You are on page 1of 14

JOURNAL OF NEUROTRAUMA 30:1781–1794 (November 1, 2013)

ª Mary Ann Liebert, Inc. Review


DOI: 10.1089/neu.2013.2932

The Effects of the Timing of Spinal Surgery


after Traumatic Spinal Cord Injury:
A Systematic Review and Meta-Analysis

Joost J. van Middendorp,1–3 Allard J.F. Hosman,4 and Suhail A.R. Doi 3

Abstract
The debate over the effects of the timing of surgical spinal decompression after traumatic spinal cord injury (tSCI) has
remained unresolved for over a century. The aim of the current study was to perform a systematic review and quality-
adjusted meta-analysis of studies evaluating the effects of the timing of spinal surgery after tSCI. Studies were searched
for through the MEDLINE database (1966 to August 2012) and a 15-item, tailored scoring system was used for assessing
the included studies’ susceptibility to bias. Random effects and quality effects meta-analyses were performed. Models
were tested for robustness using one way and criterion-based sensitivity analysis and funnel plots. Results are presented as
weighted mean differences (WMDs) and odds ratios (ORs) with 95% confidence intervals (CIs). A total of 18 studies were
analyzed. Heterogeneity was evident among the studies included. Quality effects models showed that – when compared
with ‘‘late’’ surgery – ‘‘early’’ spinal surgery was significantly associated with a higher total motor score improvement
(WMD: 5.94 points, 95% CI:0.74,11.15) in seven studies, neurological improvement rate (OR: 2.23, 95% CI:1.35,3.67) in
six studies, and shorter length of hospital stay (WMD: - 9.98 days, 95% CI: - 13.10, - 6.85) in six studies. However, one
way and criterion-based sensitivity analyses demonstrated a profound lack of robustness among pooled estimates. Funnel
plots showed significant proof of publication bias. In conclusion, despite the fact that ‘‘early’’ spinal surgery was
significantly associated with improved neurological and length of stay outcomes, the evidence supporting ‘‘early’’ spinal
surgery after tSCI lacks robustness as a result of different sources of heterogeneity within and between original studies.
Where the conduct of a surgical, randomized controlled trial seems to be an unfeasible undertaking in acute tSCI, meth-
odological safeguards require the utmost attention in future cohort studies. (Prospero registration number: PROSPERO
CRD42012003182. See also http://www.crd.york.ac.uk/NIHR_PROSPERO/)

Key words: meta-analysis; SCI; systematic review; timing of surgery

Introduction edema, increased excitatory amino acids, and lipid peroxidation.2


Pre-clinical data support the theory that persistent compression of

M ore than a century ago, Burrell reported two key


issues related to the surgical management of traumatic spinal
cord injury (tSCI): the timing of surgery and the severity of the
the spinal cord represents a cause of secondary injury and, there-
fore, may be potentially reversible.3,4 Despite positive effects of
acute spinal decompressions reported in these standardized pre-
injury.1 Nowadays, the topic of the timing of surgical spinal de- clinical studies, no neurological benefits have consistently been
compression after tSCI continues to spark vigorous debates among reported in the human, clinical setting.
specialists. A number of systematic reviews examining the timing of spinal
Advocates for early spinal decompressions refer to the concept surgery after tSCI have been published over the last decade.5–10 A
of primary versus secondary mechanisms of injury.2 The primary profound limitation of all of these reviews is that non-comparative
mechanism comprises the initial cord lesion that results from case series were also included. Furthermore, the meta-analysis
physical trauma to the tissue caused by a displacement of sur- published by La Rosa and colleagues10 was severely limited by
rounding spinal structures. The primary mechanism in turn initiates applying a single, nonspecific pooled outcome measure, namely the
a cascade of secondary injury mechanisms including ischemia, ‘‘neurological improvement rate.’’ Finally, the impact of original

1
Stoke Mandeville Spinal Foundation, National Spinal Injuries Centre, Stoke Mandeville Hospital, Aylesbury, United Kingdom.
2
Harris Manchester College, University of Oxford, Oxford, United Kingdom.
3
Clinical Epidemiology Unit, School of Population Health, University of Queensland, Herston Road, Brisbane, Australia.
4
Spine Unit, Department of Orthopaedics, Radboud University Nijmegen Medical Center, Nijmegen, The Netherlands.

1781
1782 VAN MIDDENDORP ET AL.

studies’ variable susceptibility to bias on pooled outcome estimates Eligibility criteria


has not been considered in previous reviews.
The aim of the current study was to perform a systematic review The initial screening for eligibility included the following two
and meta-analysis of the effects of the timing of spinal surgery after criteria. The study had to consist of clearly defined cohorts of
subjects who had undergone spinal surgery after tSCI. In addition,
tSCI that differs from previous reviews in two ways. First, we critically
comparative outcomes of ‘‘early’’ and ‘‘late,’’ or delayed, spinal
appraised the reporting of key methodological safeguards reducing the surgery had to be reported. Both randomized and nonrandomized
susceptibility to bias in studies comparing outcomes in patients who comparative studies were eligible for inclusion. All neurological
underwent either ‘‘early’’ or ‘‘late’’ spinal surgery. Second, based on levels and all severities of tSCI were included. All approaches for
the methodological quality score of each individual study, we subse- spinal surgery were eligible for inclusion, including anterior, pos-
quently performed a quality effects (QE) meta-analysis. terior, and circumferential open approaches, as well as closed
manual reductions. Studies combining patients with non-tSCIs with
Methods patients with tSCIs were excluded. Studies that included pa-
tients <14 years of age were also excluded. Articles published in
Data sources
languages other than English and those without an abstract were
In order to identify relevant articles on the effects of the timing excluded. Titles and abstracts of retrieved articles were screened,
of spinal surgery after tSCI, we conducted a computerized search and potentially relevant full-text articles and reports were retrieved
using the MEDLINE (1966 to August 2012) database. The search and evaluated individually and independently by two reviewers
terms used in the PubMed interface are presented in Supplementary ( J.J.v.M. and A.J.F.H.), who were trained and experienced in
Appendix 1 (see online supplementary material at http://www performing systematic literature searches. In each phase, and in all
.liebertonline.com). To ensure that no relevant studies were missed cases, disagreement concerning inclusion of articles was resolved
by the computerized search, we also performed a manual cross- by discussion and consensus agreement. Details of the litera-
reference search of the citations of each included article to obtain ture search and selection process are outlined in the flow chart in
further relevant studies.11 Figure 1.

FIG. 1. Literature search and selection process. (*References that fit into more than one exclusion criterion were categorized in a
single category at the abstractor’s discretion, {Fourteen of the 18 studies [18%] included more than one outcome measure suitable for
meta-analysis.)
TIMING OF SPINAL SURGERY AFTER tSCI 1783

Data abstraction and quality assessment studies: the Consolidated Standards of Reporting Trials of Non-
pharmacologic Treatment (CONSORT-NPT) checklist,12 the cri-
Critical appraisal of included articles was performed to examine teria list for methodological quality assessment introduced by the
studies’ susceptibility to bias. A standard data extraction form Cochrane Collaboration Back Review Group for Spinal Dis-
summarizing the study design, study population, and relevant raw orders,13 and the Strengthening the Reporting of Observational
data was completed for each article. Because a standardized as- Studies in Epidemiology (STROBE) statement.14
sessment for the methodological quality of both surgical RCTs and A 15-item, tailored scoring system was devised for the assess-
non-RCTs does not currently exist, a tailored bias assessment ment of comparative surgical studies in spinal trauma (Table 1).
scoring instrument was devised based on reworded methodological The minimum and maximum quality scores are 0 and 25 points,
items covered by the following three consensus-derived ‘‘check- respectively. Included items cover key methodological safeguards
lists’’ for reporting randomized and nonrandomized clinical that reduce studies’ susceptibility to bias. As assessment of the

Table 1. A Tailored Checklist for Assessing the Susceptibility


to Bias in Therapeutic Comparative Studies for Spinal Cord Injury

Item Question Answer (score)

Selection bias

1 Was the treatment group allocation at random? Yes (1), No/Unclear (0)
2 Was the allocation sequence generated and concealed adequately? Yes (1), No/Unclear (0)
3 Was attrition < 20% and if so, was attrition comparable between both groups? Yes (1), No/Unclear (0)
4 Was the length of follow-up equal between the groups? Yes (1), No/Unclear (0)
For case-control studies: Was the time period between the intervention/exposure and
outcome the same for cases and controls?

Information bias

5 Were data assessed and recorded prospectively in both study groups? Yes (1), No/Unclear (0)
6 Were the primary and secondary outcomes and related hypotheses pre-specified? Yes (1), No/Unclear (0)
7 Were interventions clearly defined and comparable between and/or within the groups? Yes (1), No/Unclear (0)
8 Were the following persons blinded for treatment allocation?
a) Participants Yes (1), No/Unclear (0)
b) Care providers Yes (1), No/Unclear (0)
c) Outcome assessors Yes (1), No/Unclear (0)
9 Were standardized, reproducible definitions used for:
a) Baseline characteristics Yes (1), No/Unclear (0)
b) Study outcomes Yes (1), No/Unclear (0)
c) Adverse events Yes (1), No/Unclear (0)
10 Apart from blinding, were other safeguards used for assuring the reliability of:
a) Baseline characteristics Yes (1), No/Unclear (0)
b) Study outcomes Yes (1), No/Unclear (0)
c) Adverse events Yes (1), No/Unclear (0)

Confounding

11 Were important prognostic factors comparable in both groups or stratified/


matched/adjusted for analysis?
a) Severity of SCI Yes (1), No/Unclear (0)
b) Level of SCI Yes (1), No/Unclear (0)
c) Concomitant traumatic injuries Yes (1), No/Unclear (0)
d) Pre-existing comorbidities Yes (1), No/Unclear (0)
e) Age Yes (1), No/Unclear (0)
12 Did researchers rule out any impact from a concurrent intervention or an unintended Yes (1), No/Unclear (0)
exposure in both study groups?

Analytical and reporting bias

13 Were effect sizes based on the whole data or pre-defined subgroups rather than Yes (1), No/Unclear (0)
a post hoc portion of the data?
14 Were participants analyzed within the groups they were originally assigned to or, Yes (1), No/Unclear (0)
if not, was an intention-to-treat analysis performed?
15 Was a subset of the original recorded outcome variables or an incomplete set No (1), Yes/Unclear (0)
of key point estimates and variability measures reported?

An explanation and elaboration of these items can be found in Supplementary Appendix 2 (see online supplementary material at http://
www.liebertonline.com).
The total quality score ranges from 0 to 25 points.
SCI: spinal cord injury.
1784 VAN MIDDENDORP ET AL.

quality of a study is closely intertwined with the quality of re- using MetaXL version 1.3 available from www.epigear.com.
porting;15,16 therefore, the devised quality scoring system required During the reporting of current study’s findings we adhered to the
each item to be both reported and indicative of a reduced suscep- recommendations outlined in the Preferred Reporting Items for
tibility to bias. The scoring system is applicable to both randomized Systematic Reviews and Meta-Analyses (PRISMA) statement.26,28
and nonrandomized controlled comparative surgical studies. Gui-
dance for scoring individual items is presented in an ‘‘Explanation
Results
and Elaboration document,’’ which can be found in Supplementary
Appendix 2 (see online supplementary material at http://www Search and screening results
.liebertonline.com).
The computerized search strategy resulted in 2589 citations (Fig.
1). After screening of titles and abstracts, 42 remaining potential
Outcome measures
eligible articles were obtained for full-text screening. Twenty-one
Upon completion of the critical appraisal, outcome measures articles were excluded after a more detailed evaluation. The ma-
suitable for meta-analysis were identified. The primary outcome jority of the irrelevant articles did not evaluate the effect of the
measure was neurological improvement, either measured via 1) the timing of spinal surgery specifically, or compared operative and
mean difference in Total Motor Score as defined by the Interna- nonoperative treatments. Cross-referencing resulted in one addi-
tional Standards for Neurological Classification of Spinal Cord tional study. Twenty-two studies29–50 reported on the effects of
Injury,17,18 or 2) the ‘‘neurological improvement rate’’ odds ratio
the timing spinal surgery after tSCI, of which 18 studies reported
(OR); using different scales, including the American Spinal Injury
Association (ASIA) Impairment Scale17,18 and the Frankel scale.19 usable information for meta-analysis.
Secondary outcomes included the mean difference in length of
hospital stay, the mortality OR, and the adverse events OR. Study characteristics and critical appraisal

Statistical analysis Included studies consisted of 1 randomized controlled trial


(RCT) (5%), 1 quasi-RCT (5%), 4 prospective cohort studies
All evaluated and reported effects of the timing of spinal surgery (18%), and 16 retrospective comparative cohort studies (73%)
after tSCI were abstracted and assessed for eligibility for meta- (Table 2). Early treatment was defined as spinal surgery performed
analysis (Fig. 1). Outcomes’ point estimates and measures of vari- within 24 and 72 h after injury in ten (46%) and six (27%) studies,
ability (where applicable) had to be reported in at least three studies
respectively. The maximum delay for ‘‘late’’ spinal surgery was 1
in order to be considered for meta-analysis. For continuous out-
comes, for example, the Total Motor Score,17,18 we calculated the week in one study36 (5%), 2–8 weeks in four studies31,39,43,47
weighted mean difference (WMD), that is, the weighted average of (18%), and 6–12 months in two studies33,40 (9%). A majority of 15
the differences in the individual studies, the weight being the indi- studies (68%) did not report the maximum delay for surgery.
vidual inverse variances (i.e., precision) for each study adjusted for Applied as a rough indicator for the level of SCI, half of the
heterogeneity (i.e., the variability in study effect sizes beyond that studies (11) included only tetraplegics, whereas 5 studies (23%)
caused by random error alone). This adjustment was based on as- included only paraplegics. Thirteen studies (59%) included both
sessed study deficiencies with the QE model and simply on statistical complete and incomplete SCI subjects, and seven studies (32%)
variability of effect sizes across studies with the random effects (RE) included only incomplete SCI subjects. Follow-up was notably
model. Similarly, for binary outcomes, for example, occurrence of short for studies focusing on length of stay outcomes with a min-
nonspecific neurological improvements and adverse events, we
imum reported mean duration of 10 days for early treated para-
calculated the OR, again computing a weighted average across
studies as described. The advantage of the QE meta-analysis20,21 is plegics in one study.38 Studies that focused on neurological
that it allows for redistribution of individual studies’ inverse variance outcomes reported longer follow-up periods ranging from 6 months
weights based on assessed study deficiencies rather than the usual to 9 years. Only four studies (18%) reported sufficient details on
random redistribution of weights seen with the RE model.22 The point estimates and measures of variability of the latest follow-up
derived quality score of each individual study was normalized by visits.30,33,39,40
dividing by the maximum possible quality score and indicated as the As indicated by the low Qi values presented in Table 2, included
quality index (Qi, range: 0–1). Subsequently, Qi values were entered studies demonstrated a high susceptibility to bias. A complete set of
in the quality effects model as described by Doi et al.20,21,23,24 assigned quality scores is presented in detail in Supplementary
Statistical heterogeneity was assessed using the I2 and s2 sta- Appendix 3 (see online supplementary material at http://www
tistics. Heterogeneity was regarded as substantial when the I2 sta-
.liebertonline.com).
tistic exceeded 30% and the s2 was > 0. To assess the robustness of
the meta-analysis, we performed one-way and ‘‘criterion-based’’
sensitivity analyses. One-way sensitivity analysis was conducted Quantitative synthesis
by excluding individual studies sequentially from the meta-analy- Neurological outcomes were reported in 19 studies29,31–33,35–
ses. A criterion-based sensitivity analysis looks at the pooled results 37,39–50
(86%), length of stay related outcomes were reported in 12
after altering inclusion criteria for meta-analysis. Studies were
studies30,31,34,37–39,41,42,45,47,48,50 (55%), financial outcomes were
selected and grouped using the following predefined inclusion
criteria: 1) SCI severity, 2) level of SCI, and 3) maximum delay reported in 2 studies34,41 (9%), bladder function outcomes were
‘‘early’’ treatment. The outcomes of a meta-analysis can be con- reported in 2 studies33,37 (9%), and the Functional Independence
sidered robust when sensitivity analyses in subsets of studies show Measure51 and blood loss44 were each reported in 1 study (5%)
similar pooled effect sizes. (Table 2).
To test for small-study effects,25 or potential publication bias,
the symmetry of funnel plots was assessed visually and statistically Neurological outcomes
with the Egger’s linear regression test. Although appreciating other
possible causes of funnel plot asymmetry,26 publication bias was Nine studies29,32,33,37,41,42,45,48,50 (41%) reported the Total
suspected if the intercept of Egger’s regression line deviated from 0 Motor Score, six studies31,36,39,44,49,50 (27%) reported the neuro-
with a p value of < 0$1.27 All statistical analyses were performed logical improvement rate, four studies31,36,41,50 (18%) reported the
Table 2. Study Characteristics of Studies Meeting Inclusion Criteria

Included
Study Study Timing definitions subjects Mean age in SCI SCI Latest ‘‘protocol’’
Source design period (early/late)a Subgroups (early/late) yrs (early/ late)a Level Severity Outcomes follow-upa Qi
b
McAfee et al. 1985 RCS 1973 - 1981 NA - 48 30 (16–65) P I NEU 3.4 (2–8.6) yrs 0.08
Petitjean et al. 1995 RCS 1990–1993 < 24 h/ > 24 h - 10/19 37 – 17 P I,C NEU, LOS, NR 0.08
Other
Wiberg et al. 1988 RCS 1980–1985 < 24 h/ > 24 h, < 7 d/ > 7 d - 8/2/20 NR P I,C NEU (8–72) m 0.12
Anderson et al. 2012 RCS 2000–2009 < 24 h/ > 24 h, < 48 h/ > 48 h - 14/30/25 59 (23–89) T I NEU 11 (6–60) m 0.12
Clohisy et al. 1992 RCS 1981–1990 < 48 h/ > 48 h - 11/9 30 (15–63)/ 38 (17–66) P I NEU, Bladder 42 (12–62) m/42 0.16
(6–92) m
Campagnolo et al. RCS 1990–1996 < 24 h/ > 24 h - 37/27 32/ 42 T,P I,C LOS 38 – 34 d/55 – 40 d 0.16
1997
Pollard et al. 2003 RCS 1982–2000 < 24 h/ > 24 h -c 86/243 34 – 15/ 36 – 16 T C NEU, LOS NR 0.16
Sapkas et al. 2007 RCS 1987–2000 < 72 h/ > 72 h - 31/36 36 (16–72) T I,C NEU 4 (1–9) yrs 0.16
Chen et al.2009 RCS 1999–2004 £ 4 d/ > 4 d - 21/28 56 (22–76) T I NEU 56 (25-NR) m 0.16
Stevens et al. 2010 RCS 1985–2006 < 24 h/ > 24 h - 16/34 NR T I NEU, LOS NR 0.16
Duh et al. 1994 PCS 1985–1988 < 100 h/ > 100 h -c NR NR T,P I,C NEU 1 yr/1 yr 0.20
Newton et al. 2011 RCS 1988–2000 NA{ - 57 22 (14–47) T I,C NEU NR 0.20
Levi et al. 1991 PCS 1985–1990 < 24 h/ > 24 h I 10/40 30/33 T I,C NEU, LOS 9 (6–12) m/10 0.24

1785
(6–12) m
C 35/18 25/28 11 (9–12) m/12
(9–12) m
Mirza et al. 1999 RCS 1989–1991 < 72 h/ > 72 h - 15/15 32 (14–56)/33 (21–49) T I,C NEU, LOS NR 0.24
Croce et al. 2001 RCS 1996–1999 < 72 h/ > 72 h - 71/74 34/34 T,P NR LOS, Costs 29 d/36 d 0.24
Guest et al. 2002 RCS 1986–1996 < 24 h/ > 24 h - 16/34 41 – 20/48 – 20 T I NEU, LOS, 36 (13–48) m 0.24
Bladder
Kerwin et al. 2005 RCS 1988–2001 < 72 h/ > 72 h T 59/27 37/39 T,P NR LOS 21 d/24 d 0.24
P 31/15 38/38 10 d/31 d
McKinley et al. 2004 RCS 1995–2000 < 72 h/ > 4 d -c 307/296 37 – 15/36 – 16 T,P I,C NEU, LOS, 1 yr/1 yr 0.28
Costs, Other
Vaccaro et al. 1997 RCT 1992–1995 < 72 h/ > 5 d - 35/29 40 (15–75)/39 (16–75) T I,C NEU, LOS 10 m/ 3 m 0.32
Wilson et al. 2012 PCS 2007–2009 < 24 h/ > 24 h - 35/49 42/48 T,P I,C NEU, LOS NR 0.40
Fehlings et al. 2012 PCS 2002–2009 < 24 h/ > 24 h, £ 7 d - 182/131 45 – 17/51 – 16 T I,C NEU 6 m/6 m 0.52
Cengiz et al. 2008 Quasi- 2004–2006 < 8 h/ > 72 h, < 15d - 12/15 40 – 16/43 – 14 P I,C NEU, LOS 15 (12–20) m 0.56
RCT
a
A slash (‘/’) denotes details for both early and late groups were presented. Values in parentheses are ranges of mean values, when reported. bThe timing of surgical spinal decompression was processed as a
continuous measure in this study. cVarious subgroup analyses were presented in this article. However, as no details of the subgroups were presented these analyses were not included in the current review.
RCS, retrospective cohort study; PCS, prospective cohort study; RCT, randomized controlled trial; h: hour(s); T, tetraplegia; P, paraplegia; I, incomplete; C, complete; NEU, neurological; LOS, length of stay; Qi,
‘‘quality’’ index value (see Supplementary Appendix 3), NR, not reported; NA, not applicable.
1786 VAN MIDDENDORP ET AL.

FIG. 2. Forest plot of the Total Motor Score weighted mean differences (WMDs) in individual studies and pooled estimate using the
quality effects model.

ASIA Impairment Scale, and four studies42,46–48 (14%) reported the with the QE model are shown in Figure 2. The pooled WMD of the
Frankel scale. In addition, a variety of neurological aggregate Total Motor Score between ‘‘early’’ and ‘‘late’’ surgical spinal
scores, sensory scores, and level of injury measures were each decompression was 5$94 (95% CI: 0.74, 11.15), indicating that, on
reported in one or two studies.35,37,39,41,45 The Total Motor Score average, subjects who undergo early spinal surgery gain approxi-
and the neurological improvement rate outcomes were suitable mately six motor score points more than subjects who undergo late
measures for meta-analysis. treatment (Table 3). The RE model showed a slightly smaller
The mean Total Motor Score improvement was specified for pooled WMD of 4$73 (95% CI: - 0$13, 9$59) points, with 0 now
both ‘‘early’’ and ‘‘late’’ groups in seven studies (32%), comprising being included in the CI. For details see Supplementary Appendix 4
815 pooled subjects. Studies were considered heterogeneous (see online supplementary material at http://www.liebertonline
(s2 > 0; I2 = 52%; p = 0$05), and the QE model was used to pool the .com).
data, with the RE model being used for standardized comparison. The neurological improvement rate was specified in six studies
The studies’ individual and pooled WMDs and 95% CIs obtained (27%), comprising 495 pooled subjects. As a measure of

Table 3. Pooled Effects of Analyzed Outcomes Using Quality Effect, One-Way Sensitivity Analyses
and Random Effect Models

Studies/
Outcome and statistic comparisons Model Pooled effect Excluded study

Total Motor Score improvement 7/7 QE 5. 94 (0.74, 11.15) -


WMD QE-1WS: Early + 8.16 (1.61, 14.71) McKinley et al.2004
QE-1WS: Late + 3.62 ( - 1.15, 8.40) Mirza et al.1999
RE 4.73 ( - 0.13, 9.59) -
Neurological improvement rate 6/9 QE 2.23 (1.35, 3.67) -
OR QE-1WS: Early + 3.11 (1.50, 6.44) Fehlings et al. 2012
QE-1WS: Late + 1.60 (1.02, 2.50) Cengiz et al.2008
RE 1.74 (1.04, 2.91) -
Length of hospitalization (days) 6/6 QE - 9.98 ( - 13.10, - 6.85) -
WMD QE-1WS: Early + - 11.61 ( - 16.49, - 6.73) McKinley et al.2004
QE-1WS: Late + - 8.72 ( - 11.97, - 5.47) Cengiz et al.2008
RE - 8.51 ( - 12.78, - 4.25) -
Mortality 9/14 QE 0.97 (0.40, 2.31) -
OR QE-1WS: Early + 0.81 (0.31, 2.09) Fehlings et al. 2012 (FU)
QE-1WS: Late + 1.25 (0.48, 3.28) Kerwin et al. 2005
RE 0.70 (0.35, 1.41) -
Adverse events (all) 12/28 QE 0.71 (0.49, 1.04) -
OR RE 0.86 (0.69, 1.07) -

Values in parentheses are 95% confidence intervals.


WMD, weighted mean differences; OR, odds ratio; QE, quality effects; QE-1WS, quality effects one-way sensitivity analysis; RE, random effects;
Early + , exclusion of one study resulting in a pooled effect most favoring ‘‘early’’ spinal surgery, Late + , exclusion of one study resulting in a pooled
effect most favoring ‘‘late’’ spinal surgery.
TIMING OF SPINAL SURGERY AFTER tSCI 1787

improvement, two studies applied one ASIA Impairment Scale reported two comparisons: two studies evaluated two sub-
grade,36,50 one study applied one or more ASIA Impairment Scale groups,38,39 two studies included two ‘‘late’’ comparison
grades,31 one study applied one or more Frankel scale grades,49 and groups,29,49 and one study evaluated short- and long-term follow-
two studies applied ‘‘any’’39 or ‘‘good’’44 improvement. Three up.36 This resulted in a total of 14 comparisons in the QE model,
studies performed either two subgroup analyses39,44 or included comprising 1148 pooled subjects. The ORs were considered ho-
two ‘‘late’’ comparison groups,49 resulting in a total of nine com- mogeneous (s2 = 0; I2 = 0%; p = 0$64). The pooled OR for mortality
parisons for meta-analysis. The ORs were considered heteroge- was 0$97 (95% CI: 0$40, 2$31), indicating no differences in
neous (s2 > 0; I2 = 15%; p = 0$31). The pooled OR for neurological mortality risks between subjects undergoing ‘‘early’’ and ‘‘late’’
improvement was 2$23 (95% CI: 1$35, 3$76) indicating a twice as spinal surgery (Table 3 and Fig. 4). The RE model showed a similar
high probability of improvement for subjects undergoing ‘‘early’’ pooled OR of 0$70 (95% CI: 0$35, 1$41). For details, see Sup-
intervention (Table 3). The RE model again showed a slightly plementary Appendix 7 (see online supplementary material at
lower pooled OR of 1$74 (95% CI: 1$04, 2$91). Details of both http://www.liebertonline.com).
models are presented in Supplementary Appendix 5 (see online
supplementary material at http://www.liebertonline.com). Other adverse events
Of the 19 studies (86%) reporting on adverse events, 5 studies
Length of stay outcomes specified all reported adverse events in both groups (see Supple-
mentary Appendix 3). However, 12 studies (55%) reported at least
Twelve studies30,31,34,37–39,41,42,45,47,48,50 (55%) reported the
one adverse event in both groups and, therefore, were suitable for
length of stay in hospital and/or rehabilitation center, six stud-
meta-analysis. Twenty-eight comparisons were reported in the
ies31,34,37,38,42,47 (27%) reported the length of stay in the intensive
following (post-hoc devised) subgroups: neurological deterioration
care unit, and three studies34,38,42 (14%) reported the duration of
(five studies, 23%),36,37,42,45,49 thromboembolic events (two stud-
mechanical ventilation. The length of stay in hospital was the single
ies, 9%),36,41 (other) pulmonary events (four studies,
suitable measures for meta-analysis.
23%),31,34,38,41 sepsis (three studies, 14%),31,36,49 wound and skin
The mean length of hospital stay was specified in six studies
events (three studies, 14%),33,36,41 number of patients with adverse
(27%), comprising 1103 pooled subjects. Studies were considered
events (two studies, 9%),39,47 and various other adverse events – six
heterogeneous (s2 > 0; I2 = 71%; p < 0$01). The pooled WMD of
in total – in four studies.33,36,37,41 Outcomes of analyses on all
the length of hospital stay was - 9$98 (95% CI: - 13$10, - 6$85),
adverse events combined are presented in Table 3.
indicating that on average, subjects who undergo early spinal sur-
No statistically significant differences were found for the sub-
gery spend 10 days less in hospital than do subjects who undergo
groups using the QE model. However, the RE model demonstrated
late treatment (Table 3 and Fig. 3). The RE model showed a smaller
a statistically significant pooled effect (OR: 0$67, 95% CI: 0$51,
pooled WMD of - 8$51 (95% CI: - 12$78, - 4$25). For details see
0$89) for pulmonary events, indicating that subjects who under-
Supplementary Appendix 6 (see online supplementary material at
went ‘‘late’’ surgical intervention had a higher risk of developing
http://www.liebertonline.com).
pulmonary complications, for example, pneumonia and atelectasis,
than did ‘‘early’’ treated subjects. However, caution is required
Mortality
in interpretation of such random effect results, particularly
Out of the 10 studies29,31,34,36,38,39,46–49 (46%) that reported on when considering that no significant effects were observed in the
mortality outcomes, 9 were suitable for meta-analysis. Five studies quality effect model. Details of both models are presented in

FIG. 3. Forest plot of the length of hospital stay weighted mean differences (WMDs, in days) in individual studies and pooled estimate
using the quality effects model.
1788 VAN MIDDENDORP ET AL.

FIG. 4. Forest plot of mortality odds ratios (ORs) in individual studies and pooled estimate using the quality effects model.

Supplementary Appendix 8 (see online supplementary material at Results of the ‘‘criterion-based’’ sensitivity analyses are pre-
http://www.liebertonline.com). sented in Table 4. Pooled estimates for the Total Motor Score
suggested smaller improvements for studies that included incom-
plete SCI subjects only, included both tetraplegics and paraplegics,
Sensitivity analysis and publication bias
and – unexpectedly – considered ‘‘early’’ surgery as being within
One-way sensitivity analyses with each study individually re- 24 h instead of later (Table 4). Summary estimates for the neuro-
moved was done for all performed QE models, see Table 3. The logical improvement rate suggested smaller improvements for
pooled estimate for Total Motor Score improvement varied con- studies that included incomplete SCI subjects or tetraplegics only.
siderably after having excluded a single study resulting in a pooled Pooled estimates for length of hospital stay outcomes suggested
effect most (8$16, 95% CI: 1$61, 14$71)41 and least (3$62, 95% smaller differences for studies that included incomplete SCI sub-
CI: - 1$15, 8$40)42 favoring ‘‘early’’ spinal surgery. Similar effect jects only. Mortality sensitivity analyses demonstrated substan-
modifications were seen for the neurological improvement rate OR tially dispersed pooled risks for all criteria. The summary risk
analyses. Summary tables with data from the one way sensitivity estimate for adverse events was sensitive to change when tested for
analyses are presented in Supplementary Appendix 9 (see online severity of injury (incomplete SCI: higher risk) and timing of
supplementary material at http://www.liebertonline.com). ‘‘early’’ surgery ( £ 24 h: lower risk, Table 4).
TIMING OF SPINAL SURGERY AFTER tSCI 1789

1.34 (0.38, 4.73)


0.65 (0.41, 1.02)

0.70 (0.42, 1.15)


0.68 (0.38, 1.20)

0.58 (0.34, 0.97)


0.90 (0.59, 1.38)

0.73 (0.18, 2.90)


0.71 (0.47, 1.07)
Presence of small-study effects was assessed by evaluating
funnel plots for symmetry both visually and statistically. A possi-
bility of publication bias was found for the following outcomes:

OR
events (all)
Adverse
Total Motor Score (intercept: 1$85, 95% CI: 0$66, 3$03; p = 0$01),
‘‘neurological improvement rate’’ (intercept: 0$9, 95% CI: - 0$05,
1$85; p = 0$06), and length of hospital stay (intercept: - 2$53, 95%
CI: - 4$76, - 0$29; p = 0$04) (Fig. 5a–c). The funnel plots clearly
illustrate that for all three outcomes it were the studies with smaller

5
21

13
16

16
13

6
23
n
samples that favored ‘‘early’’ spinal surgery.

1.07 (0.18, 6.49)


1.32 (0.37, 4.72)

0.90 (0.35, 2.33)


1.16 (0.19, 7.17)

1.32 (0.41, 4.29)


0.56 (0.16, 1.98)

0.94 (0.17, 5.08)


0.97 (0.36, 2.61)
Discussion
This meta-analysis demonstrated that patients who underwent

OR
Mortality
‘‘early’’ spinal surgery after tSCI had significantly greater neuro-

Table 4. Results of Criterion-Based Sensitivity Analyses Using a Quality Effects Model


logical recovery outcomes and significantly shorter hospital ad-
missions than did patients who underwent ‘‘late’’ spinal surgery.
However, this study also demonstrated that these findings are far
from robust, as estimates dispersed substantially in both one way

4b
7b

8
6

10
4

5
9
and criterion-based sensitivity analyses and funnel plots also

n
demonstrated a high likelihood of publication bias. Two major
sources of heterogeneity were identified: 1) patients with a variety

- 11.12 ( - 14.66, - 7.58)

- 8.70 ( - 13.97, - 3.43)


- 10.55 ( - 14.39, - 6.71)

- 10.96 ( - 16,36, - 5.56)


- 8.67 ( - 11.76, - 5.57)

- 8.80 ( - 15.75, - 1.86)


- 10.41 ( - 13.90, - 6.93)
of SCI severities and levels of injury were included in original

- 4.77 ( - 10.22, 0.68)


studies and 2) included studies demonstrated a high susceptibility

hospital stay (days)


to various sources of bias.
To our knowledge, this is the first literature review to present and

WMD
Length of
integrate both qualitative and quantitative data demonstrating a
profound heterogeneity among original studies reporting on the
effects of the timing of spinal surgery after tSCI. Although basic
methodological limitations have been addressed in previous re-
views and meta-analyses,5–10 no study has yet quantified the po-
tential impact of studies’ susceptibility to bias on pooled treatment

Median Quality index score = 0.20; btwo studies did not report the severity of the injury, see Table 2.
effect estimates. Previous reviews have addressed the methodo-

3
n

2
4

4
2

3
3
logical limitations of original studies, generally using an almost
instinctive, standard closing sentence referring to ‘‘the need for
3.86 (1.49, 10.03)

3.75 (0.57, 24.48)


randomized controlled trials.’’ However, clinical equipoise – 1.42 (0.88, 2.28)
1.35 (0.33, 5.44)
2.36 (1.35, 4.10)

2.13 (1.24, 3.66)


improvement rate

defined as a state of genuine uncertainty on the part of the clinical


Neurological

investigator regarding the comparative therapeutic merits of each


OR

-
-
arm in a trial52 – has lost ground, as a recent survey indicated an
outspoken preference for ‘‘early’’ spinal decompression among
spine surgeons.53 This essential aspect, combined with a number of

SCI, spinal cord injury; WMD, weight mean difference, OR, odds ratio.
other feasibility issues,54 makes conducting a properly powered,
multicenter, surgical RCT on tSCI patients a very challenging, if
3
n

2
7

9
0

4
5
not unfeasible, undertaking.54 The current study indicates that
much more can – and needs to be – done in addressing other sources
of heterogeneity than confounding by indication alone.
6.01 ( - 2.86, 14.88)

7.07 ( - 0.20, 14.34)

5.19 ( - 1.79, 12.17)


2.94 ( - 3.54, 9.43)

2.06 ( - 2.84, 6.95)


7.52 (0.44, 14.60)
5.54 (0.90, 10.18)

7.34 (0.92, 13.77)

The first identified source of heterogeneity relates to the variety


Values in parentheses are 95% confidence intervals.

of SCI severities and levels of injury included in original studies.


WMD
Total motor

SCI is a complex condition that presents with markedly heteroge-


score

neous manifestations related to the severity and anatomic level of


injury.55 Moreover, SCI patients may also present with concomi-
tant injuries and pre-existent comorbidities.56,57 Together with
age,58 these patient related factors have been demonstrated to be
strongly related to patient outcomes after tSCI.58–60 In the current
5
4
3

2
5

3
4

review, we found that 1) not a single study reported a complete


overview of patients’ age, SCI level, SCI severity, concomitant
Timing ‘‘early’’ group

injuries and co-morbidities; 2) only 2 of the 14 studies that included


Paraplegia w/wo
Tetraplegia only
Incomplete only
Complete w/wo

more than one type of severity and/or level of SCI, stratified sub-
Tetraplegia
Incomplete

Quality scorea

jects accordingly;38,39 and 3) only 4 of the 21 nonrandomized


£ 24 hours
> 24 hours

> median
SCI severity

£ median

studies (19%) applied statistical techniques to adjusted evaluated


SCI level
Criterion

outcome measures for confounding.35,36,40,50


Although beyond the scope of this review, it is worth mentioning
a

several countermeasures to address observed confounders in non-


1790 VAN MIDDENDORP ET AL.

FIG. 5. Funnel plots for Total Motor Score (a), neurological improvement rate (b), and length of hospital stay (c).
TIMING OF SPINAL SURGERY AFTER tSCI 1791

randomized comparative studies, including matching, stratifica- Although acknowledging the inherent limitations of applied
tion, and multivariate regression techniques.61 Statistical tech- sensitivity and small-study effects analyses, several findings merit
niques to adjust for unobserved, or unconsidered, confounders have further consideration. One-way sensitivity analyses indicated that
been documented, albeit such adjustments require a well-defined the pooled effects of both the Total Motor Score improvement and
high-volume cohort of patients.62 Finally, propensity scores can be the neurological improvement rate dispersed substantially after
used to balance the covariates in the two study groups, and thus excluding one single study from the analysis. In addition, criterion-
reduce the magnitude of selection bias in nonrandomized studies.63 based sensitivity analyses demonstrated that none of the pooled
The second identified source of heterogeneity relates to the in- neurological outcome estimates were robust enough after stratify-
dividual studies’ susceptibility to bias other than confounding by ing by each of the predefined criteria. These sensitivity analyses
indication. This was assessed using a tailored scoring system de- clearly demonstrate the lack of robustness in these data and,
vised based on items covered by three consensus-derived therefore, do not validate the statistically significant outcomes of
‘‘checklists.’’12–14 Where these checklists mainly focus on the the current meta-analysis. No significant differences in mortality
detail of reporting, the devised scoring system was primarily in- were observed between the ‘‘early’’ and ‘‘late’’ treatment groups.
tended as a tool to assess individual, surgical studies’ susceptibility In the light of potential selection bias, one might expect to see a
to bias, or methodological quality. Some authors state that a study’s higher mortality rate the ‘‘late’’ group. However, as only a few
true susceptibility to bias can only be assessed through its surrogate studies reported the required information to explore this hypothesis,
measure: the quality of reporting.64 Although this argument makes no conclusive findings can be drawn from the current meta-
sense from a non-reporting perspective, it does not hold from a analysis. Finally, results from the funnel plots and related Egger
‘‘do-reporting’’ perspective. To illustrate, author group ‘‘A’’ re- regression coefficients cannot be mistaken; there is a clear pre-
ports that the ‘‘treatment allocation was random, and double-blind’’ ponderance of smaller studies reporting effects favoring ‘‘early’’
and author group ‘‘B’’ reports that ‘‘treatment allocation was nei- spinal surgery. Without getting into a ‘‘chicken and egg’’ conun-
ther random, nor blind.’’ Both reports score a high ‘‘quality of drum, the likelihood of observed publication bias coincides well
reporting’’ on this item, however, only group ‘‘A’’’s report scores a with recently reported spine surgeons’ outspoken preference for
high ‘‘methodological quality.’’ Nonetheless, most of the meth- ‘‘early’’ spinal decompression after tSCI.53
odological safeguards assessed in the current review were not re- No consensus exists as to what constitutes a clinically mean-
ported in original studies, and, therefore, may have resulted in an ingful neurological improvement. Where an improvement of five
overestimation of included studies’ true susceptibility to bias. Motor Score points may seem to be trivial for clinicians and re-
The specific sources of individual studies’ susceptibility to bias searchers, minimal neurological improvements could result in
are outlined in Supplementary Appendix 3. Although the six profound functional benefits, particularly in tetraplegic patients.
prospective studies tended to present higher Qi values, the single However, only three studies related the clinical meaningfulness
included RCT48 did not reach the highest quality score. Whereas of observed neurological improvements to functional outcomes,
random treatment allocation is a key methodological safeguard, that is, bladder function33,37 and the Functional Independence
other sources of heterogeneity than confounding by indication Measure.51
alone may have an equally striking impact on studies’ validity. No Illustrated by the variety of applied definitions for ‘‘early’’ spinal
study performed and reported a sample size calculation, and only surgery in included studies, there is no consensus on what consti-
two studies (9%) did report a primary outcome measure required tutes the threshold value distinguishing ‘‘early’’ from ‘‘late’’ sur-
for determining a sample size. Whereas the manifestations of SCI gery. More than 100 years after Burrell’s problem definition,1 it is
are highly variable, even more so are its recovery patterns. Sample still unclear what the therapeutic window of opportunity after tSCI
size calculations are of crucial importance in addressing the im- is; and both clinical and pre-clinical scientists face a similar di-
pact of this disorder-specific variability on the outcomes of ther- lemma.65 The criterion-based sensitivity analyses demonstrated a
apeutic studies.59 In a recent protocol publication by the counterintuitively, greater neurological benefit in studies consid-
Prospective, Observational European Multicenter study on the ering ‘‘early’’ surgery as within 72 h instead of within 24 h. This
efficacy of acute surgical decompression after traumatic Spinal raises another discussion about the limited reliability of very early
Cord Injury (SCI-POEM) investigators, a stratified sample size neurological examinations.66 Others have stated, however, that
calculation (by ASIA Impairment Scale grades) was reported and when no concomitant injuries are present, and full cooperation can
based on reference data from a large European cohort of SCI be obtained, the timing of the examination itself is not affecting the
patients.54,58 examination’s precision.67 Given that timing of surgery was con-
The heterogeneity found in the current review may raise the sidered as a ‘‘continuous’’ factor in the retrospective study reported
question of whether the eligibility criteria that were applied were by Newton et al.43 (Qi: 0$20) it could not be included in the current
too loose. Tables 2 and 4 perfectly demonstrate, however, that if we analysis. However, using receiver operating characteristics (ROC)
had applied more strict eligibility criteria for the sake of homoge- analysis, the latter authors determined that the optimal cutoff time
neity, only a few studies would have been included, and the gen- for surgical reduction based on the outcome ‘‘complete neurolog-
eralizability of our findings would have been limited. Discussed ical recovery after tSCI’’ was £ 4 h after injury for all patients as
sources of heterogeneity and susceptibility to bias had a profound well as for patients completely paralyzed on admission.43 Once
impact on the quantitative analyses presented in the current review. confirmed in future studies, this finding could have far-reaching
However, the slight differences between the pooled effects and implications for pre-hospital management and logistics.
variance reported in the RE and QE models may suggest otherwise. The variety of approaches for spinal surgery that has been ap-
The explanation for this is straightforward: as the Qi median value plied within and between original studies has frequently been un-
of 0$20 indicates, a large majority of studies were susceptible to derappreciated in previous literature reviews. The current review
multiple sources of bias. As such, quality discounting was not demonstrated that the surgical approaches applied in both the
limited to a few studies and resulted in an inevitable ‘‘quality ho- ‘‘early’’ and ‘‘late’’ groups were reported and comparable in not
mogeneity paradox.’’ more than two studies (9%).31,39 In the remainder of the studies, it
1792 VAN MIDDENDORP ET AL.

was not clear whether study groups had a comparable number of assisted in data abstraction and statistical analysis. Dr. van Mid-
patients subjected to closed, anterior, posterior, or circumferential dendorp drafted the manuscript. All authors critically revised the
surgical approaches. In fact, 10 studies (46%) explicitly reported the manuscript and approved its final version.
term ‘‘decompression,’’ or ‘‘reduction,’’ as part of the intervention
performed in all subjects. Fewer studies reported details on more Author Disclosure Statement
specific surgical techniques believed to play a role in the decom- No competing financial interests exist.
pression of spinal canal, including laminectomies and discectomies.
Only three studies (14%) indicated having performed postsurgical References
imaging (magnetic resonance imaging in one study,36 computed
1. Burrell, H.L. (1905). Fracture of the spine. A summary of all the cases
tomography in two studies31,33) for confirming a successful de- (244) which were treated at the Boston City Hospital from 1864 to
compression of the spinal cord, although no criteria for successful 1905. Ann. Surg. 42, 481–506.
decompression were documented. Where pre-clinical scientists pay 2. Carlson, S.L., Parrish, M.E., Springer, J.E., Doty, K., and Dossett, L.
utmost attention to the standardization of injury models,68 the current (1998). Acute inflammatory response in spinal cord following impact
review clearly uncovered clinical researchers’ limited attention to injury. Exp. Neurol. 151, 77–88.
3. Dimar, J.R., 2nd, Glassman, S.D., Raque, G.H., Zhang, Y.P., and
technical, qualitative, and quantitative aspects of the actual surgical Shields, C.B. (1999). The influence of spinal canal narrowing and
decompression of the spinal cord in their final study reports. timing of decompression on neurologic recovery after spinal cord
Strengths of our study include independent screening and data contusion in a rat model. Spine 24, 1623–1633.
abstraction by two reviewers, the critical appraisal of studies’ sus- 4. Rabinowitz, R.S., Eck, J.C., Harper, C.M., Jr., Larson, D.R., Jimenez,
M.A., Parisi, J.E., Friedman, J.A., Yaszemski, M.J., and Currier, B.L.
ceptibility to bias, the inclusion of a variety of clinically relevant (2008). Urgent surgical decompression compared to methylpredniso-
outcomes, applied statistical techniques for assessing bias in this lone for the treatment of acute spinal cord injury: a randomized pro-
review, and reporting according to the PRISMA statement’s rec- spective study in beagle dogs. Spine (Phila Pa 1976) 33, 2260–2268.
ommendations.26 Nonetheless, several potential limitations of this 5. Ball, J.R., and Sekhon, L.H. (2006). Timing of decompression and
review exist. Although the tailored bias assessment scoring instru- fixation after spinal cord injury—when is surgery optimal? Crit. Care
Resusc. 8, 56–63.
ment was based on items from established consensus-derived 6. Kishan, S., Vives, M.J., and Reiter, M.F. (2005). Timing of surgery
‘‘checklists,’’12–14 no validation study has demonstrated its psy- following spinal cord injury. J. Spinal Cord Med. 28, 11–19.
chometric properties. Pooling of outcome measures, including the 7. Schinkel, C., and Anastasiadis, A.P. (2008). The timing of spinal
neurological improvement rate, may have resulted in additional stabilization in polytrauma and in patients with spinal cord injury.
Curr. Opin. Crit. Care 14, 685–689.
‘‘analytical heterogeneity.’’ Including two ‘‘late’’ surgery compari-
8. Fehlings, M.G., and Perrin, R.G. (2006). The timing of surgical in-
son groups in analyses led to doubling of ‘‘early’’ intervention group tervention in the treatment of spinal cord injury: a systematic review
data and may, therefore, have resulted in distorted outcomes. Sen- of recent clinical evidence. Spine 31, S28–36.
sitivity analyses may have limited specificity, as the predefined cri- 9. Furlan, J.C., Noonan, V., Cadotte, D.W., and Fehlings, M.G. (2011).
teria were not adjusted. We did not perform a meta-regression Timing of decompressive surgery of spinal cord after traumatic spinal
cord injury: an evidence-based examination of pre-clinical and clinical
analysis, as the number of included studies was too small and the studies. J. Neurotrauma 28, 1371–1399.
overall quality of included studies was too low.26 Finally, studies that 10. La Rosa, G., Conti, A., Cardali, S., Cacciola, F., and Tomasello, F.
did evaluate the effect of timing of surgical spinal decompression as (2004). Does early decompression improve neurological outcome of
a continuous variable could not be included in meta-analysis. spinal cord injured patients? Appraisal of the literature using a meta-
analytical approach. Spinal Cord 42, 503–512.
11. Lemeshow, A.R., Blum, R.E., Berlin, J.A., Stoto, M.A., and Colditz,
Conclusion G.A. (2005). Searching one or two databases was insufficient for meta-
analysis of observational studies. J. Clin. Epidemiol. 58, 867–873.
In conclusion, despite the fact that ‘‘early’’ spinal surgery was 12. Boutron, I., Moher, D., Altman, D.G., Schulz, K.F., and Ravaud, P.
significantly associated with improved neurological and length of (2008). Extending the CONSORT statement to randomized trials of
nonpharmacologic treatment: explanation and elaboration. Ann. In-
stay outcomes, none of these positive associations were robust, as tern. Med. 148, 295–309.
demonstrated through sensitivity analyses. Moreover, funnel plots 13. van Tulder, M.W., Assendelft, W.J., Koes, B.W., and Bouter, L.M.
showed significant proof of publication bias. Therefore, the au- (1997). Method guidelines for systematic reviews in the Cochrane
thors’ verdict is that because of different sources of heterogeneity Collaboration Back Review Group for Spinal Disorders. Spine (Phila
Pa 1976) 22, 2323–2330.
within and between original studies, there is no robust evidence
14. von Elm, E., Altman, D.G., Egger, M., Pocock, S.J., Gotzsche, P.C.,
supporting ‘‘early’’ spinal surgery after tSCI. However, what evi- and Vandenbroucke, J.P. (2007) Strengthening the Reporting of Ob-
dence there is clearly supports early intervention. Where properly servational Studies in Epidemiology (STROBE) statement: guidelines
powered RCTs have proven to be an almost unfeasible study design for reporting observational studies. BMJ 335, 806–808.
for acute, surgical tSCI management, we discussed several ap- 15. Huwiler–Muntener, K., Juni, P., Junker, C., and Egger, M. (2002).
Quality of reporting of randomized trials as a measure of methodo-
proaches that can be implemented in future nonrandomized studies, logic quality. JAMA 287, 2801–2804.
and subsequently diminish their susceptibility to bias. By endorsing 16. Juni, P., Altman, D.G., and Egger, M. (2001). Systematic reviews in
such a high-quality standard, we may well find a definitive answer health care: Assessing the quality of controlled clinical trials. BMJ
to the hypotheses postulated by Burrell more than a century ago. 323, 42–46.
17. American Spinal Injury Association (Revised 2000). International
Standards for Neurological Classification 0f Spinal Cord Injury.
Acknowledgments American Spinal Injury Association: Chicago.
18. Kirshblum, S.C., Burns, S.P., Biering–Sorensen, F., Donovan, W.,
We thank Michael Schuetz for his contributions during the early Graves, D.E., Jha, A., Johansen, M., Jones, L., Krassioukov, A.,
phases of this research project. Drs. van Middendorp and Doi Mulcahey, M.J., Schmidt–Read, M., and Waring, W. (2011). Inter-
national standards for neurological classification of spinal cord injury.
contributed to the design of this research project. Drs. van Mid- J. Spinal Cord Med. 34, 535–546.
dendorp and Hosman conducted the search and abstracted the data. 19. Frankel, H.L., Hancock, D.O., Hyslop, G., Melzak, J., Michaelis, L.S.,
Dr. van Middendorp performed the statistical analysis. Dr. Doi Ungar, G.H., Vernon, J.D., and Walsh, J.J. (1969). The value of
TIMING OF SPINAL SURGERY AFTER tSCI 1793

postural reduction in the initial management of closed injuries of the 42. Mirza, S.K., Krengel, W.F., 3rd, Chapman, J.R., Anderson, P.A.,
spine with paraplegia and tetraplegia. I. Paraplegia 7, 179–192. Bailey, J.C., Grady, M.S., and Yuan, H.A. (1999). Early versus de-
20. Doi, S.A., and Thalib, L. (2008). A quality-effects model for meta- layed surgery for acute cervical spinal cord injury. Clin. Orthop. Relat.
analysis. Epidemiology 19, 94–100. Res. 359, 104–114.
21. Doi, S.A., and Thalib, L. (2009). An alternative quality adjustor for the 43. Newton, D., England, M., Doll, H., and Gardner, B.P. (2011). The case for
quality effects model for meta-analysis. Epidemiology 20, 314. early treatment of dislocations of the cervical spine with cord involvement
22. Al Khalaf, M.M., Thalib, L., and Doi, S.A. (2011). Combining het- sustained playing rugby. J. Bone Joint Surg. Br. 93, 1646–1652.
erogenous studies using the random-effects model is a mistake and 44. Petitjean, M.E., Mousselard, H., Pointillart, V., Lassie, P., Senegas, J.,
leads to inconclusive meta-analyses. J. Clin. Epidemiol. 64, 119–123. and Dabadie, P. (1995). Thoracic spinal trauma and associated in-
23. Doi, S.A., Barendregt, J.J., and Mozurkewich, E.L. (2012). Meta- juries: should early spinal decompression be considered? J. Trauma
analysis of heterogeneous clinical trials: an empirical example. Con- 39, 368–372.
temp. Clin. Trials 32, 288–298. 45. Pollard, M.E., and Apple, D.F. (2003). Factors associated with im-
24. Doi, S.A., Barendregt, J.J., and Onitilo, A.A. (2013). Methods for the proved neurologic outcomes in patients with incomplete tetraplegia.
bias adjustment of meta-analyses of published observational studies. J. Spine (Phila Pa 1976) 28, 33–39.
Eval. Clin. Pract. 19, 653–657. 46. Sapkas, G.S., and Papadakis, S.A. (2007). Neurological outcome
25. Sterne, J.A., Egger, M., and Smith, G.D. (2001). Systematic reviews in following early versus delayed lower cervical spine surgery. J. Orthop.
health care: Investigating and dealing with publication and other Surg. (Hong Kong) 15, 183–186.
biases in meta-analysis. BMJ 323, 101–105. 47. Stevens, E.A., Marsh, R., Wilson, J.A., Sweasey, T.A., Branch, C.L.,
26. Liberati, A., Altman, D.G., Tetzlaff, J., Mulrow, C., Gotzsche, P.C., Jr., and Powers, A.K. (2010). A review of surgical intervention in the
Ioannidis, J.P., Clarke, M., Devereaux, P.J., Kleijnen, J., and Moher, setting of traumatic central cord syndrome. Spine J 10, 874–880.
D. (2009). The PRISMA statement for reporting systematic reviews 48. Vaccaro, A.R., Daugherty, R.J., Sheehan, T.P., Dante, S.J., Cotler,
and meta-analyses of studies that evaluate healthcare interventions: J.M., Balderston, R.A., Herbison, G.J., and Northrup, B.E. (1997).
explanation and elaboration. BMJ 339, b2700. Neurologic outcome of early versus late surgery for cervical spinal
27. Egger, M., Davey Smith, G., Schneider, M., and Minder, C. (1997). Bias cord injury. Spine (Phila Pa 1976) 22, 2609–2613.
in meta-analysis detected by a simple, graphical test. BMJ 315, 629–634. 49. Wiberg, J., and Hauge, H.N. (1988). Neurological outcome after
28. Moher, D., Liberati, A., Tetzlaff, J., and Altman, D.G. (2009). Pre- surgery for thoracic and lumbar spine injuries. Acta Neurochir. (Wien)
ferred reporting items for systematic reviews and meta-analyses: the 91, 106–112.
PRISMA statement. BMJ 339, b2535. 50. Wilson, J.R., Singh, A., Craven, C., Verrier, M.C., Drew, B., Ahn, H.,
29. Anderson, D.G., Sayadipour, A., Limthongkul, W., Martin, N.D., Ford, M., and Fehlings, M.G. (2012). Early versus late surgery for
Vaccaro, A., and Harrop, J.S. (2012). Traumatic central cord syn- traumatic spinal cord injury: the results of a prospective Canadian
drome: neurologic recovery after surgical management. Am. J. Or- cohort study. Spinal Cord 50, 840–843.
thop. (Belle Mead NJ) 41, E104–108. 51. Keith, R.A., Granger, C.V., Hamilton, B.B., and Sherwin, F.S. (1987).
30. Campagnolo, D.I., Esquieres, R.E., and Kopacz, K.J. (1997) Effect of The functional independence measure: a new tool for rehabilitation.
timing of stabilization on length of stay and medical complications Adv. Clin. Rehabil. 1, 6–18.
following spinal cord injury. J. Spinal Cord. Med 20, 331–334. 52. Freedman, B. (1987). Equipoise and the ethics of clinical research. N.
31. Cengiz, S.L., Kalkan, E., Bayir, A., Ilik, K., and Basefer, A. (2008). Engl. J. Med. 317, 141–145.
Timing of thoracolomber spine stabilization in trauma patients; impact 53. Fehlings, M.G., Rabin, D., Sears, W., Cadotte, D.W., and Aarabi, B.
on neurological outcome and clinical course. A real prospective (rct) (2010). Current practice in the timing of surgical intervention in spinal
randomized controlled study. Arch. Orthop. Trauma Surg. 128, 959–966. cord injury. Spine (Phila Pa 1976) 35, S166–173.
32. Chen, L., Yang, H., Yang, T., Xu, Y., Bao, Z., and Tang T. (2009). 54. van Middendorp, J.J., Barbagallo, G., Schuetz, M., and Hosman, A.J.
Effectiveness of surgical treatment for traumatic central cord syn- (2012). Design and rationale of a Prospective, Observational European
drome. J. Neurosurg. Spine 10, 3–8. Multicenter study on the efficacy of acute surgical decompression
33. Clohisy, J.C., Akbarnia, B.A., Bucholz, R.D., Burkus, J.K., and after traumatic Spinal Cord Injury: the SCI-POEM study. Spinal Cord
Backer, R.J. (1992) Neurologic recovery associated with anterior de- 50, 686–694.
compression of spine fractures at the thoracolumbar junction (T12- 55. Tuszynski, M.H., Steeves, J.D., Fawcett, J.W., Lammertse, D., Ka-
L1). Spine (Phila Pa 1976) 17, S325–330. lichman, M., Rask, C., Curt, A., Ditunno, J.F., Fehlings, M.G., Guest,
34. Croce, M.A., Bee, T.K., Pritchard, E., Miller, P.R., and Fabian, T.C. J.D., Ellaway, P.H., Kleitman, N., Bartlett, P.F., Blight, A.R., Dietz,
(2001). Does optimal timing for spine fracture fixation exist? Ann. V., Dobkin, B.H., Grossman, R., and Privat, A. (2007). Guidelines for
Surg. 233, 851–858. the conduct of clinical trials for spinal cord injury as developed by the
35. Duh, MS., Shepard, M.J., Wilberger, J.E., and Bracken, M.B. (1994) The ICCP Panel: clinical trial inclusion/exclusion criteria and ethics.
effectiveness of surgery on the treatment of acute spinal cord injury and its Spinal Cord 45, 222–231.
relation to pharmacological treatment. Neurosurgery 35, 240–249. 56. Furlan, J.C., Kattail, D., and Fehlings, M.G. (2009). The impact of co-
36. Fehlings, M.G., Vaccaro, A., Wilson, J.R., Singh, A.D.W.C., Harrop, morbidities on age-related differences in mortality after acute trau-
J.S., Aarabi, B., Shaffrey, C., Dvorak, M., Fisher, C., Arnold, P., matic spinal cord injury. J. Neurotrauma 26, 1361–1367.
Massicotte, E.M., Lewis, S., and Rampersaud, R. (2012) Early versus 57. Putz, C., Schuld, C., Akbar, M., Grieser, T., Wiedenhofer, B., Fur-
delayed decompression for traumatic cervical spinal cord injury: re- stenberg, C.H., Gerner, H.J., and Rupp, R. (2011) Neurological and
sults of the Surgical Timing in Acute Spinal Cord Injury Study functional recovery in multiple injured patients with paraplegia: out-
(STASCIS). PLoS One 7, e32037. come after 1 year. J. Trauma 70, 1078–1085.
37. Guest, J., Eleraky, M.A., Apostolides, P.J., Dickman, C.A., and 58. van Middendorp, J.J., Hosman, A.J., Donders, A.R., Pouw, M.H.,
Sonntag, V.K. (2002). Traumatic central cord syndrome: results of Ditunno, J.F., Jr., Curt, A., Geurts, A.C., and Van de Meent, H. (2011).
surgical management. J. Neurosurg. 97, 25–32. A clinical prediction rule for ambulation outcomes after traumatic
38. Kerwin, A.J., Frykberg, E.R., Schinco, M.A., Griffen, M.M., Murphy, spinal cord injury: a longitudinal cohort study. Lancet 377, 1004–
T., and Tepas, J.J. (2005). The effect of early spine fixation on non- 1010.
neurologic outcome. J. Trauma 58, 15–21. 59. Fawcett, J.W., Curt, A., Steeves, J.D., Coleman, W.P., Tuszynski,
39. Levi, L., Wolf, A., Rigamonti, D., Ragheb, J., Mirvis, S., and Robinson, M.H., Lammertse, D., Bartlett, P.F., Blight, A.R., Dietz, V., Ditunno,
W.L. (1991). Anterior decompression in cervical spine trauma: does the J., Dobkin, B.H., Havton, L.A., Ellaway, P.H., Fehlings, M.G., Privat,
timing of surgery affect the outcome? Neurosurgery 29, 216–222. A., Grossman, R., Guest, J.D., Kleitman, N., Nakamura, M., Gaviria,
40. McAfee, P.C., Bohlman, H.H., and Yuan, H.A. (1985) Anterior de- M., and Short, D. (2007). Guidelines for the conduct of clinical trials
compression of traumatic thoracolumbar fractures with incomplete for spinal cord injury as developed by the ICCP panel: spontaneous
neurological deficit using a retroperitoneal approach. J. Bone Joint recovery after spinal cord injury and statistical power needed for
Surg. Am. 67, 89–104. therapeutic clinical trials. Spinal Cord 45, 190–205.
41. McKinley, W., Meade, M.A., Kirshblum, S., and Barnard, B. (2004). 60. Kirshblum, S.C., Priebe, M.M., Ho, C.H., Scelza, W.M., Chiodo, A.E.,
Outcomes of early surgical management versus late or no surgical and Wuermser, L.A. (2007) Spinal cord injury medicine. 3. Re-
intervention after acute spinal cord injury. Arch. Phys. Med. Rehabil. habilitation phase after acute spinal cord injury. Arch. Phys. Med.
85, 1818–1825. Rehabil. 88, S62–70.
1794 VAN MIDDENDORP ET AL.

61. Grimes, D.A., and Schulz, K.F. (2002) Bias and causal associations in 67. van Middendorp, J.J., Goss, B., Urquhart, S., Atresh, S., Williams,
observational research. Lancet 359, 248–252. R.P., and Schuetz, M. (2011). Diagnosis and prognosis of traumatic
62. Groenwold, R.H., Hak, E., and Hoes, A.W. (2009) Quantitative as- spinal cord injury. Global Spine J 1, 001–008.
sessment of unobserved confounding is mandatory in nonrandomized 68. Akhtar, A.Z., Pippin, J.J., and Sandusky, C.B. (2008). Animal models
intervention studies. J. Clin. Epidemiol. 62, 22–28. in spinal cord injury: a review. Rev. Neurosci. 19, 47–60.
63. D’Agostino, R.B., Jr. (1998). Propensity score methods for bias re-
duction in the comparison of a treatment to a non-randomized control
group. Stat. Med. 17, 2265–2281. Address correspondence to:
64. Karri, V. (2006). Randomised clinical trials in plastic surgery: survey Joost J. van Middendorp, MD, PhD, MClinEpid
of output and quality of reporting. J. Plast. Reconstr. Aesthet. Surg. 59, National Spinal Injuries Centre
787–796. Stoke Mandeville Hospital
65. Blesch, A., and Tuszynski, M.H. (2009) Spinal cord injury: plasticity,
regeneration and the challenge of translational drug development. Buckinghamshire Healthcare NHS Trust
Trends Neurosci. 32, 41–47. Aylesbury, Buckinghamshire, HP21 8AL
66. Kirshblum, S.C., and O’Connor, K.C. (1998). Predicting neurologic United Kingdom
recovery in traumatic cervical spinal cord injury. Arch. Phys. Med.
Rehabil. 79, 1456–1466. E-mail: jvanmiddendorp@gmail.com

You might also like