14
Controversies in the management of aneurysmal subarachnoid hemorrhage* Neeraj S. Naval, MD; Robert D. Stevens, MD; Marek A. Mirski, MD, PhD; Anish Bhardwaj, MD, FCCM T he rupture of an intracranial aneurysm may be associated with an array of severe distur- bances in intracranial and sys- temic physiology that represent a unique challenge to the clinician. Surgical man- agement has traditionally emphasized the prevention of renewed intracranial bleed- ing by clipping or wrapping the respon- sible aneurysm. Medical management is based on the detection and treatment of cerebral and extracerebral complications of aneurysmal subarachnoid hemorrhage (aSAH). Cerebral complications of aSAH include recurrent intracranial hemor- rhage, vasospasm, cerebral infarction, hy- drocephalus, cerebral edema, and intra- cranial hypertension; extracerebral complications include respiratory failure, derangements of water and electrolyte homeostasis, myocardial dysfunction, sepsis, and thromboembolism. As many of these complications are life-threaten- ing but reversible, it is widely believed that patients with aSAH can benefit from management in an intensive care setting. Recent years have seen a considerable expansion in the use of image-guided en- dovascular therapies for aSAH, including coiling of aneurysms and balloon angio- plasty or intraarterial drug delivery for cerebral vasospasm (1– 4). These ad- vances have occurred in a general setting of increasing knowledge of aSAH epide- miology, pathophysiology, diagnosis, and prevention and of significant refinements in microsurgical technique and in medi- cal therapy. There is a widespread percep- tion that this broader understanding and expertise is yielding benefits in the form of improved outcome after aSAH. Indeed, a progressive increase in aSAH survival over the past three decades has been re- ported in several studies (5–7). However, evidence of a direct relationship between aSAH outcomes and a specific strategy or intervention is limited (2, 3, 8, 9). In several key areas of management, sup- porting data are lacking or equivocal in nature, generating uncertainty and con- troversy among clinicians. This review focuses on controversies that are central to the acute management of aSAH. These include surgical vs. endo- vascular aneurysm repair, the diagnosis and management of cerebral vasospasm, neuroprotective strategies, use of anti- thrombotic agents (thrombolytic agents, heparin, and platelet inhibitors), prophy- laxis of seizures, and the approach to ce- rebral salt wasting and to cardiac dys- function after aSAH. For each controversy, a critical evaluation of the available evidence is coupled with recom- mendations for further clinical investiga- tion. The review is selective, focusing on the principal debates at the expense of other equally important but arguably less controversial issues (e.g., post-aSAH hy- drocephalus and ventricular drainage). Controversy 1: Surgical vs. Endovascular Aneurysm Repair Endovascular coiling emerged as an alternative to surgery in patients with intracranial aneurysms who were deemed poor surgical candidates due to signifi- cant neurologic injury, the presence of severe medical co-morbidities, or difficult surgical access to the aneurysm (1). More recent work has sought to extend the indications of endovascular coiling to other patient categories. In a small ran- domized trial of 109 patients with aSAH, 3- and 12-month clinical and neuropsy- chological outcomes were the same be- tween the surgical group and the endo- vascular group (10). This was followed by *See also p. 571. From the Division of Neurosciences Critical Care, Departments of Neurology, Anesthesiology/Critical Care Medicine, and Neurological Surgery, Johns Hop- kins University School of Medicine, Baltimore, MD. Supported, in part, by U.S. Public Health Service National Institutes of Health NS NS046379 and by an Established Investigator Grant (A. Bhardwaj) from the American Heart Association. The authors have no financial interests to disclose. Copyright © 2006 by the Society of Critical Care Medicine and Lippincott Williams & Wilkins DOI: 10.1097/01.CCM.0000198331.45998.85 Background: The care of patients with aneurysmal subarach- noid hemorrhage has evolved significantly with the advent of new diagnostic and therapeutic modalities. Although it is believed that these advances have contributed to improved outcomes, consid- erable uncertainty persists regarding key areas of management. Objective: To review selected controversies in the manage- ment of aneurysmal subarachnoid hemorrhage, with a special emphasis on endovascular vs. surgical techniques for securing aneurysms, the diagnosis and therapy of cerebral vasospasm, neuroprotection, antithrombotic and anticonvulsant agents, cere- bral salt wasting, and myocardial dysfunction, and to suggest venues for further clinical investigation. Data Source: Search of MEDLINE and Cochrane databases and manual review of article bibliographies. Data Synthesis and Conclusions: Many aspects of care in patients with aneurysmal subarachnoid hemorrhage remain highly controversial and warrant further resolution with hypoth- esis-driven clinical or translational research. It is anticipated that the rigorous evaluation and implementation of such data will provide a basis for improvements in short- and long-term out- comes. (Crit Care Med 2006; 34:511–524) KEY WORDS: aneurysm; subarachnoid; hemorrhage; vasospasm; ischemia 511 Crit Care Med 2006 Vol. 34, No. 2

Controversies in the management of aneurysmal subarachnoid ... · sible aneurysm. Medical management is based on the detection and treatment of cerebral and extracerebral complications

  • Upload
    others

  • View
    4

  • Download
    0

Embed Size (px)

Citation preview

Page 1: Controversies in the management of aneurysmal subarachnoid ... · sible aneurysm. Medical management is based on the detection and treatment of cerebral and extracerebral complications

Controversies in the management of aneurysmal subarachnoidhemorrhage*

Neeraj S. Naval, MD; Robert D. Stevens, MD; Marek A. Mirski, MD, PhD; Anish Bhardwaj, MD, FCCM

T he rupture of an intracranialaneurysm may be associatedwith an array of severe distur-bances in intracranial and sys-

temic physiology that represent a uniquechallenge to the clinician. Surgical man-agement has traditionally emphasized theprevention of renewed intracranial bleed-ing by clipping or wrapping the respon-sible aneurysm. Medical management isbased on the detection and treatment ofcerebral and extracerebral complicationsof aneurysmal subarachnoid hemorrhage(aSAH). Cerebral complications of aSAHinclude recurrent intracranial hemor-rhage, vasospasm, cerebral infarction, hy-drocephalus, cerebral edema, and intra-cranial hypertension; extracerebralcomplications include respiratory failure,derangements of water and electrolytehomeostasis, myocardial dysfunction,

sepsis, and thromboembolism. As manyof these complications are life-threaten-ing but reversible, it is widely believedthat patients with aSAH can benefit frommanagement in an intensive care setting.

Recent years have seen a considerableexpansion in the use of image-guided en-dovascular therapies for aSAH, includingcoiling of aneurysms and balloon angio-plasty or intraarterial drug delivery forcerebral vasospasm (1– 4). These ad-vances have occurred in a general settingof increasing knowledge of aSAH epide-miology, pathophysiology, diagnosis, andprevention and of significant refinementsin microsurgical technique and in medi-cal therapy. There is a widespread percep-tion that this broader understanding andexpertise is yielding benefits in the formof improved outcome after aSAH. Indeed,a progressive increase in aSAH survivalover the past three decades has been re-ported in several studies (5–7). However,evidence of a direct relationship betweenaSAH outcomes and a specific strategy orintervention is limited (2, 3, 8, 9). Inseveral key areas of management, sup-porting data are lacking or equivocal innature, generating uncertainty and con-troversy among clinicians.

This review focuses on controversiesthat are central to the acute managementof aSAH. These include surgical vs. endo-vascular aneurysm repair, the diagnosis

and management of cerebral vasospasm,neuroprotective strategies, use of anti-thrombotic agents (thrombolytic agents,heparin, and platelet inhibitors), prophy-laxis of seizures, and the approach to ce-rebral salt wasting and to cardiac dys-function after aSAH. For eachcontroversy, a critical evaluation of theavailable evidence is coupled with recom-mendations for further clinical investiga-tion. The review is selective, focusing onthe principal debates at the expense ofother equally important but arguably lesscontroversial issues (e.g., post-aSAH hy-drocephalus and ventricular drainage).

Controversy 1: Surgical vs.Endovascular Aneurysm Repair

Endovascular coiling emerged as analternative to surgery in patients withintracranial aneurysms who were deemedpoor surgical candidates due to signifi-cant neurologic injury, the presence ofsevere medical co-morbidities, or difficultsurgical access to the aneurysm (1). Morerecent work has sought to extend theindications of endovascular coiling toother patient categories. In a small ran-domized trial of 109 patients with aSAH,3- and 12-month clinical and neuropsy-chological outcomes were the same be-tween the surgical group and the endo-vascular group (10). This was followed by

*See also p. 571.From the Division of Neurosciences Critical Care,

Departments of Neurology, Anesthesiology/CriticalCare Medicine, and Neurological Surgery, Johns Hop-kins University School of Medicine, Baltimore, MD.

Supported, in part, by U.S. Public Health ServiceNational Institutes of Health NS NS046379 and by anEstablished Investigator Grant (A. Bhardwaj) from theAmerican Heart Association.

The authors have no financial interests to disclose.Copyright © 2006 by the Society of Critical Care

Medicine and Lippincott Williams & Wilkins

DOI: 10.1097/01.CCM.0000198331.45998.85

Background: The care of patients with aneurysmal subarach-noid hemorrhage has evolved significantly with the advent of newdiagnostic and therapeutic modalities. Although it is believed thatthese advances have contributed to improved outcomes, consid-erable uncertainty persists regarding key areas of management.

Objective: To review selected controversies in the manage-ment of aneurysmal subarachnoid hemorrhage, with a specialemphasis on endovascular vs. surgical techniques for securinganeurysms, the diagnosis and therapy of cerebral vasospasm,neuroprotection, antithrombotic and anticonvulsant agents, cere-bral salt wasting, and myocardial dysfunction, and to suggestvenues for further clinical investigation.

Data Source: Search of MEDLINE and Cochrane databases andmanual review of article bibliographies.

Data Synthesis and Conclusions: Many aspects of care inpatients with aneurysmal subarachnoid hemorrhage remainhighly controversial and warrant further resolution with hypoth-esis-driven clinical or translational research. It is anticipated thatthe rigorous evaluation and implementation of such data willprovide a basis for improvements in short- and long-term out-comes. (Crit Care Med 2006; 34:511–524)

KEY WORDS: aneurysm; subarachnoid; hemorrhage; vasospasm;ischemia

511Crit Care Med 2006 Vol. 34, No. 2

Page 2: Controversies in the management of aneurysmal subarachnoid ... · sible aneurysm. Medical management is based on the detection and treatment of cerebral and extracerebral complications

the International Subarachnoid Aneu-rysm Trial (ISAT), a multiple-center, ran-domized study of endovascular coiling vs.surgical clipping conducted in 2,143 pa-tients with aSAH who were deemed suit-able for either therapy. Posterior circula-tion aneurysms accounted for only 58 of2,143 patients (as many of these patientswere not enrolled because coiling wasconsidered the preferred modality oftreatment). At 1 yr, endovascular coilingwas associated with dependency or deathin 23.5% of patients compared with30.9% in the surgical group, a relativerisk reduction of 22.6% (p � .001). Ofconcern, however, nonprocedural re-bleeding within 1 yr was higher in pa-tients randomized to endovascular treat-ment (40 recurrent aSAHs, with 22deaths) compared with patients allocatedto neurosurgical treatment (33 patientswith aSAHs, 30-day mortality in 21 pa-tients) (2, 3).

Although widely regarded as a land-mark trial, the ISAT has been criticizedwith regard to biases in patient selection,low rates of randomization of eligible pa-tients, the definition of clinical equipoise,expertise of the neurosurgeons and inter-ventionists, the failure to use an opera-tive microscope, the higher than ex-pected morbidity in the surgically treatedgroup, the absence of angiographic dataafter the initial treatment, the lack oflong-term (�1 yr) follow-up, and the ap-propriateness of the reported outcome as-sessment scale (11, 12). An expert panelsuggested that 1-yr outcome as reportedin the ISAT is not an appropriate endpoint for the comparison of therapies,given that endovascular coiling is be-lieved to carry a risk of aneurysmal re-bleeding that extends beyond 1 yr. Indefense of ISAT, its authors observed thatoutcomes of surgically treated patients inISAT were comparable with those in aprospective, multiple-center North Amer-ican trial (13). They acknowledge that therate of randomization of eligible patientswas low (22.4%); however, they point outthat this rate was comparable with otherlarge randomized trials of vascular ther-apy such as the North American Symp-tomatic Carotid Endarterectomy Trial(14) or the Asymptomatic Carotid Athero-sclerosis Study (15), studies that havesignificantly influenced the treatment ofpatients with carotid artery stenosis.

A recent report from the ISAT trialgroup provides further crucial informa-tion regarding long-term outcomes (3).Based on 3,258 patient years of follow-up

after the first year for the endovasculargroup and 3,107 patient years of fol-low-up for the neurosurgical group, witha mean follow-up of 4 yrs, risk of recur-rent aSAH was higher in patients ran-domized to coiling (seven patients) com-pared with clipping (two patients), butmortality related to recurrent aSAH wasequal in both groups. A higher risk forseizures and poor cognitive outcomeswas seen in the surgical group, and cu-mulative 7-yr mortality curves showedmore deaths in the surgical group com-pared with coiling. The increased risk ofre-bleeding in the coiling group did notseem to reverse the early benefit seenwith this modality. Notwithstanding itslimitations, ISAT represents level I evi-dence that in this patient population, en-dovascular coiling is associated with bet-ter 1-yr outcomes, with trends towardsuperior long-term outcomes, whencompared with surgical clipping.

Apart from the recently published in-formation stemming from the ISAT trial,randomized trials comparing long-termoutcomes after coiling vs. clipping of rup-tured aneurysms are not available. How-ever, in an analysis of patients who un-derwent coiling of unruptured aneurysmsand were followed for a median of 22.3months, annual re-bleeding rates were0.8% in the first year, 0.6% in the secondyear, and 2.4% in the third year afterembolization, with no re-bleeding in sub-sequent years (16). In a study of 29 pa-tients with giant aneurysms treated withendovascular coiling and followed up fora median of 50 months, long-term clini-cal outcomes were good in 79% of pa-tients; however, the stability of the coilover time was poor, requiring repeat coil-ing, surgery, or parent-vessel occlusionin more than half of the aneurysms ini-tially treated with coils (17). Finally,Friedman et al. (18) reported clinical(mean, 19.1 months) and angiographic(mean, 11.6 months) data in 83 patientswith aSAHs treated with endovascularcoils. Neurologic outcome was good in77% of patients; however, 26% had a“dog-ear” remnant, 35% had a residualneck, and 3% had residual aneurysm fill-ing. Two or more coiling procedures wererequired in 34% of patients.

An important question concerns theinfluence of surgical or endovasculartreatment on the prevalence of cerebralischemic complications. In several non-randomized comparisons of surgery andendovascular therapy of ruptured aneu-rysms, the rate of symptomatic vaso-

spasm was either not significantly differ-ent (19–21) or slightly higher (22, 23)among surgically treated patients. Anearly nonrandomized study of 156 pa-tients had suggested a higher rate of ce-rebral infarction in patients receiving en-dovascular vs. surgical therapy; however,the proportion of patients with poor ini-tial neurologic presentation was higherin the endovascular group (24).

Comment. The available data indicatethat in patients with good neurologicgrade aSAH who undergo treatment foraneurysms in the anterior circulation,1-yr outcomes are clearly superior afterendovascular coiling when comparedwith surgical clipping. Follow-up of pa-tients receiving endovascular treatmentin some studies suggests that despitegood clinical results, a significant propor-tion of patients may require repeat endo-vascular or surgical therapy for residualor recurrent aneurysmal lesions. How-ever, long-term (�1 yr) clinical trends inpatients enrolled in ISAT do seem to sug-gest that endovascular coiling is likely toretain its advantage over clipping as asuperior procedure, and further fol-low-up of these patients is likely to addinsight to this debate. Preliminary resultsfrom various studies have failed to con-sistently show an association betweentreatment modality and the rate of de-layed cerebral ischemia. For posteriorcirculation aneurysms and for large, an-atomically complex or wide-necked aneu-rysms, further studies are needed to de-fine the best therapeutic approach.

Controversy 2: Diagnosis ofDelayed Cerebral Ischemia

A critical concern in the managementof patients with aSAH is the predictionand accurate diagnosis of delayed cere-bral ischemia because timely institutionof therapeutic interventions may preventthe occurrence of tissue infarction.Spasm of large arteries in the circle ofWillis is a significant determinant of ce-rebral ischemia after aSAH and carries a15–20% risk of stroke or death (25).Four-vessel cerebral digital subtractionangiography is the gold standard for di-agnosing vasospasm, but given the inher-ent risks and allocation of time and re-sources required for this procedure,alternative and complementary diagnos-tic tools have been proposed (26). Theseinclude transcranial Doppler (TCD), com-puted tomographic angiography (CTA),magnetic resonance imaging, radionu-

512 Crit Care Med 2006 Vol. 34, No. 2

Page 3: Controversies in the management of aneurysmal subarachnoid ... · sible aneurysm. Medical management is based on the detection and treatment of cerebral and extracerebral complications

clide imaging, cerebral microdialysis, andelectroencephalography.

TCD is an ultrasound-based monitorthat uses the principle that the velocity ofblood flow in an artery is proportional tothe ratio of flow to the luminal surfacearea of that vessel (27). TCD is a nonin-vasive, low-risk procedure that can bereadily performed at the bedside andlends itself to repeated (e.g., daily) obser-vations, enabling trend analysis. How-ever, there is debate about the correlationbetween increased TCD flow velocitiesand a) angiographic vasospasm and b)clinically significant or “symptomatic”vasospasm. Although mean middle cere-bral artery (MCA) cerebral blood flow(CBF) velocities of �200 cm/sec accu-rately predict angiographic vasospasm,velocities in the 120–200 cm/sec rangehave a far lower predictive value (28).Moreover, it has been observed that TCDis not as reliable in estimating distal MCAvasospasm compared with the more prox-imal portions of the MCA (29). Studiesshowing a significant moment-to-mo-ment variability during continuous mea-surement of CBF velocities have height-ened concern about the accuracy of thistechnique (30).

To address these limitations, severalrefinements have been proposed. Investi-gators have suggested that incrementalincreases in CBF velocities over time aremore helpful than velocities taken in iso-lation. One study indicated that an in-crease of �50 cm/sec in 24 hrs wasclosely associated with angiographic andclinical vasospasm (31). To overcome thenumerical dependence of flow velocitiesin the measurement of CBF, Lindegaardet al. (32) developed a “hemispheric in-dex” that normalizes flow velocity in theMCA to that in the ipsilateral extracranialinternal carotid artery (an index of �3 isstrongly predictive of angiographic vaso-spasm). Torbey et al. (33), observing thatangiographic vasospasm occurs at lowerTCD velocities in older patients, sug-gested that the accuracy of TCD can beenhanced by using an age-adjusted no-mogram based on a quadratic relation-ship between age and CBF velocities.Some investigators have proposed thatthe diagnostic capability of TCD may beenhanced by transcranial color-codedsonography. Transcranial color-codedsonography is an ultrasound-based neu-roimaging technique that allows real-time visualization of intracranial vascularstructures in addition to measurement ofCBF velocities. In a prospective study

comparing transcranial color-codedsonography and conventional TCD in theprediction of angiographic spasm, sensi-tivity and specificity of transcranial color-coded sonography for MCA and internalcarotid artery vasospasm were higherthan those for TCD (34). The validity ofTCD in diagnosing angiographic vaso-spasm was summarized in a recent sys-tematic review (35). For the MCA, sensi-tivity of TCD was 67% and specificity was99%, with a positive predictive value of97% and negative predictive value of78%. The accuracy of TCD was consider-ably less for detecting spasm in vesselsother than the MCA.

Interest has developed in CTA as adiagnostic modality for detecting cerebralvasospasm. CTA may be combined withperfusion computed tomography (CT), al-lowing characterization of both vascularanatomy and associated cerebral perfu-sion abnormalities. Using CTA, a positivepredictive value of 100% for angiographicvasospasm was reported in a recent smallseries (36). Another group reported thatCTA was highly accurate in detecting se-vere cerebral vasospasm in proximal ar-terial locations but was less accurate fordetecting mild and moderate spasm indistal locations (37). A third group notedan excellent concordance between the se-verity of vasospasm as determined by CTAand conventional angiography in bothproximal and distal arterial segments(38). Of note, none of these studies in-cluded �20 patients.

Although the phenomenon of cerebralvasospasm has historically elicited muchscientific interest, recent investigationshave turned to the related question ofbrain ischemia and infarction in patientswith aSAH. The predictors of CT-definedcerebral infarction after aSAH were ana-lyzed in a recent study. Multivariate anal-ysis revealed that both TCD and angio-graphic vasospasm were independentlyassociated with cerebral infarction, how-ever there was agreement between thetwo tests only in 73% of cases, and thevalidity of the two tests alone or in com-bination was disappointing (combinedsensitivity, 0.72; specificity, 0.68; positivepredictive value, 0.67; negative predictivevalue, 0.72) (39). These results are con-sistent with data suggesting that thepathophysiologic model of cerebral isch-emia after aSAH may need to considerfactors other than large artery spasm.

Magnetic resonance diffusion-weighted imaging accurately identifiesbrain tissue that is at high risk of infarc-

tion, whereas perfusion-weighted imag-ing reveals asymmetries in regional per-fusion. In a study of 14 patients withaSAH, diffusion-weighted imaging abnor-malities were noted in all patients withvasospasm suggested by TCD, whereas nosuch abnormalities were observed in pa-tients without abnormal TCD findings(40). A study of aSAH patients using per-fusion-weighted imaging revealed areasof hypoperfusion that correlated wellwith delayed ischemic neurologic deficits(DIND) and were larger than the areas ofdiffusion-weighted imaging abnormalityperformed at the same time. Of note,whereas all 15 patients with DINDshowed alterations in perfusion-weightedimaging, TCD evidence of vasospasm wasnoted in only seven of these patients (41).

Radionuclide-based studies of CBFand metabolism include single-photonemission computed tomography (SPECT)and positron emission tomography. In aseries of 129 patients with aSAH, Rajen-dran et al. (42) found that 89 had SPECTevidence of hypoperfusion, and this cor-related with TCD evidence of vasospasmin only 64% of cases. In another study,Jabre et al. (43) observed that the sensi-tivity of SPECT for symptomatic vaso-spasm was inferior to TCD; however,SPECT was more specific. Studies usingpositron emission tomography in pa-tients with aSAH suggest this techniquecan help differentiate neurologic deficitsdue to reversible ischemia or to irrevers-ible infarction (44); however, the accu-racy of positron emission tomography(45) in the diagnosis of vasospasm is un-known.

Monitoring of neurochemical markersof ischemia with cerebral microdialysishas been proposed as a technique for de-tecting vasospasm and delayed cerebralischemia. In a study of 97 patients withaSAH, neurochemical changes indicativeof ischemia were observed before the on-set of symptoms in 83% of patients withDIND (46). In another report, an isch-emic pattern of cerebral metabolites pre-ceded the occurrence of DIND by a meaninterval of 11 hrs (47). In a comparisonwith TCD and angiography, microdialysiswas found to have the highest specificityand likelihood ratio, but lower sensitivity,as a diagnostic tool for DIND (48). In astudy of 13 patients combining cerebralmicrodialysis and positron emission to-mography, it was noted that transientreductions in regional CBF correlatedwith elevations in extracellular glutamateand glycerol, whereas the lactate/pyru-

513Crit Care Med 2006 Vol. 34, No. 2

Page 4: Controversies in the management of aneurysmal subarachnoid ... · sible aneurysm. Medical management is based on the detection and treatment of cerebral and extracerebral complications

vate ratio was sensitive only after longerperiods of hypoperfusion (49). Althoughthese results are encouraging, several in-herent limitations of cerebral microdialy-sis have been recognized, including thedifficulty of extrapolating from measure-ments made in a very restricted volumeof tissue, the development of reactive gli-osis around the catheter tip decreasingthe accuracy of measurements, the inter-subject variability in basal neurochemicalvalues, and the tissue trauma after probeimplantation (50). The use of this tech-nique as a routine diagnostic method inpatients with aSAH was not supported ina recent systematic review (51).

Several groups have assessed the roleof electroencephalography in the diagno-sis of cerebral ischemia after aSAH. In astudy of 151 patients, Rivierez et al. (52)noted that that focal areas of slowingcorrelated with angiographic vasospasmin 96% of cases. Vespa et al. (53), usingcontinuous electroencephalography,found that relative alpha variability wasdecreased in 19 patients with angio-graphically proven vasospasm; in ten ofthese patients, the relative alpha variabil-ity change preceded clinical symptoms bynearly 3 days. Finally, Claassen et al. (54),in comparative-analysis quantitative elec-troencephalographic variables in 34 pa-tients with Hunt and Hess grade IV or VaSAH, determined that a decrease in al-pha to delta ratio (ADR) was stronglyassociated with delayed cerebral ischemiadefined by clinical or CT criteria. In thisreport, a 50% decrement in alpha to deltaratio has a sensitivity of 89% and a spec-ificity of 84% for delayed cerebral isch-emia.

Comment. The prediction and accu-rate diagnosis of cerebral ischemia is acardinal goal in the critical care of pa-tients with aSAH. Using serial TCD andtranscranial color-coded sonography, in-cremental changes in flow velocities andcalculation of the hemispheric index mayprovide valuable information with regardto underlying angiographic vasospasm, inparticular, vasospasm involving the MCA.The diagnostic capability of CTA is un-clear and needs investigation in a pro-spective study. Recent studies haveshifted away from the characterization ofvessel lumen diameter to the detection ofchanges in CBF and of cerebral ischemia.Perfusion CT, diffusion- and perfusion-weighted magnetic resonance imaging,radionuclide-based perfusion studies, ce-rebral microdialysis, and electroencepha-lography hold promise as techniques that

might expand the therapeutic window fortreating ischemia in patients with aSAH;however, their introduction into clinicalpractice needs further confirmation inclinical trials.

Controversy 3: Management ofDelayed Cerebral Ischemia

The principal options for treating de-layed cerebral ischemia are hemody-namic augmentation and endovasculartherapy. Alternative therapies include in-traaortic balloon counterpulsation(IABC), therapeutic hypothermia, andbarbiturate coma.

The concept of hemodynamic aug-mentation—also referred to as hyperten-sion, hypervolemia, hemodilution or tri-ple-H therapy—in patients with aSAHevolved out of two important and (andrelated) observations. The first, suggestedin studies from the 1950s and 1960s, wasthe strong association between cardiovas-cular variables, such as intravascular vol-ume status, cardiac output, and bloodpressure in the days and weeks ensuingaSAH, and clinical outcomes, such assymptomatic vasospasm and long-termneurologic function. The second observa-tion was that cerebral vasospasm is char-acterized by a shift in cerebrovascularresistance away from the penetrating ar-terioles to the major branches of the cir-cle of Willis and their proximal branches,vessels that are incapable of effective au-toregulation. As a result, CBF becomespassively dependent on systemic bloodpressure, greatly increasing the risk ofcerebral ischemia. Using the Poiseuillerelationship, it can be predicted that ef-forts to increase systemic blood pressureor to decrease blood viscosity will ame-liorate cerebral perfusion and reverseischemia (55).

In an early case series of patients withDIND, Kosnik and Hunt noted significantimprovements in neurologic function af-ter initiating therapy with phenylephrineand colloid fluid expansion (56). This wasfollowed by several other uncontrolledstudies further substantiating the clinicalefficacy of hemodynamic augmentation(57–60). Randomized studies evaluatingthe efficacy of hemodynamic augmenta-tion therapy in aSAH are limited in num-ber. In 30 aSAH patients who were beingstudied before they underwent aneurysmclipping, Rosenwasser et al. (61) notedthat a strategy of blood pressure controlwith vasodilators in addition to volumeexpansion with packed red blood cell

transfusion and albumin was associatedwith a significantly lower rate of symp-tomatic vasospasm and death when com-pared with a strategy of blood pressurecontrol with diuretics. Lennihan et al.(62) randomly assigned 82 aSAH patientson the day after aneurysm clipping toreceive albumin fluid boluses titrated tonormal or high cardiac filling pressures(central venous and pulmonary artery di-astolic pressures). They discovered thathigher filling pressures were not associ-ated with any significant change in CBF(as measured by xenon CT) or blood vol-ume, nor were there any differences inthe rate of symptomatic vasospasm, cere-bral infarction, or 3-month Glasgow Out-come Scale. Finally, in a study of 32 aSAHpatients randomized to hypertensive/hypervolemic vs. normotensive/normo-volemic management protocols, Egge etal. (63) reported no difference in vaso-spasm rates, CBF as measured by SPECT,or 1-yr Glasgow Outcome Scale, whereasa higher rate of complications (hemor-rhage, coagulopathy, congestive heartfailure) was noted in the hypertensive/hypervolemic group. When the results ofthese three trials were pooled in a sys-tematic review, no significant effect ofprophylactic triple-H therapy on the rateof symptomatic vasospasm, DIND, ordeath was noted (64). A Cochrane meta-analysis reached a similar conclusion (9).

Triple-H therapy has many inherentlimitations, notably a) the association ofhemodynamic augmentation with severecomplications such as congestive heartfailure, noncardiogenic pulmonaryedema, myocardial ischemia, intracranialhemorrhage, global cerebral edema, anddeath; b) failure to reverse neurologicdeterioration in certain patients; and c)contraindications to its use such as thepresence of significant preexisting or ac-quired cardiopulmonary dysfunction. Al-ternative approaches to treating vaso-spasm have emerged, including a) astrategy targeting cardiac output ratherthan mean arterial pressure as a physio-logic end point, b) the endovascular ther-apies, including balloon angioplasty andintraarterial vasodilator administration,and c) the use of IABC.

To augment cardiac output, the ino-tropic agent dobutamine was adminis-tered in combination with hypervolemicpreload enhancement to 23 patients withvasospasm whose neurologic examina-tion failed to improve after preload en-hancement alone. The authors noted a52% increase in cardiac index, and clini-

514 Crit Care Med 2006 Vol. 34, No. 2

Page 5: Controversies in the management of aneurysmal subarachnoid ... · sible aneurysm. Medical management is based on the detection and treatment of cerebral and extracerebral complications

cal reversal of ischemic symptoms wasevident in 18 of the 23 patients (65). In amore recent study, xenon CT was used toevaluate CBF in 16 patients with symp-tomatic vasospasm who underwent vol-ume expansion combined with eithermean arterial pressure augmentationwith phenylephrine or cardiac outputaugmentation with dobutamine. The in-crease in mean CBF was similar in bothgroups (66). The mechanism whereby in-creases in cardiac output withoutchanges in mean arterial pressure affectCBF is unclear, and this phenomenonmay be unique to the setting of vaso-spasm, as a relationship between cardiacoutput and CBF was not observed in pa-tients with traumatic brain injury (67). Ithas been postulated that the wider pulsepressure and enhanced pulsatile flow as-sociated with the administration of ino-tropic agents may ameliorate flowthrough collateral vessels and throughthe microvasculature (68).

Recent years have seen a significantdevelopment in the use of endovasculartherapies for patients with, or at risk of,cerebral vasospasm (69). Endovasculartreatments include transluminal balloonangioplasty (TBA) and the intraarterialdelivery of vasodilating compounds.These techniques are most commonlyused in patients with symptomatic vaso-spasm that has been resistant to triple-Htherapy. TBA is highly effective in reliev-ing focal spasm involving proximal seg-ments of the circle of Willis; however,when vasospasm is diffuse or more distal,the selective intraarterial infusion of va-sodilators may be helpful. Clinical im-provement after angioplasty is well doc-umented and is generally durable,whereas the response to intraarterial va-sodilators is transitory. Complications ofTBA include trauma to the arterial wallleading to dissection, rupture, andthrombosis, with consequent cerebral in-farction or hemorrhage; in addition,reperfusion of cerebral tissue in whichinfarction has already occurred may in-duce edema and hemorrhage. The mostcommonly administered cerebral intraar-terial vasodilator is papaverine. However,papaverine is neurotoxic and has beenlinked to seizures, coma, blindness, andirreversible cortical injury (70). Vera-pamil (71), nimodipine (72), and nicardi-pine (73) have been used as alternativeintraarterial cerebral vasodilators; how-ever, data on their efficacy and safety islargely anecdotal.

Although the efficacy of TBA in treat-ing vasospasm is well documented, inter-est has developed on the preventive use ofthis technique in high-risk patients.Based on results from an animal modelshowing that TBA performed on the dayof the hemorrhage prevented the devel-opment of angiographic spasm on day 7after aSAH, Muizelaar et al. (74) evalu-ated prophylactic TBA in 13 patients withFisher grade III aSAH who had a highprobability of developing vasospasm. Ofthese patients, none developed DIND ormore than mild TCD-defined vasospasm.At 3 months posttreatment, eight pa-tients had a good recovery, two weremoderately disabled, and three had died(one because of a vessel rupture duringTBA). These authors are conducting arandomized trial (Balloon Prophylaxis ofAneurysmal Vasospasm trial; see http://www.strokecenter.org/trials for details)to determine whether the efficacy of pro-phylactic TBA is sufficient to justify therisks and to clarify which vessels need tobe dilated prophylactically

In recent years, several groups havereported on the use of IABC in patientswith delayed cerebral ischemia (75–79).IABC has been associated with reversal ofneurologic deficits and significant in-creases in CBF in patients who did notrespond to conventional triple-H therapy(79) or who had cardiopulmonary dys-function contraindicating it (75, 76). Ofnote, despite the invasive nature of thistechnique, the prevalence of severe ad-verse effects was remarkably low, and arecent series evaluated the prophylacticplacement use of IABC in six patients whowere considered at high risk for cerebralvasospasm (77). As with pharmacologiccardiac output augmentation, the benefi-cial effects of IABC are not well under-stood and might reflect augmentation ofdiastolic perfusion through the carotidand vertebral arteries and heightenedpulsatility, which has been associatedwith improved microvascular blood flow(78).

Comment. Hypovolemia and hypoten-sion after aSAH are strongly linked toadverse outcome and should be avoidedin all patients. The existing data do notsupport the prophylactic use of triple-Htherapy in patients with aSAH who do nothave clinical evidence of vasospasm. Inpatients with symptomatic vasospasm,hemodynamic augmentation may reverseneurologic deterioration; however, an ad-equately powered randomized trial isneeded to test the hypothesis that tri-

ple-H therapy has a favorable effect onneurologic outcomes or survival and issafe when compared with a strategy ofnormovolemia and normotension. Fur-ther study is needed to better understandthe effects of increased cardiac output, asopposed to hypertensive therapy, on CBFand on the reversal of symptomatic vaso-spasm. At this time, TBA and intraarterialvasodilators administration are reason-able options in treating vasospasm refrac-tory to medical management. However,the relative efficacy and harm of TBA vs.medical management needs further sub-stantiation. This might take the form of arandomized trial comparing immediateangioplasty vs. triple-H therapy in pa-tients who have developed symptomaticvasospasm. In patients with symptomaticvasospasm in whom triple-H therapy andendovascular options have either failed orare contraindicated, consideration shouldbe given to IABC or to neuroprotectiveinterventions such therapeutic hypother-mia and pentobarbital coma (see below).

Controversy 4: CerebralProtection

The development of cerebral ischemiccomplications in a significant proportionof patients after aSAH has prompted greatinterest in the possibility of preventing orlimiting irreversible brain injury. Therisk of a cerebral ischemic event is par-ticularly high during specific events as-sociated with aSAH, namely a) the initialaneurysmal rupture (risk of global cere-bral ischemia secondary to increased in-tracranial pressure, hypotension, and hy-poxemia), b) the procedure undertakenfor securing the aneurysm (risk of strokein relation to surgical clipping or endo-vascular coiling), and c) delayed cerebralischemia and vasospasm. Cerebral pro-tective strategies that have been studiedin these settings include a) pharmaco-logic agents with cytoprotective or vaso-dilatory properties, b) therapeutic hypo-thermia, and c) hemodynamicaugmentation or endovascular therapies,which are discussed above. Pharmaco-logic cerebral protectants that have beentested in clinical studies of aSAH includecalcium channel antagonists, tirilazadmesylate, glucocorticoids, magnesium,endothelin receptor antagonists, and hy-droxymethylglutaryl coenzyme A reduc-tase inhibitors.

A meta-analysis of trials using calciumchannel antagonists after aSAH was re-ported by the Cochrane group (8). Of 11

515Crit Care Med 2006 Vol. 34, No. 2

Page 6: Controversies in the management of aneurysmal subarachnoid ... · sible aneurysm. Medical management is based on the detection and treatment of cerebral and extracerebral complications

studies (2,804 patients), eight involvednimodipine (1,574 patients), two involvednicardipine (954 patients), and one in-volved AT877 (276 patients). When com-pared with placebo, calcium channel an-tagonists were associated with asignificantly reduced risk of poor out-come, with a relative risk (RR) of 0.82(95% confidence interval [CI], 0.72–0.93), an absolute risk reduction of 5.1%,and numbers needed to treat of 20. Theresults were most robust for oral nimo-dipine (RR of poor outcome, 0.70; 95%CI, 0.58–0.84). The RR of death in pa-tients treated with calcium antagonistswas 0.94 (95% CI, 0.80–1.10), that ofischemic neurologic deficits was 0.67(95% CI, 0.59–0.76), and that of CT orMR documented cerebral infarction was0.80 (95% CI, 0.71–0.89). Of note, theprevalence of angiographic vasospasmwas not influenced by treatment alloca-tion, suggesting that the benefit of nimo-dipine was linked to its cytoprotectiverather than its cerebral vasodilatory prop-erties. The effects of nimodipine on aSAHoutcome are fairly consistent across pub-lished trials. However, it is plausible thatthe results were confounded because ni-modipine-treated patients may have re-ceived greater amounts of intravenousfluids to counteract the effect of the drugon systemic blood pressure, surrepti-tiously exposing them to a more aggres-sive regimen of triple-H therapy.

Tirilazad mesylate, a non–glucocorti-coid 21-aminosteroid that inhibits lipidperoxidation, has been evaluated in fourrandomized, placebo-controlled trials ofpatients with aSAH. The first trial, con-ducted in Europe and Australasia, dem-onstrated an improvement in symptom-atic (but not angiographic) vasospasm,3-month survival, and Glasgow OutcomeScale in patients receiving 6 mg/kg tiril-azad for 10 days (80). These results werenot reproduced in a second study under-taken in North America in which tirilazadadministration had no significant effecton symptomatic vasospasm, 3-monthmortality, or Glasgow Outcome Scale(13). The divergence in these results wasbelieved to reflect differences in manage-ment protocols, including the widespreaduse of anticonvulsants (e.g., phenytoin)in the American trial, which may havediminished the bioavailability of tirilazad.Further analysis of these two studies in-dicated that any detected improvement inoutcome seemed limited to men. This ledto two other studies conducted in womenonly. The first of these demonstrated a

rate of symptomatic vasospasm that wassignificantly lower in women who re-ceived tirilazad, 15 mg/kg, without anyeffect on mortality (81). In the other trial,the rate of symptomatic vasospasm wasnot affected by tirilazad therapy; however,mortality was significantly lower intreated patients with Hunt and Hessgrades IV and V (82). When results of thefour trials were combined in a meta-analysis, it was concluded that overallmortality was decreased in patients whoreceived tirilazad, with an effect thatseemed most pronounced in patientswith poor neurologic grade (83).

Animal studies have shown that glu-cocorticoids may effectively prevent de-layed cerebral ischemia after aSAH (84–86). The potential benefit ofglucocorticoids in humans was suggestedin a nonrandomized comparative study of21 patients by Chyatte et al. in whichpatients receiving high-dose methylpred-nisolone had a significantly lower rate ofDIND (87). However, these results havenot been reproduced in any other clinicaltrial.

The neuroprotective effects of magne-sium have been reported in experimentalmodels of traumatic brain injury, cere-bral ischemia, and aSAH. Magnesium sul-fate therapy is both safe and effective inpreventing neurologic complications inobstetric patients with preeclampsia. Arandomized trial of 40 patients demon-strated that high-dose intravenous mag-nesium sulfate is safe in patients withaSAH (88). In a more recent randomized,placebo-controlled trial of 283 patients,an infusion of magnesium started within4 days of aSAH and continued up to 14days postaneurysm occlusion was associ-ated with a 34% RR reduction for theprimary outcome of delayed cerebralischemia, however, this result did notachieve statistical significance (89). Ofnote, significantly more patients treatedwith magnesium had excellent outcome(defined as a Rankin score of 0) at 3months (RR, 0.91; 95% CI, 0.84–0.98).

Following experimental evidence im-plicating the vasoconstrictor endothelinpeptides and their receptors in the patho-physiology of cerebral vasospasm, a ran-domized, placebo-controlled trial of anendothelin receptor antagonist, TAK-044,was conducted in 412 patients with aSAH(90). A nonsignificant decrease in the rateof delayed cerebral ischemia was noted inthe treated group. A more recent obser-vational study suggested that increasedCSF levels of endothelin did not correlate

with vasospasm but were a more generalmarker of neuronal damage (91).

Initially developed as cholesterol-lowering agents, the hydroxymethylglu-taryl coenzyme A reductase inhibitors orstatins are capable of modulating endo-thelial function by reducing vascular in-flammation, inhibiting vascular smoothmuscle cell proliferation, decreasingplatelet aggregation, and promoting NO-mediated cerebral vasodilation (92). In amurine model of aSAH, pretreatmentwith simvastatin was associated with areduction in vasospasm and with in-creased expression of NO synthetase (93).A retrospective review of 60 aSAH pa-tients suggested that patients who weretaking statins, when compared with acontrol group who were not, had a sig-nificantly lower rate of DIND and cerebralinfarctions of any type, but no impact onmortality or global outcome (modifiedRankin scale) was detected (94). Anotherretrospective study showed an increasedrisk of vasospasm associated with use ofstatins (95). The effects of statins havebeen further explored in two recent ran-domized, placebo-controlled trials. In thefirst, therapy with simvastatin (19 pa-tients) initiated within 48 hrs of aSAHwas linked to reduced serum levels ofbrain injury biomarkers and a decrease inthe prevalence of DIND confirmed byTCD or angiography when compared withplacebo (20 patients) (96). In the second,treatment with pravastatin started within72 hrs of aSAH ameliorated cerebral va-sospasm, improved cerebral autoregula-tion, and reduced vasospasm-relatedDIND by 83% and mortality by 75% (97).Additional agents that have generated in-terest as cerebral protectants after aSAHinclude erythropoietin (98), nitric oxidedonors (99), and potassium channel acti-vators (100); to our knowledge, there areno published clinical trials of these com-pounds in patients with aSAH.

The neuroprotective effect of hypo-thermia is supported by a large body ofexperimental and clinical evidence. Thiseffect may result from decreased excita-tory amino acid release and free-radicalproduction, reduced intracellular cal-cium accumulation, stabilization of theblood–brain barrier, and decreased cere-bral edema (101). Hypothermia also hasserious deleterious consequences, includ-ing cardiovascular depression, immunesuppression, coagulopathy, and electro-lyte abnormality. The use of moderatehypothermia (28–32°C) to protect thebrain during cerebral aneurysm surgery

516 Crit Care Med 2006 Vol. 34, No. 2

Page 7: Controversies in the management of aneurysmal subarachnoid ... · sible aneurysm. Medical management is based on the detection and treatment of cerebral and extracerebral complications

was first described in the 1950s; however,interest in this technique declined inlight of very poor clinical outcomes. Morerecently, several studies have evaluatedmild hypothermia (38–35°C) as a protec-tive strategy during aneurysm surgery. Inan early randomized trial, 114 patientswith ruptured and unruptured intracra-nial aneurysms who received mild intra-operative hypothermia had a lower rate ofneurologic deterioration at 24 and 72 hrsafter surgery, a greater frequency of dis-charge to home, and a greater rate ofgood long-term outcomes (102). How-ever, a subsequent multiple-center studyof 1,001 patients with aSAH who hadWorld Federation of Neurologic Surgeonsscores of I, II, or III failed to detect anybeneficial effect of intraoperative coolingon 3-month neurologic outcomes or onany perioperative outcomes (103). Mov-ing beyond the perioperative setting,small-scale investigations indicate thatmild hypothermia may be helpful in casesin which vasospasm is refractory to con-ventional treatment (104, 105). The ef-fects of hypothermia on the relationshipbetween CBF and cerebral metabolic ox-ygen demand in patients with aSAH arepoorly understood. Hypothermia was as-sociated with evidence of brain ischemiain one study (106), but another study didnot confirm this finding (107).

Comment. Although considerable re-search has focused on the possibility ofneuroprotection in patients with aSAH,only oral nimodipine has been clearly as-sociated with improved outcome. Meta-analysis suggests a survival benefit withhigh-dose tirilazad mesylate in patientswith poor neurologic grade. Preliminaryresults with magnesium and statins needsubstantiation in larger randomized pla-cebo-controlled trials. Finally, no benefitwas seen for intraoperative mild hypo-thermia in aSAH patients with good neu-rologic grade; however, further studiesare needed to explore other methodolo-gies and applications of therapeutic hypo-thermia, for example, more prolongedcooling protocols and its effects on pa-tients with poor neurologic grade or re-fractory vasospasm.

Controversy 5: AntithromboticAgents

Although the conventional view of an-eurysmal rupture emphasizes the patho-physiologic role of hemorrhage and theneed to prevent recurrent intracranialbleeding, data have accrued to suggest

that clot formation, both in the intravas-cular and extravascular compartments, isa key determinant of cerebral injury afteraSAH. The presence of a thick clot in thesubarachnoid space is consistently re-ported as one of the strongest predictorsof vasospasm after aSAH (108). The poorneuroanatomic correlation between ra-diologically defined vasospasm and cere-bral infarction suggests alternate or com-plementary hypotheses of DIND, amongwhich intravascular thrombosis is a pos-tulated mechanism.

Thorombolytics. Experimental andclinical evidence indicate that a sub-arachnoid clot may be removed with thehelp of thrombolytic agents. Cisternal ir-rigation with recombinant tissue plas-minogen activator was found to be safe(109), and intrathecal urokinase infusionhas been associated with a decreased rateof vasospasm and permanent neurologicdeficits (110). A systematic review of ninestudies (only one of which was random-ized) noted that cisternal thrombolysiswas associated with an absolute risk re-duction of 14.4% for DIND (p � .001),9.5% for poor Glasgow Outcome Scalescores (p � .01), and 4.5% for death (p �.05) (111). Treatment effects did not sig-nificantly differ among the studies on thebasis of the type of thrombolytic agentused (recombinant tissue plasminogenactivator vs. urokinase) or the method ofadministration (intraoperative vs. postop-erative). Studies that enrolled only pa-tients at high risk for vasospasm seemedto demonstrate greater treatment effects(109). In an intriguing recent trial,Hamada et al. (110) randomized 110 pa-tients to endovascular coiling with orwithout infusion of urokinase into thesubarachnoid space via a microcatheterinserted in the lumbar space. The rate ofsymptomatic vasospasm was 8.8% in theurokinase-treated group vs. 30.2% in theuntreated group (p � .012).

Anticoagulants. Heparin, an anti-thrombin III agonist, has biological prop-erties that include anticoagulation, modu-lation of inflammation, neuroprotection,and antiproliferative effects. In an animalmodel of aSAH, administration of heparinhas been associated with smooth musclerelaxation, increased CBF, and a reductionin proliferative angiopathy (112). A recentrandomized, placebo-controlled trial thatenrolled 120 patients indicated that low-molecular-weight heparin significantly re-duced the rate of vasospasm-related cere-bral infarction (3.5% of enoxaparin-treatedpatients and 28.3% of placebo-treated pa-

tients, p � .001), without any increase inthe rate of intracranial bleeding (113).There was a potential bias because patientsin the placebo group were more severelyaffected (worse Hunt and Hess grade). Ofnote, another larger randomized trial of170 patients suggested that enoxaparin hadno effect on the outcome of aSAH andseemed to increase the risk of intracranialhemorrhage (114).

Antiplatelet Agents. Platelet aggrega-tion and release of thromboxanes havebeen demonstrated after aSAH, and thesechanges are more pronounced in patientswho develop DIND (115). These data, incombination with the identification ofmicroembolic signals by TCD in patientswith aSAH (116), have prompted severalsmall clinical trials examining the thera-peutic potential of antiplatelet agents.Meta-analysis of these results indicatedthat in patients treated with aspirin orantiplatelet agents, the RR of DIND wassignificantly reduced (RR, 0.65; 95% CI,0.47–0.89), without a concomitant in-crease in the risk of intracranial hemor-rhage. There was also a trend toward areduced risk of poor outcome in patientsreceiving antiplatelet agents (RR, 0.87;95% CI, 0.65–1.17) (117).

Comment. Preliminary data indicatethat placement of thrombolytic agents inthe subarachnoid space may improve out-come after aSAH, but adequately pow-ered, randomized, controlled studies areneeded to substantiate these findings.The two randomized trials assessing theassociation between low-molecular-weight heparin and aSAH outcome arecontradictory. Regarding antiplatelettherapy, although a beneficial effect issuggested, the evidence does not supportthe routine use of these agents in aSAH atthe present time. Randomized trials withgreater power would be needed to clarifythe role of these therapies.

Controversy 6: SeizureProphylaxis

Aneurysmal SAH has been linked to anincreased risk of seizures, yet the role ofseizure prophylaxis is controversial. In aretrospective analysis of 217 surgicallytreated patients with aSAH over a 2-yrperiod, 20% experienced one or more sei-zures, with more than half of these oc-curring in the perioperative period; ofnote, the occurrence of perioperative sei-zures did not predict epilepsy in thisstudy (118). Another investigation re-vealed that 24 of 121 aSAH patients with

517Crit Care Med 2006 Vol. 34, No. 2

Page 8: Controversies in the management of aneurysmal subarachnoid ... · sible aneurysm. Medical management is based on the detection and treatment of cerebral and extracerebral complications

Table 1. Summary of controversies in the management of aneurysmal subarachnoid hemorrhage (SAH)

Controversy Best Available Evidence and Recommendations Future Studies

Surgical vs. endovascular aneurysmexclusion

Level I evidence in favor of endovascularmanagement (grade A)

Comparative long-term follow-up of endovascular vs.surgical patients. New randomized trials to testendovascular vs. surgical therapy in patient subsetsthat were not represented in the ISAT trial.

Diagnosis of vasospasmTranscranial Doppler (TCD) Level III evidence for high PPV and specificity of

TCD for MCA (grade C)Prospective study comparing predictive value of CTA

and TCD with angiography in detecting vasospasm.Computerized tomography angiography

(CTA)Level III comparing CTA to angiography and TCD

(grade C)Observational study to compare CTA/CT perfusion,

perfusion/diffusion weighted MRI, and conventionalangiography in detecting clinically significant vasospasm.

MRI, PET, SPECT Level V evidence (grade C)Cerebral microdialysis Level V evidence Prospective clinical studies to understand the

correlation between neurochemical abnormalities andclinical events in SAH. Outcome-based trials to assessthe effect of microdialysis-guided management.

Treatment of vasospasmVasospasm prophylaxis Level II evidence showing no effect of triple-H

prophylaxis on DIND (grade B)Prospective randomized study comparing outcomes in

patients in vasospasm receiving either triple-Htherapy or undergoing immediate endovascularintervention. Prospective studies to identify high-risk patients who may be candidates forprophylactic management of vasospasm.

Vasospasm treatment Level III evidence supporting use of hemodynamicaugmentation as treatment of vasospasm (gradeC)

Adequately powered prospective, randomized trialevaluating hemodynamic augmentation vs.conventional hemodynamic goals in symptomaticvasospasm. Prospective, randomized trials tocompare a strategy of cardiac output augmentationvs. arterial pressure augmentation.

Hemodynamic end points Level IV evidence for use of cardiac output goalsover arterial pressure goals for hemodynamicaugmentation (grade C)

Intraaortic balloon counterpulsation (IABC) Level IV evidence for use of IABCNeuroprotection

Ca� channel blockers Level I evidence favoring use of nimodipine andagainst use of nicardipine and AT877 (grade A)

Search for alternative neuroprotectants based onanimal studies for possible synergy with orsuperiority to nimodipine.

Tirilazad mesylate Level II evidence in favor of tirilazad for highgrade SAH (grade A)

Glucocorticoids, magnesium,endothelin receptor antagonists, andhydroxymethylglutaryl coenzyme Areductase inhibitors

Level II evidence for statins, magnesium (grade B) Randomized, placebo-controlled study of these agentsin patients with aneurysmal SAH; largerrandomized trials evaluating statins and magnesiumin SAH.

Level V evidence favoring use of other agents(grade C)

Hypothermia Level I evidence showing no benefit ofintraoperative hypothermia (grade A); use inother settings unclear

Prospective, randomized studies to define the role ofhypothermia in patients at high risk to developvasospasm (prophylaxis) or in patients with clinicalvasospasm.

Thrombolytics Level IIa evidence for use of thrombolytics (gradeB)

Prospective, randomized trials with sufficientstatistical power to detect the efficacy of antiplateletagents, anticoagulants, and intraoperativethrombolytics with appropriate end points(recurrent hemorrhage, DIND, mortality, andmorbidity/functional outcome).

AnticoagulationAntiplatelet agents Level IIb evidence in favor of use of antiplatelet

agents (grade B)Seizure prophylaxis Level V evidence in favor of prophylaxis Randomized, placebo-controlled studies of

anticonvulsants in aneurysmal SAH with riskstratification based on grade of SAH, location ofaneurysm, and surgical intervention (craniotomy).

Level III evidence against prophylaxis (grade C)Cardiac sequelae of SAH

Myocardial dysfunction Level III evidence in favor of adrenergic receptorblockade

Level IV evidence in favor of inotropic support or IABC

Prospective, observational studies to identifypathophysiology, diagnosis, management, and outcomeof neurocardiogenic injury and to distinguish from otherforms of myocardial dysfunction.

ISAT, International Subarachnoid Aneurysm Trial; MRI, magnetic resonance imaging; PET, positron emission tomography; SPECT, single-photonemission computed tomography; PPV, positive predictive value; MCA, middle cerebral artery; CT, computerized tomography; triple-H, hypertension/hypervolemia/hemodilution therapy; DIND, delayed ischemic neurologic deficits.

aIntervention decreased DIND and improved neurologic outcomes but not mortality rates; bintervention decreased DIND but did not improve neurologicoutcome.

518 Crit Care Med 2006 Vol. 34, No. 2

Page 9: Controversies in the management of aneurysmal subarachnoid ... · sible aneurysm. Medical management is based on the detection and treatment of cerebral and extracerebral complications

clipped aneurysms had at least one sei-zure and that ten patients had two ormore seizures after hospital discharge(119). Seizure risk has been linked to thethickness of the aSAH clot (120), to loca-tion of the aneurysm on the MCA (121),to the presence of a subdural hematoma,and to cerebral infarction (122). In onestudy, variables associated with the devel-opment of epilepsy were, in order of im-portance, a history of hypertension, cere-bral infarction, and duration of impairedconsciousness after the seizure (123).

Although empirical prophylaxis of sei-zures after aSAH might seem reasonable,the benefit of preventive anticonvulsantadministration has yet to be demon-strated in a prospective randomized trial.A cohort study of 123 aSAH patients fol-lowed up at 4 to 7 yrs found no evidencefor the effectiveness of prophylactic anti-convulsants (119). A recent retrospectiveevaluation of 527 patients with aSAH in-dicated a strong association between phe-nytoin exposure in the setting and func-tional and cognitive disability (124). In astudy of 101 patients with aSAH who hadunexplained coma or neurologic deterio-ration, eight (all of whom were receivingprophylactic anticonvulsants) were foundto be in nonconvulsive status epilepticus.Despite successful termination of sei-zures in five of eight patients, all eightpatients eventually died after a period ofprolonged coma (125).

Comment. The available evidence sug-gests that prophylaxis of seizures may beuseful in aSAH patients with stroke orother distinct focal pathology. Indiscrim-inate administration of anticonvulsantsto patients with aSAH has been linkedwith unfavorable functional and cognitiveoutcomes. A randomized, placebo-con-trolled trial is warranted to assess theeffect of anticonvulsant prophylaxis onthe prevalence of early and late seizuresin patients with aSAH. Such a trial wouldideally incorporate predefined stratifica-

tion based on focal parenchymal pathol-ogy, aneurysm location, aSAH grade, age,and history of hypertension.

Controversy 7: Cerebral SaltWasting Syndrome

Hyponatremia occurs in up to 30% ofpatients after aSAH and has been associatedwith several disorders, most notably cere-bral salt wasting (CSW) syndrome and thesyndrome of inappropriate antidiuretichormone (SIADH) secretion (126). CSW in-volves renal salt loss leading to a negativesodium balance, hyponatremia, and intra-vascular volume depletion, whereas SIADHis characterized by an inability to appropri-ately excrete free water, resulting in a euv-olemic or hypervolemic state. Becausetherapeutic interventions for CSW and SI-ADH are radically opposed, clinical differ-entiation between the two entities is essen-tial. It has been suggested thathypouricemia and an increased fractionalexcretion of uric acid are more consistentwith a diagnosis of CSW (127). However,physiologic indicators of intravascular vol-ume are generally viewed as the most reli-able way to distinguish between these con-ditions (128).

The pathogenesis of CSW is poorly un-derstood (129). The main pathologic pro-cesses that have been linked with thisderangement are a) decreased sympa-thetic input to the kidney leading to adeficient regulation of proximal tubulesodium resorption and to an inadequaterise in renin and aldosterone in responseto hypovolemia, and b) increased levels ofcirculating natriuretic peptides. The na-triuretic peptides are produced in theheart, brain, and endothelium and pro-mote vasodilation, sodium excretion, anddiuresis. Several groups have demon-strated an association between aSAH, hy-ponatremia, volume depletion, and in-creased blood natriuretic peptide levels,in particular B-type natriuretic peptide

(BNP) (130–133). Others have observed alink between increased levels of BNP andthe development of cerebral vasospasm(134, 135). Recent work suggests thatcardiac dysfunction (136) and triple-Htherapy (137) are factors that might stim-ulate BNP release after aSAH. When theseresults are considered collectively, it re-mains unclear whether the relationshipbetween increased BNP levels and the de-velopment of CSW or vasospasm is caus-ative or merely circumstantial.

Therapeutic options for CSW are lim-ited. It has been argued that increasingsalt intake during CSW only further en-hances sodium excretion. The mineralo-corticoid fludrocortisone, which acts di-rectly on the kidney tubules to enhancesodium resorption, has been shown toprevent intravascular volume depletionin patients with aSAH (138). In a relatedstudy, Hasan et al. (139) found that 0.2mg of fludrocortisone given intrave-nously or orally twice a day in 46 patientswith aSAH significantly reduced the fre-quency of a negative sodium balance andled to smaller decreases in plasma vol-ume and, consequently, less risk of cere-bral ischemia. More recently, Moro et al.(140) demonstrated that hydrocortisoneattenuated excessive natriuresis in thesetting of aSAH with development of hy-ponatremia in 43% of treated patients vs.0% in the untreated group.

Comment. Although the pathogenesisof CSW needs clarification, its conse-quences of hyponatremia and intravascu-lar volume depletion can be deleteriousin the setting of aSAH. Preliminary datasupport the careful use of salt-conservingmedications such as fludrocortisone orhydrocortisone in patients with aSAH.

Controversy 8: Management ofCardiac Dysfunction After aSAH

A spectrum of cardiac sequelae havebeen described in patients after aSAH,

Table 2. Levels of evidence and strength of recommendation

Level of evidenceLevel I Data from randomized trials with low false-positive (�) and low false-negative (�) errorsLevel II Data from randomized trials with high false-positive (�) or high false-negative (�) errorsLevel III Data from nonrandomized concurrent cohort studiesLevel IV Data from nonrandomized cohort studies using historical controlsLevel V Data from anecdotal case series

Strength of recommendationGrade A Supported by level I evidenceGrade B Supported by level II evidenceGrade C Supported by levels III–V evidence

Adapted from Cook et al. (157) and Broderick JP et al (158).

519Crit Care Med 2006 Vol. 34, No. 2

Page 10: Controversies in the management of aneurysmal subarachnoid ... · sible aneurysm. Medical management is based on the detection and treatment of cerebral and extracerebral complications

including electrocardiographic changes,reversible cardiomyopathy (stunned myo-cardium), and release of cardiac-specificmarkers (141, 142). Aneurysmal SAH hasalso been linked to hypoxemic respiratoryfailure, which may take the form of con-gestive heart failure, neurogenic pulmo-nary edema, or acute lung injury (143).Extracerebral organ dysfunction in aSAHhas been associated with a greater risk ofpoor neurologic outcome and death(144). Although there is general agree-ment regarding the importance of cardiaccomplications after aSAH, many aspectsof their pathophysiology, diagnosis, andtreatment remain undefined.

A widely held view postulates a se-quence of events involving acute cerebralinjury, derangements in autonomic func-tion, release of endogenous cat-echolamines, and activation of adrenergicreceptors, culminating in target organdamage (145). The severity of neurologiccompromise as measured by the Huntand Hess grade is highly predictive ofmyocardial necrosis after aSAH, support-ing the hypothesis that cardiac injury af-ter aSAH is a neurally mediated process(146). Significantly, impaired left ventric-ular function and low cardiac output el-evate the risk of cerebral ischemia andhave been identified as independent pre-dictors of symptomatic vasospasm (147).

Regarding diagnosis, several investi-gators have evaluated the significance ofcardiac-specific markers such as cardiactroponin I in patients after aSAH. In a7-day observation of 39 patients withaSAH, measurements of cardiac troponinI revealed a higher incidence of myocar-dial injury (21%) than predicted byCK-MB (13%), and elevations of cardiactroponin I were associated with a higher

incidence of congestive heart failure(148). Another group found that the sen-sitivity of cardiac troponin I in the detec-tion of echocardiographically demon-strated left ventricular dysfunction wassignificantly higher than CK-MB (100%compared with 29%) (149). In a thirdstudy, mild elevations in cardiac troponinI (�2.8 ng/mL) associated with depressedleft ventricular function in the absence ofsignificant electrocardiographic changeswere proposed as characteristics thatmight aid in the differentiation of neuro-genic stunned myocardium from myocar-dial infarction (150). Recently, there hasbeen increasing recognition of the impor-tance of BNP as a diagnostic and prog-nostic marker in patients with cardiacdysfunction. Increased serum levels ofBNP are highly predictive of the short-and long-term risk of cardiac deathacross the entire spectrum of acute cor-onary syndromes and in patients with de-compensated congestive heart failure(151). A correlation between increasedlevels of BNP and myocardial necrosis,pulmonary edema, and both systolic anddiastolic left ventricular dysfunction afteraSAH was demonstrated in a recent study(136). However, there is debate as to thesource—cardiac or cerebral—of BNP inpatients with aSAH (152). A recent studyevaluated serum and CSF levels of atrialnatriuretic peptide, BNP, and troponin Tin patients with aSAH. Of note, increasedlevels of atrial natriuretic peptide andBNP were detected in the serum but notCSF, leading to the conclusion that thesepeptides were exclusively of cardiac ori-gin (153). This contradicted previous re-sults in which a selective increase of BNP,but not atrial natriuretic peptide, sug-gested that the hormone was secreted inthe brain (134).

The therapeutic approach to patientswith cardiac dysfunction acquired in thesetting of aSAH is a matter of furthercontroversy. In light of the postulatedrole of catecholamines in the develop-ment of this disorder, early randomizedtrials evaluated the effect of pharmaco-logic � and � adrenergic receptor block-ade (e.g., phentolamine and propanolol)in patients with aSAH, noting a favorableeffect on the rate of myocardial necrosis,survival, and neurologic outcome (154,155). These data are at variance withmore recent studies that suggest thatneurogenically induced ventricular dys-function and its associated low cardiacoutput/pulmonary edema may exacerbatecerebral ischemia and should be treated

with the use of inotropic agents such asdobutamine (65, 147, 156). Finally, pa-tients presenting with severe neurocar-diomyopathy or cardiogenic shock andvasospasm may benefit from temporarysupport with IABC (108, 110).

Comment. Cardiac dysfunction com-plicating aSAH is common and has beenlinked to poor neurologic outcomes.These complications may contraindicate,or be exacerbated by, brain-targeted ther-apies such as hemodynamic augmenta-tion. Studies are needed to elucidate riskfactors, pathogeneses, natural history, di-agnostic markers, management strate-gies, and prognostic impact of these com-plications.

CONCLUSIONS

This review highlights some of thecontroversies that exist in the clinicalmanagement of patients with aSAH, asummary of which is presented in Table1, along with a classification of levels ofevidence (157, 158) in Table 2.

It is anticipated that these areas ofuncertainty will generate hypothesis-driven experimental and translational re-search, prospective observational studies,and clinical trials, with the goal of im-proving outcomes in these complex andchallenging patients.

ACKNOWLEDGMENT

We thank Tzipora Sofare, MA, for hereditorial assistance in preparing this ar-ticle.

REFERENCES

1. Guglielmi G, Vinuela F, Duckwiler G, et al:Endovascular treatment of posterior circu-lation aneurysms by electrothrombosis us-ing electrically detachable coils. J Neuro-surg 1992; 77:515–524

2. Molyneux A, Kerr R, Stratton I, et al: Inter-national Subarachnoid Aneurysm Trial(ISAT) of neurosurgical clipping versus en-dovascular coiling in 2143 patients withruptured intracranial aneurysms: A ran-domized trial. International SubarachnoidAneurysm Trial (ISAT) Collaborative Group.Lancet 2002; 360:1267–1274

3. Molyneux A, Kerr R, Ly-Mee Y, et al: Inter-national subarachnoid aneurysm trial(ISAT) of neurosurgical clipping versus en-dovascular coiling in 2143 patients withruptured intracranial aneurysms: A ran-domised comparison of effects on survival,dependency, seizures, rebleeding, sub-groups, and aneurysm occlusion. Lancet2005; 366:809–817

M any aspects of

care in patients

with aneurys-

mal subarachnoid hemor-

rhage remain highly contro-

versial and warrant further

resolution with hypothesis-

driven clinical or transla-

tional research.

520 Crit Care Med 2006 Vol. 34, No. 2

Page 11: Controversies in the management of aneurysmal subarachnoid ... · sible aneurysm. Medical management is based on the detection and treatment of cerebral and extracerebral complications

4. Barreau X, Pastore M, Piotin M, et al: En-dovascular treatment of cerebral vasospasmfollowing S.A.H. Acta Neurochir Suppl2001; 77:177–180

5. Hop JW, Rinkel GJ, Algra A, et al: Quality oflife in patients and partners after aneurys-mal subarachnoid hemorrhage. Stroke1998; 29:798–804

6. Ostbye T, Levy AR, Mayo NE: Hospitaliza-tion and case-fatality rates for subarachnoidhemorrhage in Canada from 1982 through1991: The Canadian Collaborative StudyGroup of Stroke Hospitalizations. Stroke1997; 28:793–798

7. Cesarini KG, Hardemark HG, Persson L:Improved survival after aneurysmal sub-arachnoid hemorrhage: Review of casemanagement during a 12-year period.J Neurosurg 1999; 90:664–672

8. Rinkel GJ, Feigin VL, Algra A, et al: Calciumantagonists for aneurysmal subarachnoidhaemorrhage. Cochrane Database Syst Rev2005; 25:CD000277

9. Rinkel G, Feigin V, Algra A, et al: Circula-tory volume expansion therapy for aneurys-mal subarachnoid haemorrhage. CochraneDatabase Syst Rev 2004; 4:CD000483

10. Koivisto T, Vanninen R, Hurskainen H, etal: Outcomes of early endovascular versussurgical treatment of ruptured cerebral an-eurysms: A prospective randomized study.Stroke 2000; 31:2369–2377

11. Raabe A, Schmiedek P, Seifert V, et al: Ger-man Society of Neurosurgery Section onVascular Neurosurgery: Position statementon the International Subarachnoid Hemor-rhage Trial (ISAT). Zentralbl Neurochir2003; 64:99–103

12. Sade B, Mohr G: Critical appraisal of theInternational Subarachnoid Aneurysm Trial(ISAT). Neurol India 2004; 52:32–35

13. Haley EC Jr, Kassell NF, Apperson-HansenC, et al: A randomized, double-blind, vehi-cle-controlled trial of tirilazad mesylate inpatients with aneurysmal subarachnoidhemorrhage: A cooperative study in NorthAmerica. J Neurosurg 1997; 86:467–474

14. Beneficial effect of carotid endarterectomyin symptomatic patients with high-gradecarotid stenosis: North American Symp-tomatic Carotid Endarterectomy Trial Col-laborators. N Engl J Med 1991; 325:445–453

15. Endarterectomy for asymptomatic carotidartery stenosis: Executive Committee forthe Asymptomatic Carotid AtherosclerosisStudy. JAMA 1995; 273:1421–1428

16. Byrne JV, Sohn MJ, Molyneux AJ, et al:Five-year experience in using coil emboli-zation for ruptured intracranial aneurysms:Outcomes and incidence of late rebleeding.J Neurosurg 1999; 90:656–663

17. Sluzewski M, Menovsky T, van Rooij WJ, etal: Coiling of very large or giant cerebralaneurysms: Long-term clinical and serialangiographic results. AJNR Am J Neurora-diol 2003; 24:257–262

18. Friedman JA, Nichols DA, Meyer FB, et al:

Guglielmi detachable coil treatment of rup-tured saccular cerebral aneurysms: Retro-spective review of a 10-year single-centerexperience. AJNR Am J Neuroradiol 2003;24:526–533

19. Charpentier C, Audibert G, Guillemin F, etal: Multivariate analysis of predictors of ce-rebral vasospasm occurrence after aneurys-mal subarachnoid hemorrhage. Stroke1999; 30:1402–1408

20. Dehdashti AR, Mermillod B, Rufenacht DA,et al: Does treatment modality of intracra-nial ruptured aneurysms influence the in-cidence of cerebral vasospasm and clinicaloutcome? Cerebrovasc Dis 2004; 17:53–60

21. Goddard AJ, Raju PP, Gholkar A: Does themethod of treatment of acutely rupturedintracranial aneurysms influence the inci-dence and duration of cerebral vasospasmand clinical outcome? J Neurol NeurosurgPsychiatry 2004; 75:868–872

22. Groden C, Kremer C, Regelsberger J, et al:Comparison of operative and endovasculartreatment of anterior circulation aneu-rysms in patients in poor grades. Neurora-diology 2001; 43:778–783

23. Rabinstein AA, Pichelmann MA, FriedmanJA, et al: Symptomatic vasospasm and out-comes following aneurysmal subarachnoidhemorrhage: A comparison between surgi-cal repair and endovascular coil occlusion.J Neurosurg 2003; 98:319–325

24. Gruber A, Ungersbock K, Reinprecht A, etal: Evaluation of cerebral vasospasm afterearly surgical and endovascular treatmentof ruptured intracranial aneurysms. Neuro-surgery 1998; 42:258–267

25. Bleck TP: Rebleeding and vasospasm afterSAH: New strategies for improving out-come. J Crit Illn 1997; 12:572–582

26. Cloft HJ, Joseph GJ, Dion JE: Risk of cere-bral angiography in patients with subarach-noid hemorrhage, cerebral aneurysm, andarteriovenous malformation: A meta-analysis. Stroke 1999; 30:317–320

27. Aaslid R, Huber P, Nornes H: Evaluation ofcerebrovascular spasm with transcranialDoppler ultrasound. J Neurosurg 1984; 60:37–41

28. Vora YY, Suarez-Almazor M, Steinke DE, etal: Role of transcranial Doppler monitoringin the diagnosis of cerebral vasospasm aftersubarachnoid hemorrhage. Neurosurgery1999; 44:1237–1247

29. Okada Y, Shima T, Nishida M, et al: Com-parison of transcranial Doppler investiga-tion of aneurysmal vasospasm with digitalsubtraction angiographic and clinical find-ings. Neurosurgery 1999; 45:443–449

30. Venkatesh B, Shen Q, Lipman J: Continu-ous measurement of cerebral blood flowvelocity using transcranial Doppler revealssignificant moment-to-moment variabilityof data in healthy volunteers and in patientswith subarachnoid hemorrhage. Crit CareMed 2002; 30:563–569

31. Grosset DG, Straiton J, McDonald I, et al:Angiographic and Doppler diagnosis of ce-

rebral artery vasospasm following subarach-noid haemorrhage. Br J Neurosurg 1993;7:291–298

32. Lindegaard KF, Nornes H, Bakke SJ, et al:Cerebral vasospasm after subarachnoidhaemorrhage investigated by means oftranscranial Doppler ultrasound. Acta Neu-rochir Suppl (Wien) 1988; 42:81–84

33. Torbey MT, Hauser TK, Bhardwaj A, et al:Effect of age on cerebral blood flow velocityand incidence of vasospasm after aneurys-mal subarachnoid hemorrhage. Stroke2001; 32:2005–2011

34. Proust F, Callonec F, Clavier E, et al: Use-fulness of transcranial color-coded sonogra-phy in the diagnosis of cerebral vasospasm.Stroke 1999; 30:1091–1098

35. Lysakowski C, Walder B, Costanza MC, et al:Transcranial Doppler versus angiography inpatients with vasospasm due to a rupturedcerebral aneurysm: A systematic review.Stroke 2001; 32:2292–2298

36. Takagi R, Hayashi H, Kobayashi H, et al:Three-dimensional CT angiography of in-tracranial vasospasm following subarach-noid haemorrhage. Neuroradiology 1998;40:631–635

37. Anderson GB, Ashforth R, Steinke DE, et al:CT angiography for the detection of cere-bral vasospasm in patients with acute sub-arachnoid hemorrhage. AJNR Am J Neuro-radiol 2000; 21:1011–1015

38. Otawara Y, Ogasawara K, Ogawa A, et al:Evaluation of vasospasm after subarachnoidhemorrhage by use of multislice computedtomographic angiography. Neurosurgery2002; 51:939–942

39. Rabinstein AA, Friedman JA, Weigand SD,et al: Predictors of cerebral infarction inaneurysmal subarachnoid hemorrhage.Stroke 2004; 35:1862–1866

40. Condette-Auliac S, Bracard S, Anxionnat R,et al: Vasospasm after subarachnoid hemor-rhage: Interest in diffusion-weighted MRimaging. Stroke 2001; 32:1818–1824

41. Hertel F, Walter C, Bettag M, et al: Perfu-sion-weighted magnetic resonance imagingin patients with Vasospasm: A useful newtool in the management of patients withsubarachnoid hemorrhage. Neurosurgery2005; 56:28–35

42. Rajendran JG, Lewis DH, Newell DW, et al:Brain SPECT used to evaluate vasospasmafter subarachnoid hemorrhage: Correla-tion with angiography and transcranialDoppler. Clin Nucl Med 2001; 26:125–130

43. Jabre A, Babikian V, Powsner RA, et al: Roleof single photon emission computed to-mography and transcranial Doppler ultra-sonography in clinical vasospasm. J ClinNeurosci 2002; 9:400–403

44. Powers WJ, Grubb RL Jr, Baker RP, et al:Regional cerebral blood flow and metabo-lism in reversible ischemia due to vaso-spasm: Determination by positron emissiontomography. J Neurosurg 1985; 62:539–546

45. Carpenter DA, Grubb RL Jr, Tempel LW, et

521Crit Care Med 2006 Vol. 34, No. 2

Page 12: Controversies in the management of aneurysmal subarachnoid ... · sible aneurysm. Medical management is based on the detection and treatment of cerebral and extracerebral complications

al: Cerebral oxygen metabolism after aneu-rysmal subarachnoid hemorrhage. J CerebBlood Flow Metab 1991; 11:837–844

46. Sarrafzadeh AS, Sakowitz OW, Kiening KL,et al: Bedside microdialysis: A tool to mon-itor cerebral metabolism in subarachnoidhemorrhage patients? Crit Care Med 2002;30:1062–1070

47. Skjoth-Rasmussen J, Schulz M, KristensenSR, et al: Delayed neurological deficits de-tected by an ischemic pattern in the extra-cellular cerebral metabolites in patientswith aneurysmal subarachnoid hemor-rhage. J Neurosurg 2004; 100:8–15

48. Unterberg AW, Sakowitz OW, SarrafzadehAS, et al: Role of bedside microdialysis inthe diagnosis of cerebral vasospasm follow-ing aneurysmal subarachnoid hemorrhage.J Neurosurg 2001; 94:740–749

49. Sarrafzadeh AS, Haux D, Ludemann L, et al:Cerebral ischemia in aneurysmal subarach-noid hemorrhage: A correlative microdialy-sis-PET study. Stroke 2004; 35:638–643

50. Bhardwaj A: In vivo regional neurochemis-try in stroke: Clinical applications, limita-tions, and future directions. Stroke 2004;35:e74–e76

51. Peerdeman SM, van Tulder MW, VandertopWP: Cerebral microdialysis as a monitoringmethod in subarachnoid hemorrhage pa-tients, and correlation with clinical events:A systematic review. J Neurol 2003; 250:797–805

52. Rivierez M, Landau-Ferey J, Grob R, et al:Value of electroencephalogram in predic-tion and diagnosis of vasospasm after intra-cranial aneurysm rupture. Acta Neurochir(Wien) 1991; 110:17–23

53. Vespa PM, Nuwer MR, Juhasz C, et al: Earlydetection of vasospasm after acute sub-arachnoid hemorrhage using continuousEEG ICU monitoring. ElectroencephalogrClin Neurophysiol 1997; 103:607–615

54. Claassen J, Hirsch LJ, Kreiter KT, et al:Quantitative continuous EEG for detectingdelayed cerebral ischemia in patients withpoor-grade subarachnoid hemorrhage. ClinNeurophysiol 2004; 115:2699–2710

55. Sen J, Belli A, Albon H, et al: Triple-Htherapy in the management of aneurysmalsubarachnoid haemorrhage. Lancet Neurol2003; 2:614–621

56. Kosnik EJ, Hunt WE: Postoperative hyper-tension in the management of patients withintracranial arterial aneurysms. J Neuro-surg 1976; 45:148–154

57. Giannotta SL, McGillicuddy JE, Kindt GW:Diagnosis and treatment of postoperativecerebral vasospasm. Surg Neurol 1977;8:286–290

58. Awad IA, Carter LP, Spetzler RF, et al: Clin-ical vasospasm after subarachnoid hemor-rhage: Response to hypervolemic hemodilu-tion and arterial hypertension. Stroke 1987;18:365–372

59. Kassell NF, Peerless SJ, Durward QJ, et al:Treatment of ischemic deficits from vaso-spasm with intravascular volume expansion

and induced arterial hypertension. Neuro-surgery 1990; 11:337–343

60. Origitano TC, Wascher TM, Reichman OH,et al: Sustained increased cerebral bloodflow with prophylactic hypertensive hyper-volemic hemodilution (“triple-H” therapy)after subarachnoid hemorrhage. Neurosur-gery 1990; 27:729–739

61. Rosenwasser RH, Delgado TE, BuchheitWA, et al: Control of hypertension and pro-phylaxis against vasospasm in cases of sub-arachnoid hemorrhage: A preliminary re-port. Neurosurgery 1983; 12:658–660

62. Lennihan L, Mayer SA, Fink ME, et al: Ef-fect of hypervolemic therapy on cerebralblood flow after subarachnoid hemorrhage:A randomized controlled trial. Stroke 2000;31:383–391

63. Egge A, Waterloo K, Sjoholm H, et al: Pro-phylactic hyperdynamic postoperative fluidtherapy after aneurysmal subarachnoidhemorrhage: A clinical, prospective, ran-domized, controlled study. Neurosurgery2001; 49:593–605

64. Treggiari MM, Walder B, Suter PM, et al:Systematic review of the prevention of de-layed ischemic neurological deficits withhypertension, hypervolemia, and hemodilu-tion therapy following subarachnoid hem-orrhage. J Neurosurg 2003; 98:978–984

65. Levy ML, Rabb CH, Zelman V, et al: Cardiacperformance enhancement from dobut-amine in patients refractory to hypervole-mic therapy for cerebral vasospasm. J Neu-rosurg 1993; 79:494–499

66. Kim D, Joseph M, Ziadi S, et al: Increases incardiac output can reverse flow deficitsfrom vasospasm independent of blood pres-sure: A study using xenon computed tomo-graphic measurement of cerebral bloodflow. Neurosurgery 2003; 53:1044–1051

67. Bouma GJ, Muizelaar JP: Relationship be-tween cardiac output and cerebral bloodflow in patients with intact and with im-paired autoregulation. J Neurosurg 1990;73:368–374

68. Milnor WR: Pulsatile blood flow. N EnglJ Med 1972; 287:27–34

69. Morgan MK, Jonker B, Finfer S, et al: Ag-gressive management of aneurysmal sub-arachnoid haemorrhage based on a papav-erine angioplasty protocol. J Clin Neurosci2000; 7:305–308

70. Smith WS, Dowd CF, Johnston SC, et al:Neurotoxicity of intra-arterial papaverinepreserved with chlorobutanol used for thetreatment of cerebral vasospasm after aneu-rysmal subarachnoid hemorrhage. Stroke2004; 35:2518–2522

71. Feng L, Fitzsimmons BF, Young WL, et al:Intraarterially administered verapamil asadjunct therapy for cerebral vasospasm:Safety and 2-year experience. AJNR Am JNeuroradiol 2002; 23:1284–1290

72. Biondi A, Ricciardi GK, Puybasset L, et al:Intra-arterial nimodipine for the treatmentof symptomatic cerebral vasospasm afteraneurysmal subarachnoid hemorrhage: pre-

liminary results. AJNR Am J Neuroradiol2004; 25:1067–1076

73. Badjatia N, Topcuoglu MA, Pryor JC, et al:Preliminary experience with intra-arterialnicardipine as a treatment for cerebral va-sospasm. AJNR Am J Neuroradiol 2004; 25:819–826

74. Muizelaar JP, Zwienenberg M, Rudisill NA,et al: The prophylactic use of transluminalballoon angioplasty in patients with FisherGrade 3 subarachnoid hemorrhage: A pilotstudy. J Neurosurg 1999; 91:51–58

75. Apostolides PJ, Greene KA, Zabramski JM,et al: Intra-aortic balloon pump counterpul-sation in the management of concomitantcerebral vasospasm and cardiac failure aftersubarachnoid hemorrhage: Technical casereport. Neurosurgery 1996; 38:1056–1059

76. Nussbaum ES, Sebring LA, Ganz WF, et al:Intra-aortic balloon counterpulsation aug-ments cerebral blood flow in the patientwith cerebral vasospasm: A xenon-enhancedcomputed tomography study. Neurosurgery1998; 42:206–213

77. Spann RG, Lang DA, Birch AA, et al: Intra-aortic balloon counterpulsation: Augmenta-tion of cerebral blood flow after aneurysmalsubarachnoid haemorrhage. Acta Neuro-chir (Wien) 2001; 143:115–123

78. Montessuit M, Chevalley C, King J, et al:The use of intra-aortic counterpulsationballoon for the treatment of cerebral vaso-spasm and edema. Surgery 2000; 127:230–233

79. Rosen CL, Sekhar LN, Duong DH: Use ofintra-aortic balloon pump counterpulsationfor refractory symptomatic vasospasm. ActaNeurochir (Wien) 2000; 142:25–32

80. Kassell NF, Haley EC Jr, Apperson-HansenC, et al: Randomized, double-blind, vehicle-controlled trial of tirilazad mesylate in pa-tients with aneurysmal subarachnoid hem-orrhage: A cooperative study in Europe,Australia, and New Zealand. J Neurosurg1996; 84:221–228

81. Lanzino G, Kassell NF, Dorsch NW, et al:Double-blind, randomized, vehicle-con-trolled study of high-dose tirilazad mesylatein women with aneurysmal subarachnoidhemorrhage: Part I. A cooperative study inEurope, Australia, New Zealand, and SouthAfrica. J Neurosurg 1999; 90:1011–1017

82. Lanzino G, Kassell NF: Double-blind, ran-domized, vehicle-controlled study of high-dose tirilazad mesylate in women with an-eurysmal subarachnoid hemorrhage: PartII. A cooperative study in North America.J Neurosurg 1999; 90:1018–1024

83. Dorsch NW, Kassell NF, Sinkula MS: Meta-analysis of trials of tirilazad mesylate inaneurysmal SAH. Acta Neurochir Suppl2001; 77:233–235

84. Chen D, Nishizawa S, Yokota N, et al: High-dose methylprednisolone prevents vaso-spasm after subarachnoid hemorrhagethrough inhibition of protein kinase C ac-tivation. Neurol Res 2002; 24:215–222

85. Shibata S, Suzuki S, Ohkuma H, et al: Ef-

522 Crit Care Med 2006 Vol. 34, No. 2

Page 13: Controversies in the management of aneurysmal subarachnoid ... · sible aneurysm. Medical management is based on the detection and treatment of cerebral and extracerebral complications

fects of intracisternal methylprednisoloneon lipid peroxidation in experimental sub-arachnoid haemorrhage. Acta Neurochir(Wien) 1999; 141:529–532

86. Chyatte D: Prevention of chronic cerebralvasospasm in dogs with ibuprofen and high-dose methylprednisolone. Stroke 1989; 20:1021–1026

87. Chyatte D, Fode NC, Nichols DA, et al: Pre-liminary report: Effects of high dose meth-ylprednisolone on delayed cerebral isch-emia in patients at high risk for vasospasmafter aneurysmal subarachnoid hemor-rhage. Neurosurgery 1987; 21:157–160

88. Veyna RS, Seyfried D, Burke DG, et al: Mag-nesium sulfate therapy after aneurysmalsubarachnoid hemorrhage. J Neurosurg2002; 96:510–514

89. Van den Bergh: Magnesium sulfate in aneu-rysmal subarachnoid hemorrhage: A ran-domized controlled trial. MASH StudyGroup (abstract). Stroke 2005; 36:434(A85)

90. Shaw MD, Vermeulen M, Murray GD, et al:Efficacy and safety of the endothelin, recep-tor antagonist TAK-044 in treating sub-arachnoid hemorrhage: A report by theSteering Committee on behalf of the UK/Netherlands/Eire TAK-044 SubarachnoidHaemorrhage Study Group. J Neurosurg2000; 93:992–997

91. Mascia L, Fedorko L, Stewart DJ, et al: Tem-poral relationship between endothelin-1concentrations and cerebral vasospasm inpatients with aneurysmal subarachnoidhemorrhage. Stroke 2001; 32:1185–1190

92. Davignon J: Beneficial cardiovascular pleio-tropic effects of statins. Circulation 2004;109(23 Suppl 1):III39–III43

93. McGirt MJ, Lynch JR, Parra A, et al: Sim-vastatin increases endothelial nitric oxidesynthase and ameliorates cerebral vaso-spasm resulting from subarachnoid hemor-rhage. Stroke 2002; 33:2950–2956

94. Parra A, Kreiter KT, Williams S, et al: Effectof prior statin use on functional outcomeand delayed vasospasm after acute aneurys-mal subarachnoid hemorrhage: A matchedcontrolled cohort study. Neurosurgery2005; 56:476–484

95. Singhal AB, Topcuoglu MA, Dorer DJ, et al:SSRI and statin use increases the risk forvasospasm after subarachnoid hemorrhage.Neurology 2005; 64:1008–1013

96. Lynch JR, Wang H, McGirt MJ, et al: Sim-vastatin reduces vasospasm after aneurys-mal subarachnoid hemorrhage: Results of apilot randomized clinical trial. Stroke 2005;36:2024–2026

97. Tseng MY, Czosnyka M, Richards H, et al:Effects of acute treatment with pravastatinon cerebral vasospasm, autoregulation, anddelayed ischemic deficits after aneurysmalsubarachnoid hemorrhage: A phase II ran-domized placebo-controlled trial. Stroke2005; 36:1627–1632

98. Grasso G, Buemi M, Alafaci C, et al: Bene-ficial effects of systemic administration ofrecombinant human erythropoietin in rab-

bits subjected to subarachnoid hemorrhage.Proc Natl Acad Sci U S A 2002; 99:5627–5631

99. Wolf EW, Banerjee A, Soble-Smith J, et al:Reversal of cerebral vasospasm using an in-trathecally administered nitric oxide donor.J Neurosurg 1998; 89:279–288

100. Kwan AL, Lin CL, Yanamoto H, et al: Sys-temic administration of the potassiumchannel activator cromakalim attenuatescerebral vasospasm after experimental sub-arachnoid hemorrhage. Neurosurgery1998; 42:347–350

101. Thome C, Schubert GA, Schilling L: Hypo-thermia as a neuroprotective strategy insubarachnoid hemorrhage: A pathophysio-logical review focusing on the acute phase.Neurol Res 2005; 27:229–237

102. Hindman BJ, Todd MM, Gelb AW, et al: Mildhypothermia as a protective therapy duringintracranial aneurysm surgery: A random-ized prospective pilot trial. Neurosurgery1999; 44:23–32

103. Todd MM, Hindman BJ, Clarke WR, et al:Mild intraoperative hypothermia duringsurgery for intracranial aneurysm: Intraop-erative Hypothermia for Aneurysm SurgeryTrial (IHAST) Investigators. N Engl J Med2005; 352:135–145

104. Nagao S, Irie K, Kawai N, et al: The use ofmild hypothermia for patients with severevasospasm: A preliminary report. J ClinNeurosci 2003; 10:208–212

105. Gasser S, Khan N, Yonekawa Y, et al: Long-term hypothermia in patients with severebrain edema after poor-grade subarachnoidhemorrhage: Feasibility and intensive carecomplications. J Neurosurg Anesthesiol2003; 15:240–248

106. Sato K, Sato K, Yoshimoto T: Systemic andcerebral haemodynamics during craniot-omy under mild hypothermia in patientswith acute subarachnoid haemorrhage.Acta Neurochir (Wien) 2000; 142:1013–1019

107. Kawamura S, Suzuki A, Hadeishi H, et al:Cerebral blood flow and oxygen metabolismduring mild hypothermia in patients withsubarachnoid haemorrhage. Acta Neuro-chir (Wien) 2000; 142:1117–1121

108. Fisher CM, Kistler JP, Davis JM: Relation ofcerebral vasospasm to subarachnoid hem-orrhage visualized by computerized tomo-graphic scanning. Neurosurgery 1980;6:1–9

109. Sasaki T, Kodama N, Kawakami M, et al:Urokinase cisternal irrigation therapy forprevention of symptomatic vasospasm afteraneurysmal subarachnoid hemorrhage: Astudy of urokinase concentration and thefibrinolytic system. Stroke 2000; 31:1256–1262

110. Hamada J, Kai Y, Morioka M, et al: Effect oncerebral vasospasm of coil embolization fol-lowed by microcatheter intrathecal uroki-nase infusion into the cisterna magna: Aprospective randomized study. Stroke 2003;34:2549–2554

111. Amin-Hanjani S, Ogilvy CS, Barker FG:Does intracisternal thrombolysis preventvasospasm after aneurysmal subarachnoidhemorrhage? A meta-analysis. Neurosur-gery 2004; 54:326–334

112. Chimowitz MI, Pessin MS: Is there a role forheparin in the management of complica-tions of subarachnoid hemorrhage? Stroke1987; 18:1169–1172

113. Wurm G, Tomancok B, Nussbaumer K, etal: Reduction of ischemic sequelae follow-ing spontaneous subarachnoid hemor-rhage: A double-blind, randomized compar-ison of enoxaparin versus placebo. ClinNeurol Neurosurg 2004; 106:97–103

114. Siironen J, Juvela S, Varis J, et al: No effectof enoxaparin on outcome of aneurysmalsubarachnoid hemorrhage: A randomized,double-blind, placebo-controlled clinicaltrial. J Neurosurg 2003; 99:953–959

115. Ohkuma H, Suzuki S, Kimura M, et al: Roleof platelet function in symptomatic cerebralvasospasm following aneurysmal subarach-noid hemorrhage. Stroke 1991; 22:854–859

116. Romano JG, Forteza AM, Concha M, et al:Detection of microemboli by transcranialDoppler ultrasonography in aneurysmalsubarachnoid hemorrhage. Neurosurgery2002; 50:1026–1030

117. Dorhout Mees SM, Rinkel GJ, Hop JW, et al:Antiplatelet therapy in aneurysmal sub-arachnoid hemorrhage: A systematic re-view. Stroke 2003; 34:2285–2289

118. Lin CL, Dumont AS, Lieu AS, et al: Char-acterization of perioperative seizures andepilepsy following aneurysmal subarach-noid hemorrhage. J Neurosurg 2003; 99:978–985

119. Ogden JA, Utley T, Mee EW: Neurologicaland psychosocial outcome 4 to 7 years aftersubarachnoid hemorrhage. Neurosurgery1997; 41:25–34

120. Rhoney DH, Tipps LB, Murry KR, et al:Anticonvulsant prophylaxis and timing ofseizures after aneurysmal subarachnoidhemorrhage. Neurology 2000; 55:258–265

121. Butzkueven H, Evans AH, Pitman A, et al:Seizures independently predict poor out-come after subarachnoid hemorrhage. Neu-rology 2000; 55:1315–1320

122. Claassen J, Peery S, Kreiter KT, et al: Pre-dictors and clinical impact of epilepsy aftersubarachnoid hemorrhage. Neurology2003; 60:208–214

123. Ohman J: Hypertension as a risk factor forepilepsy after aneurysmal subarachnoidhemorrhage and surgery. Neurosurgery1990; 27:578–581

124. Naidech AM, Kreiter KT, Janjua N, et al:Phenytoin exposure is associated with func-tional and cognitive disability after sub-arachnoid hemorrhage. Stroke 2005; 36:583–587

125. Dennis LJ, Claassen J, Hirsch LJ, et al: Non-convulsive status epilepticus after sub-arachnoid hemorrhage. Neurosurgery2002; 51:1136–1143

126. Ogawasara K, Kinouchi H, Nagamine Y, et

523Crit Care Med 2006 Vol. 34, No. 2

Page 14: Controversies in the management of aneurysmal subarachnoid ... · sible aneurysm. Medical management is based on the detection and treatment of cerebral and extracerebral complications

al: Differential diagnosis of hyponatremiafollowing subarachnoid hemorrhage. NoShinkei Geka 1998; 26:501–505

127. Maesaka JK, Gupta S, Fishbane S: Cerebralsalt-wasting syndrome: Does it exist?Nephron 1999; 82:100–9

128. Palmer BF: Hyponatremia in patients withcentral nervous system disease: SIADH ver-sus CSW. Trends Endocrinol Metab 2003;14:182–187

129. Harrigan MR: Cerebral salt wasting syn-drome: A review. Neurosurgery 1996; 38:152–160

130. Diringer M, Ladenson PW, Stern BJ, et al:Plasma atrial natriuretic factor and sub-arachnoid hemorrhage. Stroke 1988; 19:1119–1124

131. Berendes E, Walter M, Cullen P, et al: Se-cretion of brain natriuretic peptide in pa-tients with aneurysmal subarachnoid haem-orrhage. Lancet 1997; 349:245–249

132. Isotani E, Suzuki R, Tomita K, et al: Alter-ations in plasma concentrations of natri-uretic peptides and antidiuretic hormoneafter subarachnoid hemorrhage. Stroke1994; 25:2198–2203

133. Tomida M, Muraki M, Uemura K, et al:Plasma concentrations of brain natriureticpeptide in patients with subarachnoid hem-orrhage. Stroke 1998; 29:1584–1587

134. Sviri GE, Feinsod M, Soustiel JF: Brain na-triuretic peptide and cerebral vasospasm insubarachnoid hemorrhage: Clinical andTCD correlations. Stroke 2000; 31:118–122

135. McGirt MJ, Blessing R, Nimjee SM, et al:Correlation of serum brain natriuretic pep-tide with hyponatremia and delayed isch-emic neurological deficits after subarach-noid hemorrhage. Neurosurgery 2004; 54:1369–1373

136. Tung PP, Olmsted E, Kopelnik A, et al:Plasma B-type natriuretic peptide levels areassociated with early cardiac dysfunctionafter subarachnoid hemorrhage. Stroke2005; 36:1567–1569

137. Inoha S, Inamura T, Nakamizo A, et al:Fluid loading in rats increases serum brainnatriuretic peptide concentration. NeurolRes 2001; 23:93–95

138. Wijdicks EFM, Vermeulen M, van Brum-melen P, et al: The effect of fludrocortisoneacetate on plasma volume and natriuresis inpatients with aneurysmal subarachnoidhemorrhage. Clin Neurol Neurosurg 1988;90:209–214

139. Hasan D, Lindsay KW, Wijdicks EFM, et al:Effect of fludrocortisone acetate in patientswith subarachnoid hemorrhage. Stroke1989; 20:1156–1161

140. Moro N, Katayama Y, Kojima J, et al: Pro-phylactic management of excessive natri-uresis with hydrocortisone for efficient hy-pervolemic therapy after subarachnoidhemorrhage. Stroke 2003; 34:2807–2811

141. Macmillan CS, Grant IS, Andrews PJ: Pul-monary and cardiac sequelae of subarach-noid haemorrhage: Time for active manage-ment? Intensive Care Med 2002; 28:1012–1023

142. Sakr YL, Ghosn I, Vincent JL: Cardiac man-ifestations after subarachnoid hemorrhage:A systematic review of the literature. ProgCardiovasc Dis 2002; 45:67–80

143. Mayer SA, Fink ME, Homma S, et al: Car-diac injury associated with neurogenic pul-monary edema following subarachnoidhemorrhage. Neurology 1994; 44:815–820

144. Gruber A, Reinprecht A, Illievich UM, et al:Extracerebral organ dysfunction and neuro-logic outcome after aneurysmal subarach-noid hemorrhage. Crit Care Med 1999; 27:505–514

145. Masuda T, Sato K, Yamamoto S, et al: Sym-pathetic nervous activity and myocardialdamage immediately after subarachnoidhemorrhage in a unique animal model.Stroke 2002; 33:1671–1676

146. Tung P, Kopelnik A, Banki N, et al: Predic-tors of neurocardiogenic injury after sub-arachnoid hemorrhage. Stroke 2004; 35:548–551

147. Mayer SA, Lin J, Homma S, et al: Myocar-dial injury and left ventricular performanceafter subarachnoid hemorrhage. Stroke1994; 30:780–786

148. Parekh N, Venkatesh B, Cross D, et al: Car-diac troponin I predicts myocardial dys-function in aneurysmal subarachnoid hem-

orrhage. J Am Coll Cardiol 2000; 36:1328–1335

149. Deibert E, Barzilai B, Braverman AC, et al:Clinical significance of elevated troponin Ilevels in patients with nontraumatic sub-arachnoid hemorrhage. J Neurosurg 2003;98:741–746

150. Bulsara KR, McGirt MJ, Liao L, et al: Use ofthe peak troponin value to differentiatemyocardial infarction from reversible neu-rogenic left ventricular dysfunction associ-ated with aneurysmal subarachnoid hemor-rhage. J Neurosurg 2003; 98:524–528

151. Cowie MR, Jourdain P, Maisel A, et al: Clin-ical applications of B-type natriuretic pep-tide (BNP) testing. Eur Heart J 2003; 24:1710–1718

152. Clerico A, Emdin M: Diagnostic accuracyand prognostic relevance of the measure-ment of cardiac natriuretic peptides: A re-view. Clin Chem 2004; 50:33–50

153. Espiner EA, Leikis R, Ferch RD, et al: Theneuro-cardio-endocrine response to acutesubarachnoid haemorrhage. Clin Endocri-nol 2002; 56:629–635

154. Neil-Dwyer G, Walter P, Cruickshank JM, etal: Effect of propranolol and phentolamineon myocardial necrosis after subarachnoidhaemorrhage. BMJ 1978; 2:990–992

155. Walter P, Neil-Dwyer G, Cruickshank JM:Beneficial effects of adrenergic blockade inpatients with subarachnoid haemorrhage.BMJ 1982; 284:1661–1664

156. Parr MJ, Finfer SR, Morgan MK: Revers-ible cardiogenic shock complicating sub-arachnoid haemorrhage. BMJ 1996; 313:681– 683

157. Cook DJ, Guyatt GH, Laupacis A, et al:Rules of evidence and clinical recommen-dations on the use of antithromboticagents. Chest 1992; 102:305S–311S

158. Broderick JP, Adams HP Jr, Barsan W, et al:Guidelines for the management for sponta-neous intracerebral hemorrhage: State-ment for healthcare professionals from aspecial writing group of the Stroke Council,AHA. Stroke 1999; 30:905–915

524 Crit Care Med 2006 Vol. 34, No. 2