats, 3 did not show return of the SEPs up to 4 hrs
posttrauma
In the remaining 2 cats, SEPs returned on an average of 80 10 min
In this model, nimodipine Tx had a deleterious effect on the spinal cord re covery due to the significant associated hypotension
It is likely that marked hypotension in the case of traumatic loss of autoregu lation overrides the expected nimodipine-related increase in SCBF, w/
resultant additional ischemic damage

Changes in mSAP & SCBF:


Significant improvement in postinjury mSAP after adrenaline or the combina tion of nimodipine & adrenaline
No significant difference btwn the 2 Tx groups (p >0.05)
In contrast, animals given saline at an identical infusion rate had a further
deterioration in mSAP compared to postinjury values
There were no significant differences among the 3 Tx groups in postinjury
SCBF at either C-6 or T-1 spinal segments
The nimodipine + adrenaline group had the highest (p <0.05) Tx SCBF at
both C-6 & T-1 (62.0 3.8 & 26.1 3.3 ml/100 g/min, respectively)
Area & Vol of Intramedullary Hemorrhage:
Where the hemorrhage was maximum, the % of hemorrhage did not differ
significantly (p >0.05) among the 3 Tx groups, although the saline group
always had the lowest values
However, at areas remote from the site of injury, the saline group in some
sections had significantly less hemorrhage
Although the saline-treated animals had a slightly lower vol of hemorrhage
& % vol of hemorrhage, the values were not significantly different (p
>0.05) from the other 2 Tx groups

Reported Outcomes

(continued)

Comments

Translational potential of pharmacotherapy for SCI

211

212

Imamura &
Tator, 1998

Holtz et al.,
1989

SCI + saline + NSS


(before SCI;
controls)
SCI + nimodipine
SCI + ANS (before
SCI)
10 rats/group

Route: IV
Timing & dosage:
Controls: IV infusion of 0.4 ml
saline 60 min postinjury; 5
of these animals were also
given 3 ml of NSS IP 12 hrs
preinjury
Nimodipine: infusion of 1.5 g/
kg/min in a total vol of 0.4
ml of saline over 4 hrs,
starting 60 min postinjury
ANS: 3 ml of sheep ANS IP 12
hrs before injury
Animals killed: rats were killed
on Day 4

Intervention Details

Reported Outcomes

Physiological Variables:
No significant differences btwn the groups in any of the measured variables,
except that after laminectomy MABP was higher in the nimodipine-treated
group
By the time of determination of SCBF, all measured variables were similar
btwn the groups
Tx w/ ANS resulted in a reduction of the number of circulating PMNLs
Late increase in the no. of peripheral PMNLs in both ANS- & NSS-treated
groups
Control animals treated w/ NSS showed no change in the no. of circulating
PMNLs
The no. of mononuclear cells was slightly decreased by Tx w/ ANS & also w/
NSS
Neurological Dysfunction:
1 day after compression, the 3 groups exhibited equal impairment in motor
performance, & the mean capacity angle was significantly reduced, to
close to 32
Partial restoration was observed in all 3 groups during the following days, &
on Day 4 the capacity angle was close to 43
SCBF:
No differences btwn the groups either in gray or in white matter SCBF
Animal model:
Uninjured groups:
Route: IT infusion
Physiological Parameters:
Wistar rats
No SCI + placebo
Timing & dosage:
Uninjured Groups
N = 25
(PEG-400, ethanol, Placebo or nimodipine was in- pH, PaCO2, PaO2, hematocrit, & rectal temperature did not show any signifiSex: 100% M
sodium citrate, &
fused into the subarachnoid cant changes at any time of measurement
Age: NR
anhydrous citric
space for 30 min in 0.5 ml
Only the MABP in the 0.2 mg/kg nimodipine group showed a significant deWt: 351497 g
acid)
diluent, beginning 30 min
crease during infusion (p = 0.029) & postinfusion (p = 0.030)
SCI model:
No SCI + nimodipine after the baseline SCBF
Injured Groups
Laminectomy from C-1 0.05 mg/kg
was obtained for groups
pH & PaO2 showed no significant change during the expt
to T-1; acute cord
No SCI + nimodipine w/o SCI
PaCO2 in the nimodipine group significantly increased 150 min after injury
clip compression
0.2 mg/kg
SCI groups had placebo or
(p = 0.0395); however, it was still w/in normal limits
injury made at T-1 w/ 5 rats/group
nimodipine infused for
Hematocrit decreased serially in both groups, although the differences btwn
a 35-g clip for 1 min Injured groups:
30 min beginning 60 min
baseline & subsequent time intervals reached significance only in the
SCI + placebo (0.5 ml) postinjury
nimodipine groups (30 min postinjury, p = 0.0002; 90 min, p = 0.0002; &
SCI + nimodipine 0.05 SCBF was measured at 90
150 min, p = 0.0048)
mg/kg (0.5 ml)
min (at the completion of
MABP decreased significantly postinjury in both injured groups: in the place5 rats/group
infusion) & 150 min (1 hr
bo group there was no further decrease after the placebo infusion, where after completion of infusion) as in the nimodipine group the MABP continued to decline after the infupostinjury
sion of nimodipine (p = 0.0142); however, 60 min after infusion of nimodipAnimals killed: NR
ine, MABP recovered to the preinfusion level postinjury (p = 0.0768), but
did not return to baseline

Animal model:
Sprague-Dawley rats
N = 30
Sex: 100% M
Age: NR
Wt: 330400 g
SCI model:
Laminectomy at T78,
animals fixed in a
specially designed
compression device,
plate was applied
to exposed dura, &
loaded w/ 35 g for
5 min

Authors & Year Animal & Injury Models Experimental Groups

TABLE 16: Characteristics of SCI animal studies using nimodipine* (continued)

(continued)

Comments

C. H. Tator et al.

J Neurosurg: Spine / Volume 17 / September 2012

J Neurosurg: Spine / Volume 17 / September 2012

Kaynar et al.,
1998

Imamura &
Tator, 1998
(continued)

Animal model:
Sprague-Dawley rats
N = 40
Sex: 100% F
Age: NR
Wt: 230310 g
SCI model:
Laminectomy at C7T1;
clip was applied ex tradurally to spinal
cord & compressed
cord for 30 sec in
Groups 2, 3, & 4

No SCI, no Tx: sham


(Group 1)
SCI, no Tx (Group 2)
SCI + nimodipine
(Group 3)
SCI + N-acetylcyste ine (Group 4)
10 rats/group

Authors & Year Animal & Injury Models Experimental Groups

Reported Outcomes

SCBF:
Uninjured Groups
No significant changes in any of the groups during placebo or nimodipine
infusion or after infusion
Injured Groups
The decline in SCBF was not significant 30 min postinjury in the placebo
group (p = 0.094) or in the nimodipine 0.05 mg/kg group (p = 0.082), but
the decline was significant during the infusion 90 min postinjury in the
placebo & nimodipine groups (p = 0.0034 & p = 0.003)
1 hr after completion of the infusion (150 min postinjury) SCBF remained low,
w/o evidence of recovery in either group
EPs:
SSEPs in the Uninjured Groups
No significant difference among the 3 groups at any time of recording
(p >0.05), although there was a large variability btwn individual animals
CEPs in the Uninjured Groups
No significant difference among the 3 groups at any time of recording
SSEPs in the Injured Groups
The SSEPs disappeared immediately postinjury in all rats, then reappeared
in 5 rats (2 in the placebo & 3 in the nimodipine group) 30 min postinjury,
although the amplitudes were decreased & the latencies were increased
significantly (p <0.05)
The amplitude of the SSEPs decreased significantly immediately postinjury
& showed no recovery thereafter in either group (p <0.0001)
CEPs in the Injured Groups
In the placebo group, the CEPs in 3 rats disappeared immediately postinjury
In the nimodipine group, the CEPs in 4 rats remained immediately postinjury,
although the amplitude decreased significantly (p <0.0001)
There was no significant difference in the amplitude changes btwn the 2
groups at any time (p >0.05)
Route: IP inj
MDA Results:
Timing & dosage:
The effect of Tx was evaluated by assessing MDA formation in the spinal
Group 3: single dose of 0.05
cord at 1 hr postinjury
mg/kg immediately after
The MDA content was higher in Group 2 (w/o Tx) than in Group 1 (shamSCI
operated)
Group 4: single dose of 163
The difference btwn Groups 2 (no Tx) & 3 (nimodipine Tx) or Groups 2 & 4
mg/kg immediately after
(N-acetylcysteine Tx) was not statistically significant
SCI
A single dose of nimodipine & N-acetylcysteine had no effect on MDA formaAnimals killed: rats were killed tion in the spinal cord homogenates
w/ large doses of sodium
pentothal at 1 hr after clip
application

Intervention Details

TABLE 16: Characteristics of SCI animal studies using nimodipine* (continued)

(continued)

Comments

Translational potential of pharmacotherapy for SCI

213

214

Pointillart et al., Animal model:


Baboons
1993
N = 13
Sex: NR
Age: adult
Wt: 10 0.3 kg
SCI model:
Laminectomy at L-5;
Fr-4 Fogarty cathe ter inserted in epi dural space; trauma
provoked by inflating
catheter balloon
after injecting 2 ml
of Ringer solution
to develop a con trolled pressure for
5 sec

SCI + nimodipine
SCI + saline (placebo)
5 baboons/group (3 of
13 died before Day
8 & were not
included)

Authors & Year Animal & Injury Models Experimental Groups

Reported Outcomes
SCBF:
Preinjury SCBF was similar in the placebo & treated groups (40.9 16.3
ml/100 g/min vs 39.8 15.9 ml/100 g/min)
30 min posttrauma, SCBF increased 70.5 60.6% in the placebo & 85.6
60.3% in the nimodipine group; no significant difference
90 min posttrauma, SCBF decreased 90 9.7% in the placebo group & only
39.4 18.9% in the nimodipine group; values were significantly different
(p <0.05)
At 150 & 210 min posttrauma, SCBF decreased to unmeasurable values in
the placebo group, but only decreased 37.6 10.5% & 38.8 8.8% in the
treated group
On Day 8, SCBF was still low in the placebo group: a decrease of 83.8
6.4% was recorded; SCBF decreased only 36 10.2% in the nimodipine
group (p <0.05), significantly different from the placebo group, indicating
that SCBF was partially restored
SEPs:
Following trauma, SEPs showed no detectable peak in either group
On Day 8, the placebo group still revealed no SEPs, but in 2 cases in the
treated group had a partial recovery
MRI Data:
On the 1st day, MRI data were similar in the 2 groups (no significant differences)
Histological Data:
Lesions consisted mainly of interstitial bleeding, ranging from slight to de structive hemorrhagic zones
Lesions were located in both the gray & white matter
In the untreated group, average length of the lesion was 45.2 6.1 mm in
the white matter & 31.6 3.1 mm in the gray matter
In the treated group, average length of the lesion was 26.4 4.3 mm in the
white matter & 14 4.3 mm in the gray matter
These 2 differences were significant (p <0.05)

Intervention Details
Route: IP inj
Timing & dosage: infusion be gan immediately postinjury
Nimodipine: 0.02 mg/kg/hr for
2 hrs, 0.04 mg/kg/hr there after for 8 days
Placebo: saline w/ same infu sion vol
Animals killed: after measure ments on Day 8, baboons
were killed by an overdose
of anesthetic

TABLE 16: Characteristics of SCI animal studies using nimodipine* (continued)

(continued)

Comments

C. H. Tator et al.

J Neurosurg: Spine / Volume 17 / September 2012

J Neurosurg: Spine / Volume 17 / September 2012

Ross & Tator,


1993

Ross & Tator,


1991

SCI + 0 mg/kg of ni modipine (control


placebo)
SCI + nimodipine
0.005 mg/kg
SCI + nimodipine 0.01
mg/kg
SCI + nimodipine
0.025 mg/kg
SCI + nimodipine 0.05
mg/kg
5 rats/group

Route: IV
Timing & dosage: 2 hrs postin jury, 1 ml of nimodipine
solution or placebo was
infused over 35 min
Animals killed: NR

Intervention Details

Reported Outcomes

Physiological Parameters:
Although MABP dropped dramatically postinjury, from 117.6 2.7 mm Hg to
56.6 1.6 mm Hg (p <0.0001), there was no significant difference in
MABP btwn the experimental groups at any point in time (p >0.05)
SCBF:
SCBF was not improved by nimodipine at any dose
Preinjury SCBF varied significantly btwn experimental groups, but both the
postinjury pre-Tx & post-Tx SCBF values did not
Nimodipine infusion did not lead to increased SCBF in any experimental
group; in fact there was a further decline in SCBF after nimodipine infu sion in all groups (no significant difference among the 5 Tx groups in
these decreases in SCBF w/ the drug infusions)
EPs:
No difference across experimental groups in the preinjury MEP D wave amp litudes or in the preinjury SSEP N2 wave amplitudes; these waves were
completely abolished by the injury in all animals, & no return was
observed after nimodipine infusion in any animal
Histological Assessment:
In all animals, hemorrhage extending through the gray matter w/ frank areas
of necrosis were noted at the compression site
Changes in the white matter were less marked
No major difference in the severity of the changes btwn the groups
SCI + nimodipine
Animal model:
Route: IV inj
SCBF:
(0.02 mg/kg/hr)
Wistar rats
Timing & dosage:
Neither nimodipine nor MP was superior to placebo at augmenting SCBF at
(Group A)
N = 30
2 min postinjury, each rat re either 1 hr (df = 2, 27; F = 1.96; p = 0.16) or 2.5 hrs (df = 2, 27; F = 0.36;
Sex: 100% M
SCI + MP (5.4 mg/kg/ ceived a 0.6-ml bolus of
p = 0.70) postinjury
Age: adult
hr) (Group B)
25% human serum albumin Preinjury SCBF did not vary significantly btwn the experimental groups
Wt: 385650 g
SCI + placebo (PEG- 5 min postinjury, rats received EPs:
400 & ethanol)
SCI model:
1 ml of placebo, or in the
No EP responses were observed in any animal at any time after injury
Laminectomy made at (Group C)
MP-treated animals, 30 mg/ Histological Assessment:
C7T1 & T910;
10 rats/group
kg of MP dissolved in 1 ml In all rats, hemorrhage extending through the gray matter w/ frank areas of
injury made at T-1 by
of placebo
necrosis was noted at the compression site
compression of
Cont drug infusions (0.7 ml/hr) Changes in white matter were less marked
spinal cord w/ a 52-g
starting 15 min postinjury
There was no major gross difference in the severity of the changes btwn the
curved aneurysm
Infusions continued for 3 hrs
groups
clip for 60 sec
after SCI
Animals killed: NR

Animal model:
Wistar rats
N = 25
Sex: 100% M
Age: adult
Wt: 350550 g
SCI model:
Laminectomy at C7T1
& T910; injury was
made w/ the extradu ral clip compression
technique at the T-1
segment; clip closure
force was 52 g, &
duration of compres sion was 1 min

Authors & Year Animal & Injury Models Experimental Groups

TABLE 16: Characteristics of SCI animal studies using nimodipine* (continued)

(continued)

Comments

Translational potential of pharmacotherapy for SCI

215

216

Animal model:
Wistar rats
N = 44
Sex: 100% F
Age: adult
Wt: 250315 g
SCI model:
Laminectomy C-7 &
T-1; acute compres sion of T-1 segment
of the cord for 1 min,
w/ a 52-g curved,
modified, Kerr Lougheed aneurysm
clip

SCI + nimodipine (8 hr infusion, then


infusions 3/day
for 7 days) (5-min
post-SCIplacebo
[PEG-400 &
ethanol] infusions)
(Group A)
SCI + nimodipine (8
hrs) (5-min & 7
daysplacebo
infusions)
(Group B)
SCI + MP (5-min & 8 hr infusions of MP)
(7 daysplacebo
infusion) (Group C)
SCI + placebo (place bo infusions at 5
min, 8 hrs, & 7
days) (Group D)
1013 animals/group

Reported Outcomes
Physiological Parameters:
The only significant difference across Tx groups was in BP, w/ the MP group
having significantly higher MABP at the end of the 8-hr infusions (df = 3,
40; F = 4.93; p <0.01)
Functional Assessment:
Inclined-plane scores were best in Group A & worst in Group C; significant
difference btwn the 2 (p <0.05), but there was no difference btwn Group A
& placebo (Group D)
SSEPs:
Group A had the highest yield of positive responses (50%), but this was not
significantly greater than the yield in other groups
Groups B & D had 33% positive responses present at 8 wks, whereas Group
C showed a 43% yield of positive evoked responses at this time
Counts of Retrogradely Labeled Red Nucleus Neurons:
Group A had the best outcome by this measure, but the standard errors were
large & no significant difference btwn groups was found
Composite Outcome Score:
Group A performed best in all outcome measures, although not significantly
so
Mean total composite outcome scores per experimental group: A = 14.5;
B = 5.125; C = 8.375; D = 6.0 (based on performances in individual
measuressurvival, functional assessment, SSEPs, & retrogradely
labeled red nucleus neurons)
Significant difference btwn the groups (df = 3, H = 9.89, p <0.025)

Intervention Details
Route: IP inj
Timing & dosage: Txs began
15 min postinjury
Group A: 0.02 mg/kg/hr
nimodipine 8-hr infusion; 20
mg/kg nimodipine enterally
3/day for 7 days postinjury
Group B: 0.02 mg/kg/hr
nimodipine 8-hr infusion
Group C: 30 mg/kg MP 5 min
postinjury; 5.4 mg/kg/hr MP
8-hr infusion
Animals killed: 8 wks postinjury

Of the 44 rats, 27
(61%) survived for
8 wks; at least 6
rats survived in
each group

Comments

* ANS = antirat neutrophil serum; BP = blood pressure; CEP = cerebellar evoked potential; EP = evoked potential; MABP = mean arterial blood pressure; MAP = mean arterial pressure; MEP = motor
evoked potential; MP = methylprednisolone; mSAP = mean systemic arterial pressure; NSS = normal sheep serum; PMNL = polymorphonuclear leukocyte; SCBF = spinal cord blood flow; SEP = spinal
evoked potential; TRH = thyrotropin-releasing hormone.

Ross et al.,
1993

Authors & Year Animal & Injury Models Experimental Groups

TABLE 16: Characteristics of SCI animal studies using nimodipine* (continued)

C. H. Tator et al.

J Neurosurg: Spine / Volume 17 / September 2012

animal model (no./group)


rats
injury model
avulsion (cervical)
avulsion (lumbar)
blunt force (wt drop) (lumbar)
clip compression (thoracic)
inflation compression (thoracic)
transection (sacral)
timing of intervention postinjury
0 hrs
either 0 hrs or 5, 10, 14, or 16 days; then 1/
day for 1 wk, followed by 1/2 days for 2 wks
0 hrs, then 1/day for 1 wk, followed by 1/2
days for 2 wks
15 min
15 min & 2 hrs
30 min, then 2/day for 10 days
2 & 4 hrs
1/day for 3 days
1/day for 2 wks
route of intervention
IVC
IP
IT catheter
IV
microdialysis fiber
dosage
0.8 mg/kg
2 mg/kg
2.5 mg/kg
4 mg/kg
5 mg/kg
8 mg/kg
10 mg/kg
2.0 mM

Characteristic

J Neurosurg: Spine / Volume 17 / September 2012


X

90 (18)

Ates
et al.,
2007

Hama &
Sagen,
2011
Kitzman,
2009

X
X
X

X
X

39 (410) 58 (1018) 22 (1012)

Bergerot
et al.,
2004

TABLE 17: Summary of riluzole preclinical study characteristics*

54 (6)

McAdoo
et al.,
2005

36 (9)

36 (9)

Mu
Mu
et al., et al.,
200050 200049

20 (35)

Ngrdi
& Vrbov,
2001

32 (325)

Ngrdi
et al.,
2007

30 (5)

Pintr
et al.,
2010

60 (15)

Schwartz
& Fehlings,
2001

(continued)

20 (10)

Stutzmann
et al.,
1996

Translational potential of pharmacotherapy for SCI

217

218
X
NR

% animals accounted for

NR

Hama &
Sagen,
2011

* IVC = intracerebroventricular cannula; 2HPbCD = 2-hydroxypropyl-b-cyclodextrin.

100 (90
of 90)

X
X

Bergerot
et al.,
2004

>1 dose evaluated?


yes
no
control groups
SCI (saline, 2HPbCD, pluronium F68)
SCI (no Tx)
no SCI
independent or blind assessment
yes
no
NR
animals randomized to Tx groups
yes
no
NR

Characteristic

Ates
et al.,
2007

TABLE 17: Summary of riluzole preclinical study characteristics* (continued)

NR

Kitzman,
2009

NR

McAdoo
et al.,
2005

NR

NR

Mu
Mu
et al., et al.,
200050 200049

100 (32 of
32)

X
X

Ngrdi
& Vrbov,
2001

100 (20 of
20)

X
X

Ngrdi
et al.,
2007

100 (30 of
30)

X
X

Pintr
et al.,
2010

100 (60 of
60)

Schwartz
& Fehlings,
2001

100 (20 of
20)

Stutzmann
et al.,
1996

C. H. Tator et al.

J Neurosurg: Spine / Volume 17 / September 2012

Ates et al.,
2007

Animal model:
Wistar albino rats
N = 90
Sex: 100% M
Age: adult
Wt: 200250 g
SCI model:
Wt drop trauma (5 g
10 cm; impact of 50
g/cm to dorsal
surface)

Authors & Year Animal & Injury Models


SCI +
Riluzole
Mexiletine
Phenytoin
Vehicle/saline
Sham (laminectomy
only)
18 rats/Tx group

Experimental Groups

TABLE 18: Characteristics of SCI animal studies using riluzole*

Route: IP
Timing: 0 hrs post-SCI
Dosage: single dose of
Riluzole: 8 mg/kg
Mexiletine: 80 mg/kg
Phenytoin: 200 mg/kg
Vehicle/saline: 1 ml physiological saline
Animals killed:
3 subgroups killed per Tx group:
24 hrs (spinal cord edema)
24 hrs (lipid peroxidation)
6 wks (neurobehavioral & histopathological
recovery) (gentamicin administered for 3
days postinjury to protect against UTI)

Intervention Details
Motor Function Score:
SCI (riluzole) vs SCI (saline) vs Sham
1 wk: ~2 vs ~1.5 (p = NS) vs ~6
3 wks: ~4 vs ~2.5 (p = NR) vs ~6
6 wks: ~4.75 vs ~2.75 (p <0.05) vs ~6
No significant difference btwn riluzole, mexiletine, & phenytoin
Inclined-Plane Scores:
SCI (riluzole) vs SCI (saline) vs Sham
1 wk: ~22 vs ~15 (p = NR) vs ~67
3 wks: ~35 vs ~20 (p = NR) vs ~67
6 wks: ~48 vs ~32 (p <0.05) vs ~67
No significant difference btwn riluzole, mexiletine, & phenytoin
Histopathology:
SCI (riluzole) vs SCI (saline): more myelin tissue & more neurons
retained in gray matter (sham: normal)
Lesion size (% of entire spinal cord area): SCI (riluzole) vs SCI
(saline):
15.4 2.0% vs 23.5 2.6% (p <0.05)
No significant difference btwn riluzole, mexiletine, & phenytoin
Lipid Peroxidation Levels:
SCI (riluzole) vs SCI (saline) vs sham (MDA; in nmol/g wet tissue):
24 hrs: ~9 vs ~14.5 (p <0.05) vs ~6.5
Riluzole improved vs phenytoin (p <0.05); riluzole similar to
mexiletine (p = NS)
Spinal Water Content (%):
SCI (riluzole) vs SCI (saline) vs Sham
24 hrs: ~68% vs ~75% (p <0.05) vs ~67%
No significant difference btwn riluzole, mexiletine, or phenytoin
Safety/Adverse Events/Toxicology: NR

Reported Outcomes

J Neurosurg: Spine / Volume 17 / September 2012


(continued)

No independent/
blinded
assessment
of outcomes

Comments

Translational potential of pharmacotherapy for SCI

219

220
SCI +
Group 1:
VRI (n = 10)
GDNF (n = 5)
GDNF + VRI (n = 7)
Riluzole (n = 5)
Riluzole + VRI (n = 4)
GDNF + riluzole + VRI
(n = 8)
Sham (n = 8)
Control (untreated;
n =7)
Group 2:
VRI (n = 10)
VRI + GDNF (n = 4)
VRI + riluzole (n = 4)
VRI + riluzole + GDNF
(n = 8)
Control (untreated;
n = 5)
Sham: n = 8 for both
groups

Animal model:
Sprague-Dawley rats
N = 39
Sex = 100% F
Age: adult
Wt: 180250 g
SCI model:
VRA (L46 ventral tear
made using fine
forceps)

Bergerot et al.,
2004

Experimental Groups

Authors & Year Animal & Injury Models


Route: IP for riluzole, IT for GDNF
Dosage:
Riluzole: 4 mg/kg
GDNF: 12 g/day
Timing: daily for 2 wks for both drugs
Animals killed:
Group 1: at 2 wks
Group 2: at 3 mos

Intervention Details

TABLE 18: Characteristics of SCI animal studies using riluzole* (continued)

Motor & Neurological Function:


% Motoneuron Survival on Contralat Side
3 mos: SCI (riluzole + VRI) vs SCI (GDNF + riluzole + VRI) vs SCI
(untreated) vs sham: ~50 vs ~70 vs ~40 vs ~100%
VRI + riluzole Tx resulted in significant survival of motoneurons
compared to untreated group (p <0.05)
The combined GDNF + riluzole + VRI Tx had a significant impact
on motoneuron survival compared to VRI + riluzole (p <0.05)
Both VRI + riluzole & combined Tx had significantly fewer moto neurons than sham (p <0.05)
Time on Accelerating Rotarod (sec)
3 mos: SCI (riluzole + VRI) vs SCI (GDNF + riluzole + VRI) vs SCI
(untreated) vs sham: ~70 vs ~90 vs ~40 vs ~105 sec
Combined Tx resulted in significant improvement over untreated
group (p <0.05)
BBB Locomotor Rating Scale (range 021, indicating no locomo tor function to normal locomotor performance, respectively):
3 mos: SCI (riluzole + VRI) vs SCI (GDNF + riluzole + VRI) vs SCI
(untreated) vs sham: ~6 vs ~17 vs ~3 vs ~20
VRI + GDNF + riluzole resulted in significant improvement com pared to other groups (p <0.05)
Sham was significantly better than all other Tx groups, w/ the
exception of the combined Tx (p <0.05)
Motoneuron Survival Rate:
At 2 wks, SCI (riluzole) vs SCI (control): 81 9% vs 52 5%
At 2 wks, Tx w/ riluzole improved motoneuron survival compared
to injury-only group (p <0.01)
At 3 mos, riluzole + GDNF + VRI vs VRA: 80 3% vs 42 3%
Dendrite No.:
3 mos: SCI (riluzole + VRI) vs SCI (GDNF + riluzole + VRI) vs SCI
(untreated) vs sham: ~4.5 vs ~6.5 vs ~1.1 vs ~3
VRI + GDNF + riluzole significantly improved compared to other
Tx groups
VRI + riluzole showed significantly fewer dendrites when com pared to control (p <0.05)
Regenerated Axons in Sciatic Nerve:
VRI + riluzole vs VRI + riluzole + GDNF vs VRA vs intact: 10 3
vs 10 4 vs 1 1 vs 44 5 (p = NR)
Safety/Adverse Events/Toxicology: NR

Reported Outcomes

(continued)

Comments

C. H. Tator et al.

J Neurosurg: Spine / Volume 17 / September 2012

J Neurosurg: Spine / Volume 17 / September 2012

Animal model:
SCI +
Sprague-Dawley rats
Riluzole
N = 22
Vehicle/saline
Sex: 100% F
Age: adult
Wt: 200250 g
SCI model:
Spinal cord transection
(S-2 level)

SCI +
Riluzole
Vehicle/saline
Delivered via:
IVC (n = 18)
IT catheter (n = 18)
2 control groups, non injured & implanted
w/ either IVC (n = 10)
or IT catheter
(n = 12)

Animal model:
Hama &
Sagen, 2011 Sprague-Dawley rats
N = 58
Sex: 100% M
Age: adult
Wt: 125150 g
SCI model:
Microvascular clip (on
spinal cord seg ments T67 for 1
min)

Kitzman, 2009

Experimental Groups

Authors & Year Animal & Injury Models

Route: IP
Timing: 1 dose/day for 3 days
Dosage:
Group I, 8 mg/kg; n = 10
Group II, 10 mg/kg; n = 12
Initial dose for both groups was 8 mg/kg

Route: IP, IVC, or IT catheter


Dosage:
IP group: 0.8, 2.5, or 8 mg/kg
IVC group: 0.3, 1, 10, or 30 g/kg
IT group: 10 or 30 g/kg

Intervention Details

TABLE 18: Characteristics of SCI animal studies using riluzole* (continued)

von Frey Filament Test (response to filament, converted to 50%


withdrawal threshold, & measured in g):
IP injection of 8 mg/kg riluzole resulted in significant increases in
withdrawal thresholds at 120 min postinjury, & vehicle at 90 &
120 min postinjury (p <0.05)
IT injection resulted in no significant change in withdrawal
thresholds
In the instance of an IVC injection into the rt ventricle, increase of
withdrawal thresholds was observed at 1, 10, & 30 g of
riluzole vs control (p <0.05)
Tail Flick Test (time for tail to withdraw from noxious heat
stimulus):
An increase in tail flick latency was observed beginning at 30120
min postinjury following an IP injection of 8 mg/kg of riluzole vs
control (p <0.05)
IT & IVC injections had no effect on withdrawal latency
BBB Locomotor Rating Scale (range 021, indicating no locomo tor function to normal locomotor performance, respectively):
Riluzole vs Control
2 hrs postinjury: 9 1 vs 8 1
Neither riluzole nor vehicle significantly altered BBB scores
Safety/Adverse Events/Toxicology: NR
Behavioral Responses (response to each stimulus graded on a
05 scale, indicating 45 flexion to significant coiling, flexing,
extending, or writhing of the tail, respectively):
Response to Light Touch, SCI (riluzole 8 mg/kg) vs SCI (vehicle)
Preinjury: ~1.0 vs ~0.7
1 hr: ~0.4 vs ~0.9 (p <0.01)
3 hrs: ~0.5 vs ~0.7
6 hrs: ~0.9 vs ~0.85
12 hrs: ~1.1 vs ~0.8
Response to Pinch, SCI (riluzole: 8 mg/kg) vs SCI (vehicle)
Preinjury: ~5.0 vs ~5.0
1 hr: ~3.5 vs ~4.75 (p <0.05)
3 hrs: ~3.4 vs ~4.5 (p <0.05)
6 hrs: ~4.6 vs ~5.0
12 hrs: ~4.65 vs ~5.0
Response to Pinch, SCI (riluzole: 10 mg/kg) vs SCI (vehicle)
Preinjury: ~5.0 vs ~4.95
1 hr: ~2.7 vs ~4.95 (p <0.01)
3 hrs: ~2.95 vs ~5.0 (p <0.05)
6 hrs: ~4.2 vs ~5.0
12 hrs: ~5.0 vs ~5.0

Reported Outcomes

(continued)

Comments

Translational potential of pharmacotherapy for SCI

221

222

McAdoo et al.,
2005

Kitzman, 2009
(continued)

Animal model:
Sprague-Dawley rats
N = 54
Sex: 100% M
Age: adult
Wt: 240320 g
SCI model:
Wt drop trauma (10 g
24 mm; NASCIS
impactor or Infinite
Horizons impactor)

Authors & Year Animal & Injury Models

NMDA receptor blockers:


MK-801
Mematine
AMPA/kainite receptor
blockers:
NBXQ
GYKI 52466
Cyclothiazide
Sodium channel blockers:
Riluzole
Mexiletine
QX-314
Sham
See Route & Dosage;
n = 6 for both drug &
sham in each
dosage grouping

Experimental Groups

Intervention Details

Route, dosage, & timing:


NMDA receptor blockers
Mematine: 100 M; through microdialysis fiber;
administered from impact injury to end of expt
MK-801: 1.0 mM; through fiber; from impact
injury to end of expt
MK-801: 0.89 mM; through fiber; immediately
postinjury
MK-801: 4.0 mg/kg; IP; immediately postinjury
AMPA/kainite receptor blockers
NBXQ: 10.0 mM; through fiber; from impact
injury to end of expt
NBXQ: 1.8 l, 15 mM; via inj; immediately
postinjury
NBXQ: 1.8 l, 15 mM; via inj; immediately
postinjury
NBXQ: 1.8 l, 15 mM; via inj; immediately
postinjury
NBXQ: 10.0 mM; through fiber; from impact
injury to end of expt
NBXQ: 10.0 mM; through fiber; from impact
injury to end of expt
NBXQ: 10.0 mM; through fiber; from impact
injury to end of expt
NBXQ: 10.0 mM; through fiber; immediately
postinjury
Cyclothiazide: 1.8 l, 10 M; via inj outside
impact area; immediately postinjury
GYK 52466: 2.0 mM; through fiber; from impact
injury to end of expt

TABLE 18: Characteristics of SCI animal studies using riluzole* (continued)

Response to Light Touch, SCI (riluzole: 10 mg/kg) vs SCI (vehicle)


Preinjury: ~1.5 vs ~1.5
1 hr: ~0.7 vs ~1.52 (p <0.05)
3 hrs: ~0.6 vs ~1.53 (p <0.05)
6 hrs: ~1.2 vs ~1.53
12 hrs: ~1.45 vs ~1.6
Response to Stretch, SCI (riluzole: 10 mg/kg) vs SCI (vehicle)
Preinjury: ~4.95 vs ~4.5
1 hr: ~2.4 vs ~4.5 (p <0.001)
3 hrs: ~2.3 vs ~4.4 (p <0.001)
6 hrs: ~4.0 vs ~4.4
12 hrs: ~4.2 vs ~4.5
Safety/Adverse Events/Toxicology: NR
Riluzole did not have an effect on glutamate release following SCI
(no other information reported)
Safety/Adverse Events/Toxicology: NR

Reported Outcomes

(continued)

Comments

C. H. Tator et al.

J Neurosurg: Spine / Volume 17 / September 2012

Mu et al.,
200050

McAdoo et al.,
2005
(continued)

Animal model:
Long-Evans rats
N = 36
Sex: 100% F
Wt: 225250 g
SCI model:
Wt drop trauma (T-10
NYU Impactor rod,
12.5-mm drop)

Authors & Year Animal & Injury Models

SCI +
Riluzole
MP
Riluzole + MP
Vehicle/saline
9 rats/group

Experimental Groups

Intervention Details
Route, dosage, & timing (continued):
Sodium channel blockers
Riluzole: 2.0 mM; through fiber; from impact
injury to end of expt
Mexiletine: 80 mg/kg IP; immediately postinjury
QX-314: 2 ml, 5 mM; inj; immediately postinjury
Route: IP
Dosage:
Riluzole: 8 mg/kg
MP: 30 mg/kg
Timing: 15 min & 2 hrs postinjury
Animals killed: 4 hrs postinjury for all groups

TABLE 18: Characteristics of SCI animal studies using riluzole* (continued)

Mitochondrial Function (% of control):


SCI (Riluzole) vs SCI (Riluzole + MP) vs SCI (Control): ~130% vs
~135% vs ~100%
Riluzole significantly increased measures of mitochondrial func tion compared to control (p <0.001)
Riluzole + MP increased mitochondrial function in synaptosomes
(p <0.01)
Reactive Oxygen Species Levels in Synaptosomes (% of control):
SCI (Riluzole) vs SCI (Riluzole + MP) vs SCI (Control): ~90% vs
~75% vs ~100%
Reactive oxygen species levels were significantly reduced, by
33% (p <0.01) in riluzole + MP group compared to control
Neither riluzole nor MP alone had any effect on reactive oxygen
species levels
MDA Levels (% of control):
SCI (Riluzole) vs SCI (Riluzole + MP) vs SCI (Control): ~90% vs
~70% vs 100%
Thiobarbituric acid reactive product levels in riluzole group were
no different than control
In riluzole + MP group, MDA levels were decreased by 29%
(p <0.01)
Glutamate Uptake (% of control):
SCI (Riluzole) vs SCI (Riluzole + MP) vs SCI (Control): ~190% vs
~160% vs 100%
In riluzole-treated group, glutamate uptake was increased by 87%
(p <0.01)
In riluzole + MP group, glutamate uptake was increased by 75%
(p <0.01)
Glucose Uptake (% of control):
SCI (Riluzole) vs SCI (Riluzole + MP) vs SCI (Control): ~140% vs
190% vs 100%
Glucose uptake was increased by 43.8% in riluzole group
(p <0.025) compared to control
Glucose uptake was increased by 85% in the riluzole + MP group
(p <0.025) as opposed to control group
Safety/Adverse Events/Toxicology: NR

Reported Outcomes

(continued)

Comments

Translational potential of pharmacotherapy for SCI

J Neurosurg: Spine / Volume 17 / September 2012

223

224
SCI +
Riluzole
MP
Riluzole + MP
Vehicle/saline
9 rats/group

SCI +
Reimplantation of nerve
dorsolaterally from
spinal cord; n = 4
Reimplantation +
Riluzole; n = 4
Grafted embryonic moto neurons; n = 4
Riluzole + grafted moto neurons: n = 5
Control/intact; n = 3

Animal model:
Long-Evans rats
N = 36
Sex: 100% F
Age: adult
Wt: 225250 g
SCI model:
Wt drop trauma (T-10
SCI Impactor rod,
12.5-mm drop)

Animal model:
Wistar rats
N = 20
Sex: NR
Age: adult
Wt: NR
SCI model:
L-4 avulsion & reimplantation

Ngrdi &
Vrbov,
2001

Experimental Groups

Mu et al.,
200049

Authors & Year Animal & Injury Models

Intervention Details

Route: IP
Dosage: 4 mg/kg
Timing: 1 dose at 0 hrs IP; then 1 dose/day for
1 wk; then 1 dose/2 days for 2 wks
Animals killed: at 3 mos

Route:
Riluzole: IP
MP: IV at femoral vein
Dosage:
Riluzole: 8 mg/kg
MP: 30 mg/kg
Timing: 2 & 4 hrs postinjury

TABLE 18: Characteristics of SCI animal studies using riluzole* (continued)

BBB Locomotor Rating Scale (range 021, indicating no locomo tor function to normal locomotor performance, respectively):
SCI (Riluzole) vs SCI (Riluzole + MP) vs SCI (Control)
Before trauma: ~20 vs ~20 vs ~21
Wk 1: ~0 vs ~0 vs ~1
Wk 2: ~4 vs ~4 vs ~3
Wk 3: ~6 vs ~7 vs ~5.5
Wk 4: ~12 vs ~13 vs ~9
Wk 5: ~12.5 vs ~14 vs ~10
Wk 6: ~13 vs ~14 vs ~10
Histopathology:
Mean Myelin Index, SCI (Riluzole) vs SCI (Riluzole + MP)
0.91 0.08 vs 0.84 0.09
Control: NR
Safety/Adverse Events/Toxicology: NR
No. of L-4 Motoneurons (3 mos):
Untreated vs SCI (reimplantation + riluzole) vs SCI (reimplanation
+ graft + riluzole) vs control
3 mos: 723 26 vs 645 36 vs 1181 40
Safety/Adverse Events/Toxicology: NR

Reported Outcomes

(continued)

Comments

C. H. Tator et al.

J Neurosurg: Spine / Volume 17 / September 2012

Ngrdi et al.,
2007

Animal model:
Sprague-Dawley rats
N = 32
Sex: NR
Age: adult
Wt: 180200 g
SCI model:
L-4 avulsion & reim plantation of nerve
dorsolaterally into
spinal cord

Authors & Year Animal & Injury Models


SCI +
Riluzole (n = 25)
No Tx (n = 4)
Control/intact (n = 3)

Experimental Groups

Intervention Details
Route: IP
Dosage: 4 mg/kg
Timing: started either at 0 hrs postinjury or 5,
10, 14, & 16 days postinjury (5 rats/group);
1 dose/day for 1 wk; then 1 dose/2 days for
2 wks
Animals killed: 3 mos

TABLE 18: Characteristics of SCI animal studies using riluzole* (continued)

Behavioral:
Animals treated w/ riluzole w/in 14 days postop started to recover
from paralysis during the 3rd & 4th wk postinjury, & by end of
period were almost able to walk normally, & flexed ankle joint
during locomotion, whereas the no-Tx group never recovered
dorsiflexion of ankle joint or the ability to walk at any time
Muscle Wt Loss (% of muscle wt on the control [unoperated]
side):
Riluzole, EDL & TA Muscles
Immediate: 9 1.5% & 15.4 1.8%
5-day delay: 9.8 1.16% & 17 1.15%
10-day delay: 9.9 2% & 18.5 2.2%
14-day delay: 24.3 1.3% & 30.7 2.8%
16-day delay: 21.7 1.8% & 29.3 4.9%
Untreated: 39.1 4.5% & 48.5 5.3%
Motoneuron Count:
Riluzole
0 hrs postinjury: 763 36
5-day delay: 815 50.6
10-day delay: 722 39.1
14-day delay: 67 3.9
16-day delay: 52 3
Untreated: 20.4 1.6
Control: 1164 29
Significant difference btwn animals that received riluzole &
untreated group (p = 0.016)
Presence of Fast Blue Dye vs ChAT (ChAT levels indicate
presence of cholinergic cells in the spinal cord, whereas Fast
Blue levels indicate reinnervating neurons):
5-day delay: 70 4.3 vs 90.7 2.1
10-day delay: 66.3 3.3 vs 85.4 2.4
14-day delay: 5.76 0.34 vs 41.4 2.1
16-day delay: 4.5 0.26 vs 38.3 2.5
Safety/Adverse Events/Toxicology: NR

Reported Outcomes

(continued)

Comments

Translational potential of pharmacotherapy for SCI

J Neurosurg: Spine / Volume 17 / September 2012

225

226

Pintr et al.,
2010

Animal model:
Sprague-Dawley rats
N = 30
Sex: 100% F
Age: adult
Wt: 180200 g
SCI model:
C-7 VRA

Authors & Year Animal & Injury Models


SCI +
No Tx
Reimplantation
Dural nerve graft implantation
Riluzole
Riluzole + reimplantation
5 rats/group

Experimental Groups

Intervention Details
Route: IP
Dosage: Riluzole 4 mg/kg
Timing: 0 hrs postinjury, then 1/day for 1 wk,
& 1/2 days for 2 wks
Animals killed:
For the no-Tx & riluzole + reimplantation group,
rats were killed at 5 wks
For the other 3 groups, rats were killed at 3
mos + 6 days

TABLE 18: Characteristics of SCI animal studies using riluzole* (continued)

Movement: Dorsiflexion of Wrist Joint (dorsiflexion graded on a


scale of 03, indicating no dorsiflexion & dorsiflexion >30,
respectively):
SCI (avulsion + reimplantation) vs SCI (avulsion, reimplantation
+ riluzole)
1 wk: ~0 vs ~0
2 wks: ~0 vs ~0
3 wks: ~0 vs ~0.1
4 wks: ~0.1 vs ~0.2
5 wks: ~0.3 vs ~1.1
6 wks: ~0.7 vs ~1.6
7 wks: ~1.2 vs ~1.9
8 wks: ~1.3 vs ~2.0
9 wks: ~1.5 vs ~2.5 (p 0.05)
10 wks: ~1.8 vs ~2.9 (p 0.05)
11 wks: ~1.8 vs ~2.9 (p 0.05)
12 wks: ~2.0 vs ~2.9 (p 0.05)
Movement: Contracture of Wrist Joint (contracture graded on a
scale of 03, indicating no contraction & severe contraction,
respectively):
SCI (avulsion + reimplantation) vs SCI (avulsion, reimplantation +
riluzole)
1 wk: ~0 vs ~0
2 wks: ~0 vs ~0
3 wks: ~0 vs ~0
4 wks: ~0 vs ~0.4
5 wks: ~0 vs ~0.5
6 wks: ~0 vs ~1.2
7 wks: ~0.1 vs ~1.3
8 wks: ~0.1 vs ~1.8 (p 0.01)
9 wks: ~0.1 vs ~2.2 (p 0.01)
10 wks: ~0.1 vs ~2.5 (p 0.01)
11 wks: ~0.1 vs ~3.0 (p 0.01)
12 wks: ~0.1 vs ~3.0 (p 0.01)
Intact Motoneurons:
SCI (no Tx) vs SCI (riluzole) vs Control (no injury)
5 wks: 65 8 vs 637 26 (p 0.001) vs 875 21
SCI (reimplantation) vs SCI (reimplantation + riluzole) vs control
(no injury):
5 wks: 211 15 vs 573 9 (p 0.001) vs 875 21
Expression of ChAT:
No groups were significantly different w/ regard to the proportion
of fast bluepositive/ChAT-positive neurons
Safety/Adverse Events/Toxicology: NR

Reported Outcomes

(continued)

Comments

C. H. Tator et al.

J Neurosurg: Spine / Volume 17 / September 2012

Schwartz &
Fehlings,
2001

Animal model:
Wistar rats
N = 60
Sex: 100% F
Age: adult
Wt: 225280 g
SCI model:
Compression (53 g for
1 min at C7T1)

Authors & Year Animal & Injury Models


SCI +
Riluzole
Phenytoin
CNS5546A
Vehicle/2HPbCD
15 rats/group

Experimental Groups

Intervention Details
Route: IP
Dosage:
Riluzole: 5 mg/kg
Phenytoin: 30 mg/kg
CNS5546A: 15 mg/kg
Vehicle: 5 mg/kg
Timing: 15 min postinjury for all Txs

TABLE 18: Characteristics of SCI animal studies using riluzole* (continued)

BBB Locomotor Rating Scale (range 021, indicating no locomo tor function to normal locomotor performance, respectively):
SCI (riluzole) vs SCI (vehicle)
Wk 1: ~2 vs ~2
Wk 2: ~4.5 vs ~3
Wk 3: ~7.5 vs ~4
Wk 4: ~9 vs ~6
Wk 5: ~10 vs ~7.5
Wk 6: ~11 vs ~8
Riluzole-treated animals showed significantly higher inclined plane scores compared w/ control animals at 4, 5, & 6 wks
(p <0.05)
Inclined-Plane Performance Scores (maximal angle at which
animal could maintain position for 5 sec):
SCI (riluzole) vs SCI (vehicle)
Wk 1: ~24 vs ~15
Wk 2: ~35 vs ~20
Wk 3: ~41 vs ~27
Wk 4: ~45 vs ~35
Wk 5: ~47 vs ~37
Wk 6: ~49 vs ~39
Riluzole-treated rats showed significantly higher inclined-plane
scores compared w/ phenytoin-treated & control rats at all
follow-up intervals (except 5 wks) (p <0.05)
Histopathology (7 wks):
Neuron Counts: SCI + Riluzole vs SCI + Vehicle (range)
Red Nucleus: 1442.5 299.7 (6261904) vs 733.8 273.5
(2561392)
Raphe Nuclei: 576.8 129.2 (293859) vs 467.8 115.4
(161721)
Reticular Formation: 1819.3 281.2 (11082320) vs 1359.5
228.0 (9761947)
Vestibular Nuclei: 591.5 240.6 (2451296) vs 102.0 171.8
(138902)
RVLM: 86.8 25.8 (26134) vs 82.5 42.6 (20201)
Significant differences btwn Tx groups:
Red Nucleus: p = 0.02
Raphe nuclei: p = 0.74
Reticular formation: p = 0.63
Vestibular nuclei: p = 0.94
RVLM: p = 0.54
Riluzole group had significantly higher mean neuron counts in the
red nucleus (p <0.05)
Mean Cell Diameters: SCI + Riluzole vs SCI + Vehicle
26.06 m vs 18.73 m (p <0.05)

Reported Outcomes

(continued)

Comments

Translational potential of pharmacotherapy for SCI

J Neurosurg: Spine / Volume 17 / September 2012

227

228

Schwartz &
Fehlings,
2001
(continued)

Authors & Year Animal & Injury Models

Experimental Groups

TABLE 18: Characteristics of SCI animal studies using riluzole* (continued)


Intervention Details
Normalized Residual Tissue Area: SCI + Riluzole vs SCI +
Vehicle
7 wks: increased residual tissue area in riluzole-treated animals
vs control (p <0.05)
Normalized Epicenter Cavity Area: SCI + Riluzole vs SCI + Vehicle
7 wks: ~0.3 vs ~0.5 mm (p <0.05)
Riluzole significantly increased residual tissue & decreased cavity
area compared to other Txs (p <0.05)
At 1 & 1.5 mm caudal to epicenter, riluzole also increased tissue
sparing compared to other groups (p <0.05)
White Matter Vol
At 1.5 mm rostral to epicenter, riluzole increased white matter vol
compared to CNS5546A & control (p <0.05)
At 2 mm rostral to epicenter, both riluzole & phenytoin increased
vols compared to control (p <0.05)
Gray Matter Vol
At 1 mm rostral, 1.5 & 2 mm caudal, & at 1 mm caudal compared
to other Txs or control, riluzole had a marked effect on gray
matter vol (p <0.05)
At 1.5 & 2 mm rostral, control had increased gray matter com pared to riluzole; at 1 & 2 mm rostral, control had greater gray
matter compared to phenytoin; & at 1 mm rostral, control had
increased gray matter compared to CNS5546A
Cavity Vol
Significant effects were seen at all caudal increments (p 0.005)
At 1.5 mm, riluzole had a greater effect on reduced cavity vol
when compared to phenytoin (p <0.05), control (p <0.05), &
CNS5546A (p <0.05)
At 1 mm caudal, CNS5546A also decreased cavity vol compared
to control (p <0.05)
At each increment, the overall difference btwn components was
found to be significant (p <0.00001). Gray matter vol increased
compared w/ that of the cavity (p <0.05). Overall, significantly
greater gray matter & reduced cavity vols emerged (p = 7.84
1011 & p = 6.20 107, respectively). Riluzole had the greatest
effect on both vols (p <0.05). Significant interactions among
tissue & cavity vols emerged btwn increments (p = 2.40
1036) & drug Txs (p = 1.02 1012)
Safety/Adverse Events/Toxicology: NR

Reported Outcomes

(continued)

Comments

C. H. Tator et al.

J Neurosurg: Spine / Volume 17 / September 2012

J Neurosurg: Spine / Volume 17 / September 2012

SCI +
Animal model:
Riluzole
Wistar rats
N = 20
Control/Vehicle
Sex: 100% M
Age: adult
Wt: 260300 g
SCI model:
Compression (Fogarty
balloon catheter, 2.5
bars for 8 min)

Experimental Groups
Route: IV
Dosage:
Riluzole: 2 mg/kg
Vehicle: 1.5% pluronic F68 in 0.9% NaCl
Timing:
Riluzole: 30 min postinjury, then 2/day for 10
days
Vehicle: 30 min postinjury, then 2/day for 10
days
Animals killed: at 10 days

Intervention Details
Behavioral:
After 6 2 days, 70% (7 of 10) of animals treated w/ riluzole could
use paws to sit upright; 30% (3 of 10) maintained marked motor
deficit beyond that point
SSEP Amplitude Scores (in V):
SCI + Riluzole vs SCI + Vehicle
Control: ~10 vs ~13
Before trauma: ~10 vs ~12
Day 1: ~0 vs ~0
Day 3: ~2 vs ~0
Day 5: ~4 vs ~0
Day 7: ~5 vs ~0
SSEP Duration (in V):
SCI + Riluzole vs SCI + Vehicle
Control: ~4.2 vs ~3.8
Before trauma: ~4.2 vs ~3.9
Day 1: ~0 vs ~0
Day 3: ~3.0 vs ~0
Day 5: ~5.0 vs ~0
Day 7: ~5.4 vs ~0
SSEP Latency (in V):
SCI + Riluzole vs SCI + Vehicle
Control: ~12 vs ~16
Before trauma: ~12 vs ~13
Day 1: ~30 vs ~30
Day 3: ~15 vs ~30
Day 5: ~15 vs ~30
Day 7: ~17 vs ~30
Riluzole group showed recovery of SSEP amplitude, duration, &
latency
Vehicle-treated animals showed no recovery
Histopathological Assessment:
Hemorrhagic zone: SCI + riluzole vs SCI + vehicle: 1.03 0.12
mm3 vs 1.99 0.23 mm3 (p <0.05)
Protection by riluzole was most obvious in the white matter
Safety/Adverse Events/Toxicology: NR

Reported Outcomes

Comments

* ChAT = choline acetyltransferase; EDL = extensor digitorum longus; GDNF = glial cell linederived neurotrophic factor; IVC = intraventricular catheter; NASCIS = National Acute Spinal Cord Injury
Study; NMDA = N-methyl-d-aspartate; RVLM = rostral ventrolateral medulla oblongata; TA = tibialis anterior; UTI = urinary tract infection; VRA = ventral root avulsion; VRI = ventral root reimplantation.

Stutzmann et
al., 1996

Authors & Year Animal & Injury Models

TABLE 18: Characteristics of SCI animal studies using riluzole* (continued)

Translational potential of pharmacotherapy for SCI

229

You might also like