Você está na página 1de 16

Brain Injury, May 2010; 24(5): 706–721

REVIEW

Acute management of acquired brain injury part II:


An evidence-based review of pharmacological interventions

MATTHEW J. MEYER1, JOSEPH MEGYESI2,3, JAY MEYTHALER4,5,


MANUEL MURIE-FERNANDEZ6, JO-ANNE AUBUT1, NORINE FOLEY1,
KATHERINE SALTER1, MARK BAYLEY7,8, SHAWN MARSHALL9,10, &
ROBERT TEASELL1,6,11
Brain Inj Downloaded from informahealthcare.com by HINARI on 03/21/11

1
Aging, Rehabilitation and Geriatric Care Program, Lawson Health Research Institute, London, Ontario, Canada,
2
Department of Clinical Neurological Sciences, University of Western Ontario, London, Ontario, Canada, 3London
Health Sciences Centre, University Hospital, London, Ontario, Canada, 4Department of Physical Medicine and
Rehabilitation, Wayne State University School of Medicine, Detroit, MI, USA, 5Rehabilitation Institute of Michigan,
Detroit, MI, USA, 6Department of Physical Medicine and Rehabilitation, St Joseph’s Health Care, London, Ontario,
Canda, 7Division of Physical Medicine and Rehabilitation, University of Toronto, Toronto, Ontario, Canada,
8
Toronto Rehabilitation Institute, Toronto, Ontario, Canada, 9The Ottawa Hospital Rehabilitation Centre, Ottawa,
Ontario, Canada, 10Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Cananda, and 11Department of
For personal use only.

Physical Medicine and Rehabilitation, Schulich School of Medicine and Dentistry, University of Western Ontario,
London, Ontario, Canada

(Received 22 July 2009; accepted 8 February 2010)

Abstract
Primary objective: To review the research literature on pharmacological interventions used in the acute phase of acquired
brain injury (ABI) to manage ICP and improve neural recovery.
Main outcomes: A literature search of multiple databases (CINAHL, EMBASE, MEDLINE and PSYCHINFO) and hand
searched articles covering the years 1980–2008 was performed. Peer reviewed articles were assessed for methodological
quality using the PEDro scoring system for randomized controlled trials (RCTs) and the Downs and Black tool for RCTs
and non-randomized trials. Levels of evidence were assigned and recommendations were made.
Results: In total, 11 pharmacological interventions used in the acute management of ABI were evaluated. These included
propofol, barbiturates, opioids, midazolam, mannitol, hypertonic saline, corticosteroids, progesterone, bradykinin
antagonists, dimethyl sulphoxide and cannabinoids. Of these interventions, corticosteroids were found to be contra-
indicated and cannabinoids were reported as ineffective. The other nine interventions demonstrated some benefit for
treatment of acute ABI. However, rarely did these benefits result in improved long-term patient outcomes.
Conclusions: Substantial research has been devoted to evaluating the use of pharmacological interventions in the acute
management of ABI. However, much of this research has focused on the application of individual interventions in small
single-site trials. Future research will need to establish larger patient samples to evaluate the benefits of combined
interventions within specific patient populations.

Keywords: Brain injury, acute, pharmacological management

Correspondence: Robert Teasell, Parkwood Hospital, 801 Commissioners Rd East, London, Ontario, Canada N6C 5J1. Tel: 519-685-4559. Fax: 519-685-
4023. E-mail: robert.teasell@sjhc.london.on.ca
ISSN 0269–9052 print/ISSN 1362–301X online ß 2010 Informa Healthcare Ltd.
DOI: 10.3109/02699051003692126
Acute ABI: Pharmacological 707

Introduction ABI [3]. Stated case definitions deemed to constitute


an ABI included TBI, head injury, brain injury,
Immediately following a serious brain injury, there is
anoxia, drug or alcohol induced toxicity and infec-
a variable degree of irreversible damage to the central
tion. Case definitions of cerebrovascular accident,
nervous system, commonly known as the primary
malignant/metastatic tumours, congenital or devel-
injury. Subsequently, a chain of events often leads to
opmental problems (e.g. cerebral palsy, autism) or
ongoing neural injury caused by oedema, inflamma-
progressive degenerative diseases (e.g. Alzheimer’s,
tion, hypoxia, ischemia, release of toxic amounts
Parkinson’s) were not included [4].
of excitatory neurotransmitters and impaired ionic
Relevant studies cited in review articles,
homeostasis across neuronal cell membranes [1].
meta-analyses, systematic reviews or in selected
Treatment of acute brain injury focuses on prevent-
study articles but not identified through the original
ing or minimizing the extent of secondary injury by
literature search were hand searched and included.
controlling intracranial hypertension, maintaining
Studies published before 1980 that held significant
adequate cerebral oxygenation and promoting ionic
clinical importance were also included.
homeostasis.
Studies were assessed for methodological quality
Pharmacological strategies for the management of
using the Physiotherapy Evidence Database
intracranial hypertension generally fall under one
(PEDro) [5] scoring system for randomized con-
of three types: osmotic agents (diuretics), analgesics
trolled trials (RCTs) and the Downs and Black [6]
Brain Inj Downloaded from informahealthcare.com by HINARI on 03/21/11

or sedatives. Osmotic agents draw fluid from the


tool for both RCTs and non-randomized trials. The
cranial cavity, thereby reducing increased intracra-
PEDro scale is comprised of 11 yes or no quality
nial pressure. Analgesics and sedatives reduce
items; 10 of which are used to calculate the final
the metabolic demands placed on injured or at risk
PEDro score (0–10). The Downs and Black scale
neurons by limiting nociceptive stimulation and
consists of 27 one-point questions and one two-point
reducing overall brain activity.
question (scored from 0–2) resulting in a final score
ranging from 0–28. For both tools, higher scores are
For personal use only.

indicative of greater methodological quality. Levels


Methods
of evidence were generated based on methodological
This review is the second in a series of three articles scores, which were then used to summarize the study
and is also a component of a larger review process. results [4]:
Detailed methods have been published separately
(1) Level 1 evidence: Findings supported by the
[2]. An exploratory search of multiple databases
results of one or more RCT of at least good
(CINAHL, EMBASE, MEDLINE and PsycINFO)
quality (PEDro score  6).
was performed to identify pharmacological interven-
(2) Level 2 evidence: Findings supported by a single
tions commonly used in acute ABI management.
RCT of at least fair quality (PEDro score < 6).
Interventions used to lower elevated intracranial
(3) Level 3 evidence: Findings supported by at least
pressure (ICP) and/or preserve neural functioning
one case-control study.
in the acute stages following an acquired brain injury
(4) Level 4 evidence: Findings supported by at least
(ABI) were considered. Interventions reported as
one:
standard practice or indicated as having potential
future benefits were selected for review. Extensive (a) Pre–post test: A prospective trail with a
literature reviews were then performed for each baseline measure and a post intervention
intervention individually using the same databases measure in a single group of subjects;
covering the years 1980–2008. (b) Post-test: A prospective study using a post
Title and keyword searches were performed using intervention measure only (no pre-test or
the name (or names) of the intervention in combi- baseline measurement with one or more
nation with ‘head injury’, ‘brain injury’ or ‘cranio- groups); or
cerebral injury’. Studies were considered for (c) Case series: A retrospective study, usually a
inclusion if at least 50% of the patient population chart audit.
consisted of patients with ABI, n  5, and the study
(5) Conflicting evidence: Findings in direct contra-
evaluated a targeted intervention with measurable
diction in at least two studies of similar meth-
outcomes.
odological quality.
For the purpose of this review, the Toronto
Acquired Brain Injury Network definition of ABI Interventions were evaluated individually. Each
was adopted [2]. This was chosen to reflect local care section contains a brief background regarding the
practices, to allow for inclusion of a large number of intervention, a table summarizing the details of each
pertinent trials and to coincide with definitions used study and a general overview. Bibliographical infor-
in reviews of other pharmacological interventions for mation, methodology and outcome information are
708 M. J. Meyer et al.

summarized in Tables I–XI. Levels of evidence and blocking agents, benzodiazepines, pentobarbital and
conclusions are presented in Table XII. CSF drainage when compared to patients receiving
morphine alone (Table XII). However, there were
no significant between-group differences reported in
Main outcomes and results mortality rates or GOS scores at 6-month follow-up.
The administration of morphine in conjunction with
Propofol propofol infusion made conclusions regarding pro-
Propofol is a powerful, fast-acting, short-duration pofol alone problematic. The other two studies also
sedative that provides potentially neuroprotective suggested propofol was a safe and effective sedative
effects through decreases in peripheral vascular agent but resulted in little or no effect on ICP or
tension. Several in vivo and in vitro animal studies CPP [9, 10]. Based on the results of this review,
have shown positive effects on cerebral physiology there was strong (level 1) evidence that propofol only
following an ABI including reductions in cerebral reduces elevated ICP and the need for other ICP and
blood flow, cerebral oxygen metabolism, EEG sedative interventions when used in conjunction with
activity and ICP [7]. One RCT and two morphine (Table XII).
non-RCTs examining propofol use in patients with Anecdotally, it has been reported that administra-
brain injuries were evaluated (Table I). tion of high dose propofol can result in propofol
Brain Inj Downloaded from informahealthcare.com by HINARI on 03/21/11

In the only RCT identified, Kelly et al. [8] infusion syndrome, characterized by severe meta-
compared propofol sedation to morphine for safety bolic acidosis, rhabdomyolosis, cardiac dysrhythmias
and efficacy. Propofol, in combination with mor- and potential cardiovascular collapse [11]; espe-
phine, reduced ICP and the need for neuromuscular cially in children [12]. In a letter to the Lancet,

Table I. Propofol for the acute management of ABI.


For personal use only.

Study n Study design PEDro D&B Interventions Results

[8] 42 RCT 8 25 Propofol & morphine vs. morphine (þ) ICP control
(ND) GOS (6M)
[10] 15 Non-RCT NS 10 Propofol vs. (þ) AVDO2
morphine/midazolam (ND) MABP, ICP, CPP, GOS (6M)
[9] 10 Case series NS 10 Propofol (þ) ICP, CPP

PEDro ¼ Physiotherapy Evidence Database rating scale score [5]; D&B ¼ Downs and Black [6] quality assessment scale score;
(þ), beneficial result; (ND), no difference; (), negative result; (NS), no score; ICP, intracranial pressure; GOS, Glasgow outcome scale;
AVDO2, arterio-venous difference oxygen; MABP, mean arterial blood pressure; CPP, cerebral perfusion pressure.

Table II. Barbiturates for the acute management of ABI.

Study n Study design PEDro D&B Interventions Results

[23] 53 RCT 6 18 Pentobarbitol vs. control (ND) Mortality, ICP, GOS


() MABP
[25] 20 RCT 5 20 Thiopental vs. pentobarbital (ND) ICP, Mortality
[24] 59 RCT 5 17 Pentobarbitol vs. mannitol () ICP, Mortality (patients with no haematoma)
[22] 73 RCT 4 18 Pentobarbitol vs. control (þ) Mortality (if ICP responded to barbiturate)
[100] 7 Non-RCT NS 17 Pentobarbitol vs. control (þ) Energy expenditure, 24 h nitrogen excretion
(ND) 3-MH excretion
[20] 40 Non-RCT NS 16 Barbiturate coma vs. control () adrenal insufficiency
[21]
52 Non-RCT NS 15 Thiopental vs. control () reversible leucopenia, granulocytopenia,
infection rate, bone marrow function
[26] 38 Non-RCT NS 15 Thiopental & hypothermia vs. (þ) GOS
control
[101] 12 Case series NS 13 Barbiturate coma (ND) ICP, MABP, CPP, PtiO2, PRx
[102] 38 Case series NS 11 Thiopentone (þ) ICP
() MAPB

PEDro ¼ Physiotherapy Evidence Database rating scale score [5]; D&B ¼ Downs and Black [6] quality assessment scale score;
(þ), beneficial result; (ND), no difference; (), negative result; (NS), no score; ICP, intracranial pressure; GOS, Glasgow Outcome Scale;
MABP, mean arterial blood pressure; 3-MH, 3-methylhistidine; CPP, cerebral perfusion pressure; PTiO2, brain tissue oxygenation;
PRx, autoregulation.
Acute ABI: Pharmacological 709

Table III. Opioids for the acute management of ABI.

Study n Study design PEDro D&B Interventions Results

[36] 30 RCT 8 23 Morphine vs. fentanyl (ND) ICP, MABP, CPP


[35] 9 RCT 7 19 Fentanyl vs. sufentanil (ND) ICP, MABP
[40] 161 RCT 5 19 Remifentanil & propofol/midazolam if (þ) awakening for assessment
necessary vs. propofol/midazolam &
fentanyl/morphine if necessary
[38] 15 RCT 5 19 Fentanyl vs. sufentanil vs. morphine (ND) ICP
() MAP (suf vs. fen & mor)
[33] 6 Case series NS 15 Sufentanil, alfentanil or fentanyl () ICP, MABP, CPP
[39] 10 Case series NS 15 Sufentanil & sufentanil/midazolam (þ) ICP
() MABP
[34] 10 Case series NS 15 Sufentanil () ICP, MABP, CPP
[37] 20 Case series NS 14 Remifentanil (ND) ICP, CPP, CBFV
[30] 30 Case series NS 14 Sufentanil () MAP, ICP

PEDro ¼ Physiotherapy Evidence Database rating scale score [5]; D&B ¼ Downs and Black [6] quality assessment scale score;
(þ), beneficial result; (ND), no difference; (), negative result; (NS), no score; ICP; intracranial pressure; MABP, mean arterial blood
pressure; CPP, cerebral perfusion pressure; CBFV, cerebral blood flow velocity.
Brain Inj Downloaded from informahealthcare.com by HINARI on 03/21/11

Table IV. Midazolam for the acute management of ABI.

Study n Study design PEDro D&B Interventions Results

[44] 59 RCT 5 17 Midazolam vs. propofol vs. (þ) Hypertrigliceridemia (mid vs. prop)
midazolam & propofol (ND) haemodynamics, ICP, CPP, SjvO2
For personal use only.

[43] 184 Chart review NS NS Midazolam () hypotension (in greater doses)
[45] 12 Case series NS 12 Midazolam (ND) ICP
() CPP, MABP

PEDro ¼ Physiotherapy Evidence Database rating scale score [5]; D&B ¼ Downs and Black [6] quality assessment scale score;
(þ), beneficial result; (ND), no difference; (), negative result; (NS), no score; ICP, intracranial pressure; CPP, cerebral perfusion
pressure; SjvO2, jugular vein O2 saturation; MABP, mean arterial blood pressure.

Table V. Mannitol for the acute management of ABI.

Study n Study design PEDro D&B Interventions Results

[60] 41 RCT 7 20 Mannitol vs. saline () Urine output


(þ) Serum sodium
(ND) BP
[57] 20 RCT 7 22 Mannitol vs. hypertonic saline () ICP clinical failure
(ND) Mortality, GOS
[59] 20 RCT 6 22 Mannitol vs. Hypertonic saline (ND) ICP
[47] 9 RCT cross-over 5 18 Mannitol vs. saline & dextran-70 () ICP
[50] 44 RCT 5 18 High dose mannitol vs. conventional-dose (þ) Papillary widening, Mortality,
mannitol GOS (6M)
[52] 141 RCT 5 18 High dose mannitol vs. conventional-dose (þ) Papillary widening, Mortality,
mannitol GOS (6M)
[51] 178 RCT 4 17 High dose mannitol vs. conventional-dose (þ) Papillary widening, Mortality,
mannitol GOS (6M)
[55] 80 RCT 4 16 Mannitol (only if ICP > 25 mmHg) vs. (ND) Mortality, ICP, neurological
mannitol outcome
[58] 13 Case series NS 10 Mannitol vs. hypertonic saline () Duration of ICP reduction
(ND) ICP
[53] 28 Case series NS 12 Mannitol 0.1 g Kg1 (100 kg patient ¼ 1.0 mmHG
ICP drop
[56] 11 Case series NS 9 Mannitol (þ) ICP, CPP (if ICP > 20 mmHg)
(ND) ICP (if < 20 mmHg)

PEDro ¼ Physiotherapy Evidence Database rating scale score [5]; D&B ¼ Downs and Black [6] quality assessment scale score;
(þ), beneficial result; (ND), no difference; (), negative result; (NS), no score; BP, blood pressure; ICP, intracranial pressure;
GOS, Glasgow outcome scale; CPP, cerebral perfusion pressure.
710 M. J. Meyer et al.

Table VI. Hypertonic saline for the acute management of ABI.

Study n Study design PEDro D&B Interventions Results

[65] 460 RCT 10 20 Saline vs. albumin (þ) Mortality


(ND) GOS-E
[66] 229 RCT 9 23 Hypertonic saline vs. Ringer’s lactate (ND) Mortality, GOS
[57] 20 RCT 7 22 Hypertonic saline vs. mannitol (þ) ICP clinical failure
(ND) Mortality, GOS
[67] 32 RCT 6 21 Hypertonic saline vs. Ringer’s lactate (þ) ARDS, LOS, complications
[47] 9 RCT 5 18 Saline & dextran-70 vs. mannitol (þ) ICP
[64] 41 RCT 5 19 Hypertonic saline vs. Ringer’s lactate (ND) ICP, GOS
() # treatments
[62] 10 Case series NS 15 Saline (þ) ICP, CPP
[68] 14 Case series NS 12 Hypertonic saline (þ) ICP, non-contused volume
() contused hemispheric volume
[69] 10 Case series NS 12 Hypertonic saline/acetate solution (þ) ICP, lateral displacement, GCS
[61] 10 Case series NS 11 Hypertonic saline. (þ) ICP, CPP
[58] 13 Case series NS 10 Hypertonic saline vs. mannitol (þ) Duration of ICP reduction
(ND) ICP
[70] 6 Case series NS 10 Hypertonic saline (þ) ICP
Brain Inj Downloaded from informahealthcare.com by HINARI on 03/21/11

[71] 12 Case series NS 6 Hypertonic saline (þ) ICP, CPP, Pbt O2.

PEDro ¼ Physiotherapy Evidence Database rating scale score [5]; D&B ¼ Downs and Black [6] quality assessment scale score;
(þ), beneficial result; (ND), no difference; (), negative result; (NS), no score; GOS[-E], Glasgow outcome scale [extended];
ICP, intracranial pressure; ARDS, acute respiratory distress syndrome; LOS, length of stay; CPP, cerebral perfusion pressure; GCS,
Glasgow coma scale; Pbt O2, partial pressure of brain tissue O2.
For personal use only.

Table VII. Corticosteroids for the acute management of ABI.

Study n Study design PEDro D&B Interventions Results

[75] 10 008 RCT 10 27 Methylprednisolone vs. control () Mortality


[73] 396 RCT 9 19 Triamcinolone acetonide vs. placebo (þ) GOS (if GCS < 8)
[74] 76 RCT 8 19 High dose dexamethasone vs. low dose (ND) ICP, good outcomes
dexamethasone vs. placebo
[77] 88 RCT 7 20 High-dose methylprednisolone vs. low dose (ND) GOS, morbidity
methylprednisolone vs. placebo
[78] 130 RCT 4 19 Dexamethasone vs. placebo (þ) ICP (if >20 mmHg)
[79] 161 RCT 4 20 High-dose dexamethasone vs. placebo (ND) Mortality, GOS (6 Mo)
[76] 100 RCT 4 16 Methylprednisolone vs. control (ND) GOS
[80] 76 RCT 4 15 High-dose dexamethasone vs. low-dose (ND) ICP
dexamethasone vs. placebo
[81] 404 Non-RCT NS 15 Glucocorticoids vs. control () Early seizures (<2 days)

PEDro ¼ Physiotherapy Evidence Database rating scale score [5]; D&B ¼ Downs and Black [6] quality assessment scale score;
(þ), beneficial result; (ND), no difference; (), negative result; (NS), no score; GOS, Glasgow outcome scale; ICP, intracranial pressure.

Table VIII. Progesterone for the acute management of ABI.

Study n Study design PEDro D&B Interventions Results

[84] 100 RCT 10 26 IV progesterone vs. placebo (þ) Mortality (30-day), GOS-E (if GCS 9–18)
[85] 159 RCT 7 23 IV progesterone vs. placebo (þ) Mortality, GOS, mFIM,
(ND) ICP

PEDro ¼ Physiotherapy Evidence Database rating scale score [5]; D&B ¼ Downs and Black [6] quality assessment scale score;
(þ), beneficial result; (ND), no difference; (), negative result; (NS), no score; GOS-E, Glasgow outcome scale extended; GCS, Glasgow
coma scale; GOS, Glasgow outcome scale; mFIM, modified Functional Independence Measure; ICP, intracranial pressure.

Cremer et al. [13] assessed all patients treated in their they identified a crude odds ratio of 1.93 (95% CI
facility from 1996–1999 retrospectively and identi- 1.12–3.32, p ¼ 0.018) for developing propofol infu-
fied seven patients who died of apparent propofol sion syndrome per unit (mg kg1 h1) increase in
infusion syndrome. Using logistic regression analysis, propofol dose. Similarly, Otterspoor et al. [14]
Acute ABI: Pharmacological 711

Table IX. Bradykinin antagonist for the acute management of ABI.

Study n Study design PEDro D&B Interventions Results

[86] 139 RCT 8 22 BradycorTM vs. placebo (þ) ICP (days 4 & 5), mortality
(ND) GOS
[87] 20 RCT 6 21 BradycorTM vs. Ringer’s lactate (þ) ICP, GCS
[90] 25 RCT 4 17 Anatibant vs. placebo (þ) GOS (3, 6 Mo)

PEDro ¼ Physiotherapy Evidence Database rating scale score [5]; D&B ¼ Downs and Black [6] quality assessment scale score;
(þ), beneficial result; (ND), no difference; (), negative result; (NS), no score; ICP, intracranial pressure; GOS, Glasgow outcome scale;
GCS, Glasgow coma scale.

Table X. DMSO for the acute management of ABI.

Study n Study design PEDro D&B Interventions Results

[92] 10 Case series NS 10 DMSO (þ) ICP (30 min)


[91] 10 Case series NS 8 DMSO (if ICP  25 mmHg) (þ) ICP, CPP (short term)
Brain Inj Downloaded from informahealthcare.com by HINARI on 03/21/11

[93] 5 Case studies NS NS DMSO (þ) ICP (15 min)


() hypernaetremia

PEDro ¼ Physiotherapy Evidence Database rating scale score [5]; D&B ¼ Downs and Black [6] quality assessment scale score;
(þ), beneficial result; (ND), no difference; (), negative result; (NS), no score; ICP, intracranial pressure; CPP, cerebral perfusion
pressure.

Table XI. Cannabinoids for the acute management of ABI.


For personal use only.

Study n Study design PEDro D&B Interventions Results

[99] 861 RCT 10 25 Dexanabinol vs. placebo (ND) Mortality, GOS, ICP, CPP, BI, SF36, CIQ
[98] 67 RCT 8 22 Dexanabinol vs. placebo (þ) ICP, CPP, GOS (1 Mo), DRS

PEDro ¼ Physiotherapy Evidence Database rating scale score [5]; D&B ¼ Downs and Black [6] quality assessment scale score;
(þ), beneficial result; (ND), no difference; (), negative result; (NS), no score; ICP, intracranial pressure; CPP, cerebral perfusion
pressure; DRS, disability rating scale; BI, Barthel index; SF36, short form 36; CIQ, community reintegration questionnaire.

reviewed case reports to assess risk factors associated in patients with ABI [17, 18] and suggested
with propofol infusion syndrome. They identified barbiturates reduced elevated ICP, even in patients
large cumulative doses, young age, acute neurological unresponsive to other ICP management interven-
injury, low carbohydrate intake, high fat intake, tions (mannitol and hyperventilation) [19].
catecholamine infusion, corticosteroid infusion, crit- However, most early investigations were uncon-
ical illness and inborn errors of mitochondrial fatty trolled comparisons of small patient cohorts.
acid oxidation as potential risk factors. They recom- More recent studies have suggested barbiturate-
mended that until further research is conducted, induced coma may result in adverse effects including
propofol infusion should not exceed 4 mg kg1 h1 in adrenal insufficiency [20] and bone marrow sup-
patients with any of the above risk factors and that pression [21]. Four RCTs and six non-RCTs
infusion of 5 mg kg1 h1 should never be exceeded. evaluating barbiturate use in ABI were found
(Table II).
Conflicting evidence has been reported regarding
Barbiturates
many aspects of barbiturate use in patients with ABI.
Barbiturates have long been used in the management Eisenberg et al. [22] reported high dose pentobarbi-
of elevated ICP. It is believed that barbiturates tal was an effective adjunctive therapy for the
reduce increased ICP through the suppression management of elevated ICP refractory to conven-
of cerebral metabolism, thereby reducing meta- tional therapeutic measures and supported the use
bolic demands and cerebral blood volume [15]. of high dose barbiturates for a small sub-group of
Barbiturates are also utilized to prevent the devel- severe ABI patients (GCS  7). In contrast, Ward
opment of seizures in acute ABI, despite conflicting et al. [23] reported pentobarbital was no better
claims regarding their efficacy [16]. Early reports than conventional ICP management measures
supported the therapeutic benefit of barbiturate use in a similar group of patients (GCS  7), while
712 M. J. Meyer et al.

Table XII. Recommendations regarding pharmacological interventions for acute ABI management.

Treatment Conclusion Level of Evidence

Propofol  Propofol reduces ICP and the need for other ICP and sedative interventions when 1
used in conjunction with morphine
Barbiturates  There is no difference between thiopental and pentobarbital for control of elevated 1
ICP
 Pentobarbital is less effective than mannitol for control of elevated ICP 2
 Barbiturate therapy may cause reversible leukopenia, granulocytopenia and 3
systemic hypotension
 A combination barbiturate therapy and hypothermia may result in improved 3
clinical outcomes up to 1 year post-injury
 Pentobarbital is more effective than conventional ICP management measures Conflicting
Opioids  Bolus opioid administration results in elevated ICP 1
 Remifentanil results in faster arousal compared to hypnotic-based sedation 2
regimens
 Infusion of opioids results in elevations in ICP Conflicting
Midazolam  Midazolam has no effect on ICP in brain injured patients 2
 Midazolam negatively affects CPP and MAP Conflicting
Mannitol  Higher dose mannitol is superior to conventional mannitol in improving mortality 1
Brain Inj Downloaded from informahealthcare.com by HINARI on 03/21/11

rates and clinical outcomes


 Mannitol is less effective than hypertonic saline for reducing ICP 1
 Out-of-hospital administration of mannitol does not adversely affect blood pressure 1
compared to treatment with saline
 Prophylactic mannitol does not result in decreased ICP Conflicting
Hypertonic saline  Hypertonic saline improves ICP and CPP in brain injured patients 1
 Hypertonic saline reduces ICP more effectively than mannitol 1
 Hypertonic saline results in similar improvements in ICP and GOS outcomes 1
compared with Ringer’s lactate solution
For personal use only.

 Hypertonic saline solution results in improved mortality rates but not GOS scores 1
compared to albumin
 Hypertonic saline may increase the volume of contused brain tissue 4
Corticosteroids  Methylprednisolone increases mortality rates in ABI patients and should not be 1
used
 Triamcinolone may improve outcomes in patients with a GCS < 8 and a focal 1
lesion
 Glucocorticoid administration may increase the risk of developing first late seizures 3
 Corticosteroids reduce ICP Conflicting
Progesterone  Progesterone decreases mortality rates in ABI patients and improves clinical 1
outcomes up to 6 months post-treatment
Bradykinin  Bradycor is effective in preventing acute elevations in ICP post-ABI 1
Antagonists  Bradykinin antagonists improve functional clinical outcomes Conflicting
Dimethyl sulphoxide  Dimethyl sulphoxide transiently reduces ICP elevations 4
Cannabinoids  Dexanabinol does not provide acute improvement in ICP or long-term clinical 1
benefits post-ABI

Schwartz et al. [24] suggested pentobarbital is less separate meta-analysis, Roberts [15] reported
effective than mannitol in the treatment of elevated that, while barbiturates may reduce elevated ICP,
ICP. Perez-Barcena et al. [25] reported thiopental there was little evidence to link this with reductions
may be superior to pentobarbital in controlling in mortality or disability. In the same meta-analysis,
ICP; however, Llompart-Pou et al. [20] reported Roberts [15] noted that barbiturate therapy was
thiopental led to increased risk of developing adrenal associated with substantial hypotension that could
insufficiency compared to control. offset any ICP-lowering effect. Similarly, this
Nordby and Nesbakken [26] reported thiopental review located several studies linking barbiturate
combined with mild hypothermia resulted in better use to adverse effects, including decreased cerebral
clinical outcomes on the Glasgow Outcome scale 1 perfusion pressure due to decreased arterial pres-
year post-injury when compared with conventional sure [27] and reduced production of white blood
ICP management measures (including hyperventila- cells [21]. Stover and Sticker [21] noted interactions
tion, steroids and mannitol). However, because with bone marrow suppressing antibiotics (specifi-
this study used a combination of thiopental and cally tazobactum/piperacillin) can further exacer-
hypothermia, it was not possible to attribute bated the suppression of white blood cell
better clinical outcomes to thiopental alone. In a production.
Acute ABI: Pharmacological 713

In summary, there was conflicting evidence as to primary sedative agent and then a hypnotic agent,
whether barbiturates are more effective than other while patients in the control groups received fentanyl
conventional ICP management strategies and strong or morphine in conjunction with a hypnotic agent.
(level 1) evidence of no difference between thiopen- Therefore, remifentanil’s efficacy can be compared
tal and pentobarbital for ICP control. There was to hypnotic-based sedation but not fentanyl or
level 2 evidence that mannitol is superior to morphine. There was level 2 evidence that remifen-
pentobarbital for ICP control and level 3 evidence tanil results in faster arousal compared to hypnotic
that barbiturates may cause reversible leukopenia, based sedation (Table XII).
granulocytopenia and systemic hyotension. There
was also level 3 evidence that barbiturate use in Midazolam
conjunction with hypothermia improves long-term
Midazolam is a fast-acting benzodiazepine with a
patient outcomes (Table XII).
short half-life and inactive metabolites [41]. It is
anxiolytic and displays anti-epileptic, sedative, and
Opioids amnestic properties. It is a protein-bound, highly
Opioids are substances that have morphine-like lipid soluble drug which crosses the blood–brain
actions. They work by binding to opioid receptors, barrier and has a rapid onset of action within
found principally in the central nervous system and 1–5 minutes in most patients [42]. However, delayed
Brain Inj Downloaded from informahealthcare.com by HINARI on 03/21/11

the gastrointestinal tract. Each opioid has a distinct elimination of midazolam, resulting in prolonged
binding affinity to group(s) of opioid receptors that sedation, has been demonstrated in some critically
then determines its pharmacodynamic response. ill patients.
Morphine has been the most commonly used Studies in the operating room or intensive care
opioid following an ABI, while fentanyl and its unit have demonstrated midazolam to be relatively
derivatives have gained popularity owing to their safe in euvolemic patients or in the presence of
more rapid onset and shorter duration of effect [28]. continuous haemodynamic monitoring for early
For personal use only.

Controversy persists regarding the effect of opioids detection of hypotension [43]. Midazolam has been
on ICP and CPP. It has been reported that opioids found to reduce cerebrospinal fluid pressure in
can increase cerebral blood flow (CBF), which may patients without intracranial mass lesions as well as
lead to an increase in ICP [29–32] in the presence decrease cerebral blood flow and cerebral oxygen
of intracranial pathology. This review found four consumption [42]. One RCT and two non-RCTs on
RCTs and five non-RCTs evaluating opioid use in midazolam use in acute ABI management were
acute ABI management (Table III). reviewed (Table IV).
Analgesic sedation with opioids is commonly used Infusions of midazolam or propofol were reported
in conjunction with hypnotic agents (i.e. midazolam, to provide similar quality sedation in patients with
propofol) to reduce nociceptive stimulation. This severe head trauma, although propofol was asso-
makes it difficult to evaluate the effects of opioids ciated with a high incidence of hypertriglyceridemia
in isolation. Five studies reported increases in ICP [44]. In both studies evaluating midazolam and ICP,
after opioid administration [30, 33–36], while two no significant difference was seen after midazolam
found no increase [37, 38] and one reported a administration [44, 45]. However, significant hypo-
tension related to increased doses of midazolam [43]
decrease [39]. However, mode of administration has
and decreases in MAP resulting in decreased CPP
been suggested as a determining factor for increases
(especially in patients with initial ICP  18 mmHg)
in ICP [33, 34]. In those studies where patients
[45] were also reported. In summary, there was level
received only bolus injections of opioids, significant
2 evidence that midazolam has no effect on ICP, but
increases in ICP were seen [30, 35, 36]. There was
conflicting evidence regarding midazolam’s effect on
strong (level 1) evidence that bolus opioid adminis-
MAP and CPP (Table XII). Hypotension should be
tration resulted in increased ICP and conflicting
monitored as a potential side-effect based on current
evidence regarding the effects of opioid infusion on
research.
ICP levels (Table XII).
Fentanyl and its derivatives have been suggested
Mannitol
as more ideally suited for sedation in patients with
brain injury due to their rapid onset and short Mannitol is an osmotic agent that draws excess fluid
duration [28]. In this review, one study found from the cranial cavity, thereby decreasing ICP.
remifentanil resulted in significantly faster arousal However, with prolonged dosage, mannitol may
compared to propofol or midazolam [40]. The penetrate the blood–brain barrier, potentially exacer-
authors suggested that this allowed for prompt bating any elevation in ICP [46]. Mannitol has also
neurological assessment. However, patients in been associated with significant diuresis, acute renal
the treatment group received remifentanyl as the failure, hyperkalemia, hypotension and rebound
714 M. J. Meyer et al.

increments in ICP [47, 48]. For these reasons, it has pentobarbital but detrimental effects when com-
been recommended that mannitol only be used pared to hypertonic saline [46]. The authors report
when signs of elevated ICP or deteriorating neuro- consensus agreement that mannitol is effective
logical status suggest the benefits of mannitol may in reducing ICP, but has potentially harmful side-
outweigh potential complications or adverse effects effects that need to be taken into consideration.
[49]. Eight RCTs and three non-RCTs assessing the In conclusion, this review resulted in strong
efficacy of mannitol in brain injury management (level 1) evidence that mannitol reduces elevated
were reviewed (Table V). ICP, but equally strong evidence that mannitol is
All of the studies reviewed reported that mannitol less effective than hypertonic saline for ICP reduc-
reduced ICP. However, there remains considerable tion (Table XII). Given mannitol’s effectiveness in
uncertainty regarding how and when it should be reducing elevated ICP, further research into specific
used. Cruz et al. [50–52] conducted three separate indications for its use relative to other osmotic agents
RCTs in ABI patients to investigate the effects of would be beneficial.
high dose mannitol on clinical outcomes 6 months
post-injury. All three trials indicated that high dose Hypertonic saline
mannitol (1.4 g kg1) was superior to conventional
mannitol therapy (0.7 g kg1) in improving mortality Hypertonic saline is an osmotic agent that has
rates and clinical outcomes. This finding was traditionally been used as an adjunct to mannitol
Brain Inj Downloaded from informahealthcare.com by HINARI on 03/21/11

further supported by Sorani et al. [53] who found or in individuals who have become tolerant to
a 1.0 mmHg drop in ICP for every 0.1 g Kg1 mannitol. However, recent studies have examined
increase in mannitol dosage. However, the hypertonic saline as a primary measure for ICP
American Association of Neurological Surgeons control [58, 61]. Hypertonic saline exerts its effect
(AANS) has recommended mannitol for ICP control primarily by increasing serum sodium and osmolar-
at 0.25–1 g Kg1 body weight, but that systolic BP ity, thereby establishing an osmotic gradient. Water
should not be allowed to drop below 90 mmHg [54]. diffuses passively from cerebral intracellular and
For personal use only.

While improved outcomes may be obtained with interstitial space into capillaries resulting in a reduc-
higher doses, decisions regarding mannitol adminis- tion in ICP [62]. Although mannitol works similarly,
tration should be made on a case-by-case basis. sodium chloride has a better reflection coefficient
Three studies addressed the effects of timing and (1.0) than mannitol (0.9) [63], making it a better
ICP level on mannitol administration. In the only osmotic agent. Hypertonic saline may also normalize
RCT evaluating timing of administration, Smith resting membrane potential and cell volume by
et al. [55] reported no significant difference in terms restoring normal intracellular electrolyte balance in
of mortality rates or neurological outcome between injured cells [62]. Six RCTs and seven non-RCTs
patients receiving mannitol irrespective of ICP mea- examining the use of hypertonic saline were reviewed
surement and those who received mannitol only after (Table VI).
the onset of intracranial hypertension (>25 mmHg). Based on the above studies, hypertonic saline
However, the lack of a non-treatment control group proved beneficial in many aspects of acute ABI
makes it unclear whether or not prophylactic management. There was strong (level 1) evidence
mannitol was effective. Hartl et al. [56] reported hypertonic saline is effective in reducing ICP as all
that mannitol was only effective in diminishing ICP of the above trials demonstrated decreases in ICP
when initial ICP was elevated (>20 mmHg); this associated with hypertonic saline administration [47,
was corroborated by Sorani et al. [53]. Therefore, 57, 58, 61, 62, 64–71]. Three studies also reported
there appears to be conflicting evidence regarding improvements in CPP [61, 62, 71] and one reported
the benefit of prophylactic mannitol administration. improvements in cerebral oxygenation [71].
Five studies and a review were located that Compared to mannitol, hypertonic saline was
assessed the efficacy of mannitol therapy relative to shown to induce a significantly greater decrease
other osmotic agents. Three studies reported man- in ICP over time [47], with fewer incidences of
nitol resulted in poorer reductions in ICP when persistent ICP [57]. Hypertonic saline was also
compared to hypertonic saline [47, 57, 58]. shown to decrease mortality rates in severely injured
Francony et al. [59] found mannitol and saline patients (GCS 3–8) compared with albumin infu-
were comparable in reducing ICP in stable patients sions [65] and resulted in similar reductions in
with intact autoregulation and Sayre et al. [60] ICP when compared to Ringer’s lactate solution
reported mannitol administration did not signifi- [64, 66].
cantly affect blood pressure relative to saline when Despite these positive findings, some limitations
administered out of hospital. However, a 2008 to hypertonic saline use were also noted. Shackford
Cochrane review concluded mannitol may have et al. [64] reported that more medical interventions
beneficial effects on mortality when compared to were required to reduce ICP with hypertonic saline
Acute ABI: Pharmacological 715

compared to Ringer’s lactate, although a lack of steroid group compared to controls (1.18, 95% CI
baseline comparability between groups may have 1.09–1.27; p ¼ 0.0001). Further analysis showed
biased the results. Lescot et al. [68] applied CT no differences in outcomes between eight CT
technology to assess the effectiveness of hypertonic sub-groups or between patients with major extra-
saline on volume, weight and specific gravity of cranial injury compared to those without. The
contused and non-contused brain tissue. Three days authors also conducted a systematic review and
after TBI, the blood–brain barrier remained meta-analysis of existing trials using corticosteroids
semi-permeable in non-contused areas but not in for head injury. Before the CRASH trial, a 0.96
contused areas. Contused tissue increased in volume (95% CI 0.85–1.08) relative risk of death was
following administration of hypertonic saline. The reported in the corticosteroid group. The addition
authors recommended further study was needed to of the CRASH trial increased the relative risk to
assess the effects of hypertonic saline on different 1.12 (95% CI 1.05–1.2). The authors concluded
tissue types so contusion site and size could be that corticosteroids should not be used in head
factored into clinical decisions. injury care. The meta-analysis performed by Roberts
This review resulted in the following conclusions et al. [75] included all but two of the studies in this
regarding hypertonic saline use in acute ABI man- review.
agement. There was level 1 evidence that hypertonic Of the studies in this review, two assessed methyl-
Brain Inj Downloaded from informahealthcare.com by HINARI on 03/21/11

saline improves ICP and CPP in patients with prednisolone use and both reported no improve-
acute brain injury, reduces ICP more effectively ments in GOS outcome at 6 months [76, 77].
than mannitol, results in similar reductions in ICP However, Giannotta et al. [77] reported patients less
and GOS outcomes compared with Ringer’s lactate than 40 years old demonstrated decreased mortality
solution and decreases mortality rates but not GOS rates. Four studies evaluated dexamethasone treat-
scores compared to albumin. There was level 4 ment [74, 78–80], none of which reported significant
evidence that hypertonic saline may increase the improvements compared with placebo.
volume of contused brain tissue (Table XII). Watson et al. [81] retrospectively evaluated
For personal use only.

the influence of four different corticosteroids on


Corticosteroids post-traumatic seizure rates. They noted patients
receiving glucocorticoid treatment on the first day
Corticosteroids used in brain injury care have post-injury were at increased risk of developing a
included dexamethasone, methylprednisolone, pre- first late seizure when compared to patients receiving
dnisolone, betamethasone, cortisone, hydrocorti- no treatment. They also saw no improvement in
sone, prednisone and triamcinolone [72]. The use patients receiving glucocorticoids after the first day.
of such a broad spectrum of agents within diverse In the only study reporting positive results,
patient groups has complicated one’s understanding Grumme et al. [73] found the synthetic corticoster-
of corticosteroid efficacy in acute ABI. This problem oid triamcinolone improved outcomes in patients
has been exacerbated by a lack of understanding of with both a GCS < 8 and a focal lesion.
the mode of steroid action in this setting. Laboratory In summary, there was level 1 evidence that
studies have reported reductions in wet brain weight, methylprednisolone increases mortality and should
facilitation of synaptic transmission, reduction of not be used in acute ABI management and there was
lipid peroxidation, enhanced blood flow, preserva- conflicting evidence regarding the effect of corticos-
tion of electrolyte distribution and membrane stabi- teroids on ICP. There was level 1 evidence based on
lization with corticosteroid use [73]. Research one study that triamcinolone may improve outcomes
has noted focal lesions appear to respond well to in patients with a GCS < 8 and a focal lesion,
corticosteroid therapy while diffuse intracerebral but further study should be carried out cautiously
lesions and haematomas appear less responsive [73, given the results of a larger trial. There was level 3
74]. Nine studies (eight RCTS and one non-RCT) evidence that glucocorticoid administration may
on corticosteroid use in acute ABI were reviewed increase the risk of developing first late seizures
(Table VII). (Table XII).
In what has become the definitive study of
corticosteroid use to date, Roberts et al. [75]
Progesterone
conducted a large, multinational, randomized con-
trolled collaborative study of early methylpredniso- Progesterone has been identified as a potential
lone use after severe head injury. This study, neuro-protective agent. Animal research suggests
the Corticosteroid Randomization After Severe progesterone modulates excito-toxicity, reconstitutes
Head Injury (CRASH) trial, was halted after the blood–brain barrier, reduces cerebral oedema,
10 008 patients had been randomly allocated as the regulates inflammation and decreases apoptosis
reported relative risk of death was greater in the [82]. In some cases, women have been excluded
716 M. J. Meyer et al.

from studies due to concerns that hormonal inter- need for other therapeutic interventions to control
actions will improve their outcomes and bias study ICP was markedly lower in those treated with
results [83]. However, few clinical studies have BradycorTM [87]. A larger RCT conducted by
shown this to be true, while several epidemiological Marmarou et al. [86] reported that, compared with
studies have indicated that female patients may those assigned to a placebo group, patients treated
actually do worse [83]. Only two published clinical with BradycorTM experienced a significant reduction
studies of progesterone use in ABI have been in intracranial hypertension (ICP > 15 mmHg).
performed to date (Table VIII). However, despite trends favouring the BradycorTM
In a phase II trial, Wright et al. [84] assessed treatment, there were no significant differences
progesterone vs. placebo for patients acutely follow- in mortality rates, improvements in GOS scores at
ing ABI. Patients in the progesterone group demon- 3 and 6 months or the intensity of therapeutic
strated no increase in complication rates and a interventions needed to control ICP [86].
decreased 30-day mortality rate. In addition, utiliz- In a more recent study, severe head injury patients
ing a post-hoc analysis, patients with moderate– were randomized to receive placebo or Anatibant,
severe brain injury showed significantly greater a more potent bradykinin antagonist than
rates of moderate-to-good GOS-E scores. Despite BradycorTM [90]. The authors were unable to
limitations with sample size, the authors noted that report any conclusive evidence to support or refute
Brain Inj Downloaded from informahealthcare.com by HINARI on 03/21/11

these results were encouraging and warranted inves- the efficacy of Anatibant in preventing brain oedema
tigation in a larger and more thorough clinical trial. or deteriorations in ICP and CPP, which they
Xiao et al. [85] provided intravenous progesterone attributed to a small sample size and lack of baseline
to TBI patients with a GCS < 9. Despite no change comparability between groups [90]. However, the
in ICP compared to controls, significant improve- authors did report that those patients who received
ments in Glasgow Outcome Scale, mortality and the higher dose of this drug showed more favourable
modified FIM scores were reported at 3 and 6 outcomes on the GOS at 3 and 6 months compared
month follow-up. Based on these trials, there was with the other groups [90].
For personal use only.

level 1 evidence that progesterone can decrease In summary, there was level 1 evidence that
mortality rates in ABI patients and improve clinical Bradycor is effective in preventing acute elevations
outcomes (Table XII). Given these beneficial in ICP post-ABI but conflicting evidence that
results and the lack of complications associated bradykinin antagonists improve functional clinical
with progesterone administration, further study is outcomes such as the GOS (Table XII).
warranted.
Dimethyl sulphoxide
Bradykinin antagonists
Dimethyl sulphoxide (DMSO) has been reported to
Tissue injury and subsequent cell death act as strong result in strong diuresis, protect cells from mechan-
triggers for the initiation of an inflammatory ical damage, reduce oedema in tissue by stabilizing
response. The kinin-kallikrein pathway is one of cell membranes and act as a free radical scavenger
the components of this acute inflammatory cascade [91]. DMSO is also believed to increase tissue
following traumatic brain injury [86, 87]. The perfusion, thereby improving cell oxygenation, neu-
generation of bradykinin leads to a detrimental tralizing metabolic acidosis and decreasing intracel-
cascade of events ultimately ending in altered lular fluid retention [91]. This study identified three
vascular permeability and tissue oedema [88]. studies examining the effects of DMSO in the
Animal research using BK2 receptor knockout mice management of ICP and brain swelling post ABI
has demonstrated direct involvement of this receptor (Table X).
in the development of inflammatory-induced sec- In a study using a single group intervention
ondary damage and subsequent neurological deficits design, Kulah et al. [91] demonstrated that, for the
resulting from diffuse TBI [89]. These findings majority of participants, DMSO was effective in
suggest that specific inhibition of the BK2 receptor controlling ICP elevations within minutes of injec-
could prove to be an effective therapeutic strategy tion, which was accompanied by a concomitant
following brain injury. Three studies evaluating the increase in CPP. Unfortunately, these benefits
efficacy of BradycorTM and Anatibant (two bradyki- appear to be transient, since continuous infusions
nin antagonists) in the acute treatment of ABI were of DMSO for up to 7 days failed to control
reviewed (Table IX). elevations in ICP. In a similar study, Karaca et al.
Narotam et al. [87] reported treatment with [92] reported that, although reductions in ICP were
BradycorTM resulted in significant reductions in seen within the first 30 minutes of DMSO adminis-
ICP elevation and decreased deterioration in GCS tration, the effect was not sustained and most
scores over the course of the study. Furthermore, the patients required maintenance doses for 2–10 days
Acute ABI: Pharmacological 717

to minimize fluctuations in ICP. Marshall et al. [93] that propofol infusion greater than 4 mg kg1 h1
reported that rapid infusions of DMSO were effec- may result in propofol infusion syndrome, a danger-
tive in controlling elevated ICP in patients for whom ous and potentially fatal adverse effect.
ICP could not be controlled by standard measures. Barbiturate sedation remains questionable in
All three studies scored low on methodological ABI care. There is conflicting evidence regarding
quality, limiting the strength of conclusions. the benefits of barbiturate use in ICP management
Therefore, there is level 4 evidence that dimethyl and some indication that mannitol may be more
sulphoxide transiently reduces ICP elevations, but effective in this regard. Also, there is evidence to
further study is required (Table XII). suggest that barbiturate use can lead to negative
side-effects including reversible leukopenia, granu-
Cannabinoids locytopenia and systemic hypotension. Barbiturates
have increasingly limited indication and, given
Dexanabinol (HU-211) is a synthetic, non-
the benefits noted with use of alternate sedatives
psychotropic cannabinoid [94] thought to act as
including propofol and midazolam, it is questionable
a non-competitive N-methyl-D-aspartate receptor
as to whether further study into the role of
antagonist to decrease glutamate excito-toxicity
barbiturates in ABI management is appropriate.
[95]. Dexanabinol is believed to possess antioxidant
Opioid administration leads to increases in ICP
properties [96] and has shown encouraging neuro-
if given via bolus injection; however, there was
Brain Inj Downloaded from informahealthcare.com by HINARI on 03/21/11

protective effects in animal models of TBI [97].


conflicting evidence regarding the effects of infusion
Two studies on cannabinoid use in humans were
administration on ICP levels. One study also
reviewed (Table XI).
suggested that remifentanil may allow for more
Knoller et al. [98] reported that the active drug
rapid arousal relative to hypnotic-based sedation.
group showed significant improvements in ICP and
Given the prevalence of opioid administration in
CPP. However, despite showing initially significant
patients with brain injury, further research into their
improvements on the GOS and DRS at 1 month
effect on ICP is necessary.
For personal use only.

post-treatment, these benefits were no longer signif-


Midazolam was found to provide similar quality
icant at 6-month follow-up. Subsequently, Maas
sedation to propofol with no increase in ICP.
et al. [99] conducted a large-scale multi-centre RCT
However, midazolam has been linked to significant
and reported that, compared with placebo, dexana-
hypotension resulting in decreases in CPP that
binol did not significantly improve outcomes
require monitoring upon administration.
(i.e. extended GOS, mortality rates, Barthel
Mannitol is a common treatment for management
index or quality of life measures (SF36, CIQ)) at
of elevated ICP and this review resulted in strong
6-months. Moreover, dexanabinol failed to provide
evidence that mannitol is effective in reducing ICP.
acute control of derangements in ICP or CPP [99].
However, these benefits may only be seen when
These negative findings suggest that the initial
ICP is already elevated. Moreover, there was strong
benefits reported by Knoller et al. [98] could have
evidence that mannitol was less effective than
been due to the small sample size in this earlier
hypertonic saline in reducing elevated ICP.
study. Therefore, there was level 1 evidence that
Hypertonic saline was shown to be very effective in
dexanabinol does not result in acute improvement
reducing elevated ICP and may reduce mortality
in ICP or benefits in long-term outcomes
rates in severely injured patients. For this reason,
(Table XII).
hypertonic saline may be the optimal osmotic agent
for use in acute ABI care. However, one study found
that hypertonic saline may result in greater swelling
Discussion
of contused tissue. Further study into this finding
This review evaluated a total of 11 pharmacological appears warranted.
interventions used in the acute management of Based on a large scale international trial, corticos-
acquired brain injury. Of these, corticosteroids teroid use in patients with ABI was found to increase
were found to be contraindicated and cannabinoids mortality rates. Similarly, this review found the
were shown to be ineffective. Each of the other majority of studies demonstrated a lack of improve-
interventions showed promise in acute ABI care. ment associated with corticosteroid use and evidence
However, limited efficacy was found for improved that they may lead to increases in early onset
long-term outcomes and specific applications for seizures. Only triamcinolone was found to improve
many of the interventions require further study. patient outcomes in severely injured patients with
Evidence suggests that propofol sedation is safe focal lesions; however, in light of the other studies,
and effective; however, its benefit without concom- these findings should be interpreted cautiously. This
itant morphine administration requires further inves- review supports the recommendation that corticos-
tigation. There is also consensus evidence suggesting teroids should not be used in acute ABI care.
718 M. J. Meyer et al.

Progesterone has gained interest as a possible 2. Teasell R, Bayona N, Marshall S, Cullen N, Bayley M,
neuroprotective agent post-ABI. This review located Chundamala J, Villamere J, Mackie D, Rees L, Hartridge C,
et al. A systematic review of the rehabilitation of moderate to
two trials into progesterone use, both of which severe acquired brain injuries. Brain Injury 2007;21:
resulted in beneficial outcomes and decreased mor- 107–112.
tality. Although one trial was a preliminary phase II 3. Fleminger S, Greenwood RJ, Oliver DL. Pharmacological
trial, there is evidence to suggest that progesterone management for agitation and aggression in people with
is safe and effective, warranting further study. acquired brain injury. Cochrane Database System Reviews
2006;4:CD003299.
Three other less common interventions for ICP
4. ERABI group. Evidence-based review of moderate to severe
management were also reviewed. There is an indi- acquired brain injury. Accessed October 2008 October.
cation that the bradykinin antagonists Bradycor and Available online at: www.abiebr.com
Anatibant result in transient reductions in ICP; 5. Moseley AM, Herbert RD, Sherrington C, Maher CG.
however, further study into their effect on long-term Evidence for physiotherapy practice: A survey of the
outcomes is necessary. Dimethyl sulphoxide Physiotherapy Evidence Database (PEDro). Australian
Journal of Physiotherapy 2002;48:43–49.
was shown to transiently reduce ICP levels in the 6. Downs SH, Black N. The feasibility of creating a checklist
short-term and may be beneficial in instances where for the assessment of the methodological quality both of
ICP is unresponsive to other treatments. Finally, randomised and non-randomised studies of health care
despite initial promise, larger studies suggest canna- interventions. Journal of Epidemiology and Community
Health 1998;52:377–384.
Brain Inj Downloaded from informahealthcare.com by HINARI on 03/21/11

binoids are not effective in reducing ICP or


7. Adembri C, Venturi L, Pellegrini-Giampietro DE.
improving outcomes post-ABI.
Neuroprotective effects of propofol in acute cerebral injury.
Pharmacological interventions will continue to CNS Drug Review 2007;13:333–351.
play a key role in the acute management of ABI. 8. Kelly DF, Goodale DB, Williams J, Herr DL, Chappell ET,
Excluding corticosteroids and cannabinoids, each of Rosner MJ, Jacobson J, Levy ML, Croce MA, Maniker AH,
the other nine interventions showed promise in some et al. Propofol in the treatment of moderate and severe head
injury: A randomized, prospective double-blinded pilot trial.
aspect of acute care. Given the diversity of complica-
Journal of Neurosurgery 1999;90:1042–1052.
tions associated with brain injury, treatment often
For personal use only.

9. Farling PA, Johnston JR, Coppel DL. Propofol infusion for


requires combinations of medications that make sedation of patients with head injury in intensive care.
isolating the effect of a single intervention difficult. A preliminary report. Anaesthesia 1989;44:222–226.
Future research should aim to establish large scale 10. Stewart L, Bullock R, Rafferty C, Fitch W, Teasdale GM.
trials similar to the CRASH trial for corticosteroid Propofol sedation in severe head injury fails to control high
ICP, but reduces brain metabolism. Acta Neurochirgica
use so that stronger and more definitive conclusions Supplementum (Wien) 1994;60:544–546.
can be drawn. Also, further research into the effects 11. Corbett SM, Moore J, Rebuck JA, Rogers FB, Greene CM.
of interventions within different patient groups will Survival of propofol infusion syndrome in a head-injured
allow for more individualized treatment strategies patient. Critical Care Medicine 2006;34:2479–2483.
resulting in improved patient outcomes. 12. Sabsovich I, Rehman Z, Yunen J, Coritsidis G. Propofol
infusion syndrome: A case of increasing morbidity with
traumatic brain injury. American Journal of Critical Care
2007;16:82–85.
Conclusions
13. Cremer OL, Moons KG, Bouman EA, Kruijswijk JE,
Substantial research has been devoted to evaluating de Smet AM, Kalkman CJ. Long-term propofol infusion
the use of pharmacological interventions in the acute and cardiac failure in adult head-injured patients. Lancet
2001;357:117–118.
management of ABI. Based on the results of this 14. Otterspoor LC, Kalkman CJ, Cremer OL. Update on the
review, corticosteroids were found to be contra- propofol infusion syndrome in ICU management of patients
indicated and cannabinoids were reported as inef- with head injury. Current Opinion in Anaesthesiology
fective. The other nine interventions demonstrated 2008;21:544–551.
some benefit for treatment of acute ABI. However, 15. Roberts I. Barbiturates for acute traumatic brain injury.
Cochrane Database System Reviews 2000;2:CD000033.
rarely did these benefits result in improved
16. Brain Injury Special Interest Group of the American
long-term patient outcomes. Academy of Physical Medicine and Rehabilitation. Practice
parameter: Antiepileptic drug treatment of posttraumatic
seizures. Archives of Physical Medicine and Rehabilitation
1998;79:594–597.
Declaration of interest: The authors report no
17. Rea GL, Rockswold GL. Barbiturate therapy in uncontrolled
conflict of interest. The authors alone are responsi- intracranial hypertension. Neurosurgery 1983;12:401–404.
ble for the content and writing of the paper. 18. Rockoff MA, Marshall LF, Shapiro HM. High-dose barbi-
turate therapy in humans: A clinical review of 60 patients.
Annals of Neurology 1979;6:194–199.
19. Marshall LF, Smith RW, Shapiro HM. The outcome with
References aggressive treatment in severe head injuries. Part II: Acute
1. Zasler ND, Katz DI, Zafonte RD. Brain injury medicine. and chronic barbiturate administration in the management of
New York: Demos Medical Publishing; 2007. head injury. Journal of Neurosurgery 1979;50:26–30.
Acute ABI: Pharmacological 719

20. Llompart-Pou JA, Perez-Barcena J, Raurich JM, Burguera B, 36. de Nadal M, Munar F, Poca MA, Sahuquillo J, Garnacho A,
Ayestaran JI, Abadal JM, et al. Effect of barbiturate coma on Rossello J. Cerebral hemodynamic effects of morphine
adrenal response in patients with traumatic brain injury. and fentanyl in patients with severe head injury: Absence of
Journal of Endocrinology Investigations 2007;30:393–398. correlation to cerebral autoregulation. Anesthesiology
21. Stover JF, Stocker R. Barbiturate coma may promote 2000;92:11–19.
reversible bone marrow suppression in patients with severe 37. Engelhard K, Reeker W, Kochs E, Werner C. Effect of
isolated traumatic brain injury. European Journal of Clinical remifentanil on intracranial pressure and cerebral blood flow
Pharmacology 1998;54:529–534. velocity in patients with head trauma. Acta Anaesthesiologica
22. Eisenberg HM, Frankowski RF, Contant CF, Marshall LF, Scandinavica 2004;48:396–399.
Walker MD. High-dose barbiturate control of elevated 38. Lauer KK, Connolly LA, Schmeling WT. Opioid sedation
intracranial pressure in patients with severe head injury. does not alter intracranial pressure in head injured patients.
Journal of Neurosurgery 1988;69:15–23. Canadian Journal of Anaesthesia 1997;44:929–933.
23. Ward JD, Becker DP, Miller JD, Choi SC, Marmarou A, 39. Scholz J, Bause H, Schulz M, Klotz U, Krishna DR, Pohl S,
Wood C, Newlon PG, Keenan R. Failure of prophylactic Schulte AM, Esch J. Pharmacokinetics and effects on
barbiturate coma in the treatment of severe head injury. intracranial pressure of sufentanil in head trauma patients.
Journal of Neurosurgery 1985;62:383–388. British Journal of Clinical Pharmacology 1994;38:369–372.
24. Schwartz ML, Tator CH, Rowed DW, Reid SR, Meguro K, 40. Karabinis A, Mandragos K, Stergiopoulos S, Komnos A,
Andrews DF. The University of Toronto head injury Soukup J, Speelberg B, Kirkham AJ. Safety and efficacy of
treatment study: A prospective, randomized comparison analgesia-based sedation with remifentanil versus standard
of pentobarbital and mannitol. Canadian Journal of hypnotic-based regimens in intensive care unit patients with
Neurological Science 1984;11:434–440. brain injuries: A randomised, controlled trial
Brain Inj Downloaded from informahealthcare.com by HINARI on 03/21/11

25. Perez-Barcena J, Barcelo B, Homar J, Abadal JM, Molina FJ, [ISRCTN50308308]. Critical Care 2004;8:R268–R280.
de la Pena A, Sahuquillo J, Ibanez J. [Comparison of the 41. McCollam JS, O’Neil MG, Norcross ED, Byrne TK,
effectiveness of pentobarbital and thiopental in patients with Reeves ST. Continuous infusions of lorazepam, midazolam,
refractory intracranial hypertension. Preliminary report of and propofol for sedation of the critically ill surgery trauma
20 patients]. Neurocirugia 2005;16:5–12. patient: A prospective, randomized comparison. Critical Care
26. Nordby HK, Nesbakken R. The effect of high dose Medicine 1999;27:2454–2458.
barbiturate decompression after severe head injury. 42. McClelland M, Woster P, Sweasey T, Hoff JT. Continuous
A controlled clinical trial. Acta Neurochirgica (Wien) midazolam/atracurium infusions for the management of
1984;72:157–166.
For personal use only.

increased intracranial pressure. Journal of Neuroscience


27. Schalen W, Messeter K, Nordstrom CH. Complications and
Nursing 1995;27:96–101.
side effects during thiopentone therapy in patients with severe
43. Davis DP, Kimbro TA, Vilke GM. The use of midazolam
head injuries. Acta Anaesthesiologica Scandinavica 1992;36:
for prehospital rapid-sequence intubation may be associated
369–377.
with a dose-related increase in hypotension. Prehospital
28. Metz C, Gobel L, Gruber M, Hoerauf KH, Taeger K.
Emergency Care 2001;5:163–168.
Pharmacokinetics of human cerebral opioid extraction:
44. Sanchez-Izquierdo-Riera JA, Caballero-Cubedo RE, Perez-
A comparative study on sufentanil, fentanyl, and alfentanil
Vela JL, Ambros-Checa A, Cantalapiedra-Santiago JA,
in a patient after severe head injury. Anesthesiology 2000;92:
ted-Lopez E. Propofol versus midazolam: Safety and efficacy
1559–1567.
for sedating the severe trauma patient. Anesthesia and
29. de Nadal M, Munar F, Poca MA, Sahuquillo J, Garnacho A,
Analgesia 1998;86:1219–1224.
Rossello J. Cerebral hemodynamic effects of morphine
45. Papazian L, Albanese J, Thirion X, Perrin G, Durbec O,
and fentanyl in patients with severe head injury: Absence of
Martin C. Effect of bolus doses of midazolam on intracranial
correlation to cerebral autoregulation. Anesthesiology
pressure and cerebral perfusion pressure in patients with
2000;92:11–19.
severe head injury. British Journal of Anaesthesia 1993;71:
30. Werner C, Kochs E, Bause H, Hoffman WE, Schulte am EJ.
Effects of sufentanil on cerebral hemodynamics and intra- 267–271.
46. Wakai A, Roberts I, Schierhout G. Mannitol for acute
cranial pressure in patients with brain injury. Anesthesiology
1995;83:721–726. traumatic brain injury. Cochrane Database System Reviews
31. Marx W, Shah N, Long C, Arbit E, Galicich J, Mascott C, 2005;4:CD001049.
Mallya K, Bedford R. Sufentanil, alfentanil, and fentanyl: 47. Battison C, Andrews PJ, Graham C, Petty T. Randomized,
Impact on cerebrospinal fluid pressure in patients with brain controlled trial on the effect of a 20% mannitol solution and a
tumors. Journal of Neurosurgery and Anesthesiology 1989;1: 7.5% saline/6% dextran solution on increased intracranial
3–7. pressure after brain injury. Critical Care Medicine 2005;33:
32. Bunegin L, Albin MS, Ernst PS, Garcia C. Cerebrovascular 196–202.
responses to sufentanil citrate (SC) in primates with and 48. Doyle JA, Davis DP, Hoyt DB. The use of hypertonic saline
without intracranial hypertension. Journal of Neurosurgery in the treatment of traumatic brain injury. Journal of Trauma
and Anesthesiology 1989;1:138–139. 2001;50:367–383.
33. Albanese J, Viviand X, Potie F, Rey M, Alliez B, Martin C. 49. Task force of the American Association of Neurological
Sufentanil, fentanyl, and alfentanil in head trauma patients: Surgeons and Joint Section in Neurotrauma and Critical
A study on cerebral hemodynamics. Critical Care Medicine Care. Guidelines for the management of severe head injury.
1999;27:407–411. Brain Trauma Foundation. 1995. Available online at: http://
34. Albanese J, Durbec O, Viviand X, Potie F, Alliez B, www.guideline.gov/summary/summary.aspx?doc_id¼3794&
Martin C. Sufentanil increases intracranial pressure in nbr¼003020&string¼3020
patients with head trauma. Anesthesiology 1993;79:493–497. 50. Cruz J, Minoja G, Okuchi K, Facco E. Successful use of the
35. Sperry RJ, Bailey PL, Reichman MV, Peterson JC, new high-dose mannitol treatment in patients with Glasgow
Petersen PB, Pace NL. Fentanyl and sufentanil increase Coma Scale scores of 3 and bilateral abnormal pupillary
intracranial pressure in head trauma patients. Anesthesiology widening: A randomized trial. Journal of Neurosurgery
1992;77:416–420. 2004;100:376–383.
720 M. J. Meyer et al.

51. Cruz J, Minoja G, Okuchi K. Improving clinical outcomes 65. Myburgh J, Cooper DJ, Finfer S, Bellomo R, Norton R,
from acute subdural hematomas with the emergency Bishop N, Kai Lo S. Saline or albumin for fluid resuscitation
preoperative administration of high doses of mannitol: in patients with traumatic brain injury. New England Journal
A randomized trial. Neurosurgery 2001;49:864–871. of Medicine 2007;357:874–884.
52. Cruz J, Minoja G, Okuchi K. Major clinical and physiological 66. Cooper DJ, Myles PS, McDermott FT, Murray LJ,
benefits of early high doses of mannitol for intraparenchymal Laidlaw J, Cooper G, Tremayne AB, Bernard SS,
temporal lobe hemorrhages with abnormal pupillary Ponsford J. Prehospital hypertonic saline resuscitation of
widening: A randomized trial. Neurosurgery 2002;51: patients with hypotension and severe traumatic brain injury:
628–637. A randomized controlled trial. Journal of the American
53. Sorani MD, Morabito D, Rosenthal G, Giacomini KM, Medical Association 2004;291:1350–1357.
Manley GT. Characterizing the dose–response relationship 67. Simma B, Burger R, Falk M, Sacher P, Fanconi S.
between mannitol and intracranial pressure in traumatic A prospective, randomized, and controlled study of fluid
brain injury patients using a high-frequency physiological management in children with severe head injury: Lactated
data collection system. Journal of Neurotrauma 2008;25: Ringer’s solution versus hypertonic saline. Critical Care
291–298. Medicine 1998;26:1265–1270.
54. Bratton SL, Chestnut RM, Ghajar J, Connell Hammond FF, 68. Lescot T, Degos V, Zouaoui A, Preteux F, Coriat P,
Harris OA, Hartl R, et al. Guidelines for the management of Puybasset L. Opposed effects of hypertonic saline on
severe traumatic brain injury. XI. Anesthetics, analgesics, contusions and noncontused brain tissue in patients with
and sedatives. Journal of Neurotrauma 2007;24(Suppl 1): severe traumatic brain injury. Critical Care Medicine
S71–S76. 2006;34:3029–3033.
55. Smith HP, Kelly Jr DL, McWhorter JM, Armstrong D, 69. Qureshi AI, Suarez JI, Bhardwaj A, Mirski M, Schnitzer MS,
Brain Inj Downloaded from informahealthcare.com by HINARI on 03/21/11

Johnson R, Transou C, Howard G. Comparison of mannitol Hanley DF, Ulatowski JA. Use of hypertonic (3%) saline/
regimens in patients with severe head injury undergoing acetate infusion in the treatment of cerebral edema: Effect on
intracranial monitoring. Journal of Neurosurgery 1986;65: intracranial pressure and lateral displacement of the brain.
820–824. Critical Care Medicine 1998;26:440–446.
56. Hartl R, Bardt TF, Kiening KL, Sarrafzadeh AS, 70. Schatzmann C, Heissler HE, Konig K, Klinge-Xhemajli P,
Schneider GH, Unterberg AW. Mannitol decreases ICP but Rickels E, Muhling M, Borschel M, Samii M. Treatment of
does not improve brain-tissue pO2 in severely head-injured elevated intracranial pressure by infusions of 10% saline in
patients with intracranial hypertension. Acta Neurochirgica severely head injured patients. Acta Neurochirgica
For personal use only.

Supplementum 1997;70:40–42. Supplementum 1998;71:31–33.


57. Vialet R, Albanese J, Thomachot L, Antonini F, 71. Pascual JL, Maloney-Wilensky E, Reilly PM, Sicoutris C,
Bourgouin A, Alliez B, Martin C. Isovolume hypertonic Keutmann MK, Stein SC, LeRoux PD, Gracias VH.
solutes (sodium chloride or mannitol) in the treatment of Resuscitation of hypotensive head-injured patients: Is hyper-
refractory posttraumatic intracranial hypertension: 2 mL/kg tonic saline the answer? American Surgery 2008;74:253–259.
7.5% saline is more effective than 2 mL/kg 20% mannitol. 72. Alderson P, Roberts I. Corticosteroids for acute traumatic
Critical Care Medicine 2003;31:1683–1687. brain injury. Cochrane Database System Reviews 2005;1:
58. Ware ML, Nemani VM, Meeker M, Lee C, Morabito DJ, CD000196.
Manley GT. Effects of 23.4% sodium chloride solution in 73. Grumme T, Baethmann A, Kolodziejczyk D, Krimmer J,
reducing intracranial pressure in patients with traumatic Fischer M, von Eisenhart RB, Rothe B, Pelka R,
brain injury: A preliminary study. Neurosurgery 2005;57: Bennefled H, Pollauer E. Treatment of patients with severe
727–736. head injury by triamcinolone: A prospective, controlled
59. Francony G, Fauvage B, Falcon D, Canet C, Dilou H, multicenter clinical trial of 396 cases. Research in
Lavagne P, Jacquot C, Payen JF. Equimolar doses of Experimental Medicine (Berlin) 1995;195:217–229.
mannitol and hypertonic saline in the treatment of increased 74. Cooper PR, Moody S, Clark WK, Kirkpatrick J, Maravilla K,
intracranial pressure. Critical Care Medicine 2008;36: Gould AL, Drane W. Dexamethasone and severe head injury.
795–800. A prospective double-blind study. Journal of Neurosurgery
60. Sayre MR, Daily SW, Stern SA, Storer DL, van Loveren HR, 1979;51:307–316.
Hurst JM. Out-of-hospital administration of mannitol to 75. Saul TG, Ducker TB, Salcman M, Carro E. Steroids in
head-injured patients does not change systolic blood pres- severe head injury: A prospective randomized clinical trial.
sure. Acadamy of Emergency Medicine 1996;3:840–848. Journal of Neurosurgery 1981;54:596–600.
61. Horn P, Munch E, Vajkoczy P, Herrmann P, Quintel M, 76. Giannotta SL, Weiss MH, Apuzzo ML, Martin E. High dose
Schilling L, Schmiedek P, Schurer L. Hypertonic saline glucocorticoids in the management of severe head injury.
solution for control of elevated intracranial pressure in Neurosurgery 1984;15:497–501.
patients with exhausted response to mannitol and barbitu- 77. Dearden NM, Gibson JS, McDowall DG, Gibson RM,
rates. Neurology Research 1999;21:758–764. Cameron MM. Effect of high-dose dexamethasone on
62. Khanna S, Davis D, Peterson B, Fisher B, Tung H, outcome from severe head injury. Journal of Neurosurgery
O’Quigley J, Deutsch R. Use of hypertonic saline in the 1986;64:81–88.
treatment of severe refractory posttraumatic intracranial 78. Braakman R, Schouten HJ, Blaauw-van DM,
hypertension in pediatric traumatic brain injury. Critical Minderhoud JM. Megadose steroids in severe head injury.
Care Medicine 2000;28:1144–1151. Results of a prospective double-blind clinical trial. Journal of
63. Suarez JI. Hypertonic saline for cerebral edema and elevated Neurosurgery 1983;58:326–330.
intracranial pressure. Cleveland Clinic Journal of Medicine 79. Kaktis JV, Pitts LH. Complications associated with use of
2004;71(Suppl 1):S9–S13. megadose corticosteroids in head-injured adults. Journal of
64. Shackford SR, Bourguignon PR, Wald SL, Rogers FB, Neurosurgery Nursing 1980;12:166–171.
Osler TM, Clark DE. Hypertonic saline resuscitation of 80. Watson NF, Barber JK, Doherty MJ, Miller JW, Temkin NR.
patients with head injury: A prospective, randomized clinical Does glucocorticoid administration prevent late seizures after
trial. Journal of Trauma 1998;44:50–8. head injury? Epilepsia 2004;45:690–694.
Acute ABI: Pharmacological 721

81. Stein DG. Progesterone exerts neuroprotective effects after 93. Shohami E, Novikov M, Bass R. Long-term effect of
brain injury. Brain Research Review 2008;57:386–397. HU-211, a novel non-competitive NMDA antagonist, on
82. Vagnerova K, Koerner IP, Hurn PD. Gender and the injured motor and memory functions after closed head injury in the
brain. Anesthesia and Analgesia 2008;107:201–214. rat. Brain Research 1995;674:55–62.
83. Marmarou A, Nichols J, Burgess J, Newell D, Troha J, 94. Knoller N, Levi L, Shoshan I, Reichenthal E, Razon N,
Burnham D, Pitts L. Effects of the bradykinin antagonist Rappaport ZH, Biegon A. Dexanabinol (HU-211) in the
Bradycor (deltibant, CP-1027) in severe traumatic brain treatment of severe closed head injury: A randomized,
injury: Results of a multi-center, randomized, placebo- placebo-controlled, phase II clinical trial. Critical Care
controlled trial. American Brain Injury Consortium Study Medicine 2002;30:548–554.
Group. Journal of Neurotrauma 1999;16:431–444. 95. Maas AI, Murray G, Henney III H, Kassem N, Legrand V,
84. Narotam PK, Rodell TC, Nadvi SS, Bhoola KD, Troha JM, Mangelus M, Muizelaar JP, Stocchetti N, Knoller N.
Parbhoosingh R, van Dellen JR. Traumatic brain contusions: Efficacy and safety of dexanabinol in severe traumatic
A clinical role for the kinin antagonist CP-0127. Acta brain injury: Results of a phase III randomised, placebo-
Neurochirgica (Wien) 1998;140:793–802. controlled, clinical trial. Lancet Neurology 2006;5:38–45.
85. Francel PC. Bradykinin and neuronal injury. Journal of 96. Fried RC, Dickerson RN, Guenter PA, Stein TP,
Neurotrauma 1992;9(Suppl 1):S27–S45. Gennarelli TA, Dempsey DT, Buzby GP, Mullen JL.
86. Hellal F, Pruneau D, Palmier B, Faye P, Croci N, Barbiturate therapy reduces nitrogen excretion in acute
Plotkine M, Marchand Verrecchiac. Detrimental role of head injury. Journal of Trauma 1989;29:1558–1564.
bradykinin B2 receptor in a murine model of diffuse brain 97. Thorat JD, Wang EC, Lee KK, Seow WT, Ng I. Barbiturate
injury. Journal of Neurotrauma 2003;20:841–851. therapy for patients with refractory intracranial hypertension
87. Marmarou A, Guy M, Murphey L, Roy F, Layani L, following severe traumatic brain injury: Its effects on tissue
Brain Inj Downloaded from informahealthcare.com by HINARI on 03/21/11

Combal JP, Marquer C. A single dose, three-arm, placebo-


oxygenation, brain temperature and autoregulation. Journal
controlled, phase I study of the bradykinin B2 receptor
of Clinical Neuroscience 2008;15:143–148.
antagonist Anatibant (LF16-0687Ms) in patients with severe
98. Schalen W, Sonesson B, Messeter K, Nordstrom G,
traumatic brain injury. Journal of Neurotrauma 2005;22:
Nordstrom CH. Clinical outcome and cognitive impairment
1444–1455.
in patients with severe head injuries treated with barbiturate
88. Kulah A, Akar M, Baykut L. Dimethyl sulfoxide in the
coma. Acta Neurochirgica (Wien) 1992;117:153–159.
management of patient with brain swelling and increased
99. Roberts I, Yates D, Sandercock P, Farrell B, Wasserberg J,
intracranial pressure after severe closed head injury.
Lomas G, Cottingham R, Svaboda P, Brayley N,
Neurochirurgia (Stuttgart) 1990;33:177–180.
For personal use only.

Mazairac G, et al. Effect of intravenous corticosteroids on


89. Marshall LF, Camp PE, Bowers SA. Dimethyl sulfoxide for
the treatment of intracranial hypertension: A preliminary death within 14 days in 10008 adults with clinically
trial. Neurosurgery 1984;14:659–663. significant head injury (MRC CRASH trial): Randomised
90. Mechoulam R, Feigenbaum JJ, Lander N, Segal M, placebo-controlled trial. Lancet 2004;364:1321–1328.
Jarbe TU, Hiltunen AJ, Consroe P. Enantiomeric cannabi- 100. Wright DW, Kellermann AL, Hertzberg VS, Clark PL,
noids: Stereospecificity of psychotropic activity. Experientia Frankel M, Goldstein FC, Salomone JP, Dent LL,
1988;44:762–764. Harris OA, Ander DS, et al. ProTECT: A randomized
91. Feigenbaum JJ, Bergmann F, Richmond SA, Mechoulam R, clinical trial of progesterone for acute traumatic brain injury.
Nadler V, Kloog Y, Sakolovsky M. Nonpsychotropic Annals of Emergency Medicine 2007;49:391–402.
cannabinoid acts as a functional N-methyl-D-aspartate 101. Xiao G, Wei J, Yan W, Wang W, Lu Z. Improved outcomes
receptor blocker. Proceedings of the National Academy of from the administration of progesterone for patients with
Sciences (USA) 1989;86:9584–9587. acute severe traumatic brain injury: A randomized con-
92. Eshhar N, Striem S, Kohen R, Tirosh O, Biegon A. trolled trial. Critical Care 2008;12:R61.
Neuroprotective and antioxidant activities of HU-211, 102. Karaca M, Bilgin UY, Akar M, de la Torre JC. Dimethly
a novel NMDA receptor antagonist. European Journal of sulphoxide lowers ICP after closed head trauma. European
Pharmacology 1995;283:19–29. Journal of Clinical Pharmacology 1991;40:113–114.

Você também pode gostar