Transl. Stroke Res. DOI 10.1007/s12975-015-0398-6

EDITORIAL

Aneurysmal Subarachnoid Hemorrhage—Status Quo and Perspective Nima Etminan 1

Received: 26 March 2015 / Accepted: 31 March 2015 # Springer Science+Business Media New York 2015

Aneurysmal subarachnoid hemorrhage (SAH) accounts for about 5 % of all strokes and continues to be a major cause of morbidity and mortality, especially in younger patients (mean age of 52). The global incidence of SAH is 9 per 100, 000 per year with some regional variations [1]. Data from population-based studies suggests a distinct decrease in case fatality (0.9 % per year) over the past four decades to about 30 %, which is likely originated in earlier and more advanced aneurysm repair as well as improved neurocritical care of SAH patients following aneurysm repair [2]. Nevertheless, the incidence of poor functional outcome in SAH patients remains high (about one third of patients). The main determinants for poor outcome after SAH are the degree of early brain injury (EBI) and the incidence of delayed cerebral ischemia (DCI) (see below) [3, 4]. The clinical course of SAH is complex and can be divided into an acute stage (the initial 72 h after SAH ictus) and a subacute stage, which lasts up until 20 days after SAH ictus. The potential main complications in the acute phase are EBI, rebleeding from the ruptured aneurysms, with a frequency of up to 23 % within the first 72 h, and acute hydrocephalus, occurring in about 20 % of the patients, whereas the main complications occurring in the subacute phase are DCI (formerly referred to as Bvasospasm,^ see below), cardiopulmonary or other medical complications, as well as permanent hydrocephalus [5–7]. The term EBI refers not only to direct

* Nima Etminan [email protected] 1

Department of Neurosurgery, Medical Faculty, Heinrich-HeineUniversity, Moorenstraße 5, 40225 Düsseldorf, Germany

mechanical damage to the brain tissue due to the hemorrhage but also to a cascade of different pathomechanisms, incl. a temporary increase of intracranial pressure, consecutive reduction of cerebral blood flow and/or microcirculatory spasm, transient global ischemia, neuronal injury or apoptosis, and edema formation [8]. The degree of EBI is a strong and independent predictor for clinical outcome after SAH [9, 3, 4]. The term DCI describes a complex phenomenon as a consequence of several pathomechanisms, incl. (1) macrovascular or angiographic vasospasm, i.e., narrowing of proximal cerebral arteries, (2) microvascular spasm, (3) microthromboembolism, (4) cortical spreading depolarization/ischemia, (5) autoregulatory dysfunction, and (6) inflammation [10–13, 7, 14–17]. The radiological term vasospasm has been widely replaced with the clinical term DCI, which describes a secondary neurological deterioration in the course SAH, excluding other causes of neurological deterioration; an explicit definition for DCI has been proposed by a multidisciplinary research group [16]. This Bparadigm shift^ came along with the insight that angiographic vasospasm occurs in up to 70 % of SAH patients, whereas only 30 % of SAH patients develop clinical features of DCI and, moreover, because pharmaceutical treatments exclusively targeting of angiographic vasospasm failed to improve neurological outcome after SAH [18–20]. However, in those clinical studies where cerebral infarction due to DCI was effectively reduced by experimental pharmaceutical treatments, a significant improvement of clinical outcome was also seen. These data further underlined the concept that DCI comprises several, pro-ischemic pathomechanisms, which ultimately result in cerebral infarction, but the individual importance of each of these pro-ischemic pathomechanisms remains to elucidated [21, 22]. The general shift in scientific focus, i.e., away from ‘vasospasm’ towards DCI and EBI, can also be

Transl. Stroke Res.

appreciated in the research topics of more recent SAH studies: Recent experimental studies investigated mechanisms of EBI and potential pharmaceutical or interventional means to limit or even reverse the extent of EBI with the aim to reduce the incidence of DCI or improve neurological outcome [8, 23–26]. The relevance of apoptosis inhibitors or attenuation of other causes of early neuronal injury and inflammation (e.g., p53, caspase, matrix metalloproteinase-9, aquaporin-1, VEGF, etc.) have been reported in the setting of experimental SAH [23, 25, 27–30]. Pharmacokinetics and in vivo characteristics of intrathecally or intracisternally administered, sustained-release formulations for prevention of DCI (e.g., nicardipine, nimodipine) have gained increasing interest [31, 32]. The same holds true for experimental studies investigating pathomechanisms involved in microcirculatory impairment or the role of the neurovascular unit in the pathogenesis of DCI [18, 33, 34]. Consequently, existing experimental SAH models have also been refined or modified to allow for more comprehensive experimental studies on mechanisms of EBI and DCI [35–39]. Thus, it will be interesting to see whether the promising data from these experimental studies on EBI and DCI could also be reproduced in future clinical studies. Recent clinical studies have further investigated radiological means to detect EBI and DCI, the appropriate neurocritical management of SAH patients as well as novel pharmaceutical concepts for prevention of DCI or poor outcome. Further, the efficacy of antifibrinolytic drugs (e.g., aminocaproic acid, tranexamic acid) for prevention of early aneurysm rerupture remains unclear and is currently being investigated in a clinical trial [40, 41, 6]. Interestingly and despite the increasing data on the merits of perfusion CT imaging as a more sensitive radiological means to detect DCI, a recent decision analysis based on Markov modeling underlined that the neurological status of SAH patients seems to remain the most sensitive surrogate for the presence of DCI [42–44]. Here, automated analyses and harmonized PCT algorithms may help to further establish the standardized use of perfusion CT imaging for the more accurate detection or even prediction of DCI [45]. For patients with clinical features of DCI, induced hypertension and euvolemia instead of hypervolemia have been proposed as rescue therapy, but the beneficial use of such strategies remains to be proven in a randomized, controlled trial [46–48]. In case of refractory DCI, some studies have suggested pharmaceutical or balloon angioplasty, but these treatments remain controversial since robust evidence is lacking [49]. Numerous clinical trials studied novel pharmaceutical treatments (i.e., nicardipine, tirilazad, statins, clazosentan, magnesium, etc.) against DCI, but oral nimodipine remains the only drug to improve neurological outcome in SAH patients [50, 19, 51, 20, 52]. Whether the lack of efficacy of these drugs originated from small sample sizes, insensitive

outcome measures, insensitive pharmacological targets, or detrimental systemic effects is uncertain [53]. To further investigate this, a multidisciplinary research group has been established to build a Subarachnoid Hemorrhage International Trialists (SAHIT) data repository based on prospectively collected SAH patient data from over 14,000 patients; these data may help to establish a novel SAH outcome prediction model based on this very large prospective patient cohort [54–56]. In line with the improved clinical outcomes of SAH patients, a growing body of data highlights that frequently used SAH outcome grading scales (e.g., modified Rankin Scale or Glasgow Outcome Scale) may be somewhat insensitive to detect neuropsychological (depression, anxiety, etc.) or cognitive deficits in SAH patients [57–59]. In summary, the ongoing SAH research seems to address (a) the further analysis of pathomechanisms of early brain injury and DCI; (b) Bbridge the gap^ between experimental and clinical research; (c) the optimization and harmonization of neurocritical care in SAH patients; (d) new imaging techniques or biomarkers to detect and measure EBI or DCI as surrogates for clinical outcome; (e) the efficacy of systemic versus local administration of vasodilators; (f) mechanisms, sequelae, and potential treatments of neuropsychological deficits after SAH; and (g) novel pharmaceutical targets strategies to limit or reverse the extent of early brain injury and DCI. New insights are to be expected from the ongoing clinical trials investigating the effect of systemic (prostacyclin, ketamine) or local (EG-1962) pharmaceutical treatment for prevention of DCI, pharmacologic (hemodynamic therapy, NCT 01613235) or interventional rescue therapy (intra-arterial vasodilators, NCT 01996436 or balloon angioplasty), or ischemic preconditioning (NCT01158508) or hypercapnia (NCT00930072) [60–62]. Ultimately, all the studies briefly discussed here underline that a large variety of promising scientific angles are being pursued from SAH research groups around the globe to build upon and expand on the already striking progress in the research and treatment of SAH. Conflict of Interest NE served as a scientific advisor for Edge Therapeutics Inc.

References 1.

2.

3.

de Rooij NK, Linn FH, van der Plas JA, Algra A, Rinkel GJ. Incidence of subarachnoid haemorrhage: a systematic review with emphasis on region, age, gender and time trends. J Neurol Neurosurg Psychiatry. 2007;78(12):1365–72. doi:10.1136/jnnp.2007.117655. Lovelock CE, Rinkel GJ, Rothwell PM. Time trends in outcome of subarachnoid hemorrhage: population-based study and systematic review. Neurology. 2010;74(19):1494–501. doi:10.1212/WNL. 0b013e3181dd42b3. Fergusen S, Macdonald RL. Predictors of cerebral infarction in patients with aneurysmal subarachnoid hemorrhage. Neurosurgery. 2007;60(4): 658–67. doi:10.1227/01.NEU.0000255396.23280.31. discussion 67.

Transl. Stroke Res. 4.

5.

6.

7.

8.

9.

10.

11.

12.

13.

14.

15.

16.

17.

18.

Vergouwen MD, Ilodigwe D, Macdonald RL. Cerebral infarction after subarachnoid hemorrhage contributes to poor outcome by vasospasm-dependent and -independent effects. Stroke. 2011;42(4):924–9. doi:10.1161/STROKEAHA.110.597914. Bruder N, Rabinstein A, Participants in the International MultiDisciplinary Consensus Conference on the Critical Care Management of Subarachnoid H. Cardiovascular and pulmonary complications of aneurysmal subarachnoid hemorrhage. Neurocrit Care. 2011;15(2):257–69. doi:10.1007/s12028-011-9598-4. Larsen CC, Astrup J. Rebleeding after aneurysmal subarachnoid hemorrhage: a literature review. World Neurosurg. 2013;79(2): 307–12. doi:10.1016/j.wneu.2012.06.023. Macdonald RL. Delayed neurological deterioration after subarachnoid haemorrhage. Nat Rev Neurol. 2014;10(1):44–58. doi:10. 1038/nrneurol.2013.246. Fujii M, Yan J, Rolland WB, Soejima Y, Caner B, Zhang JH. Early brain injury, an evolving frontier in subarachnoid hemorrhage research. Transl Stroke Res. 2013;4(4):432–46. doi:10.1007/s12975013-0257-2. Etminan N, Beseoglu K, Eicker SO, Turowski B, Steiger HJ, Hanggi D. Prospective, randomized, open-label phase II trial on concomitant intraventricular fibrinolysis and low-frequency rotation after severe subarachnoid hemorrhage. Stroke. 2013;44(8): 2162–8. doi:10.1161/STROKEAHA.113.001790. Dreier JP, Korner K, Ebert N, Gorner A, Rubin I, Back T, et al. Nitric oxide scavenging by hemoglobin or nitric oxide synthase inhibition by N-nitro-L-arginine induces cortical spreading ischemia when K+ is increased in the subarachnoid space. J Cereb Blood Flow Metab. 1998;18(9):978–90. doi:10.1097/00004647199809000-00007. Dreier JP, Major S, Manning A, Woitzik J, Drenckhahn C, Steinbrink J, et al. Cortical spreading ischaemia is a novel process involved in ischaemic damage in patients with aneurysmal subarachnoid haemorrhage. Brain. 2009;132(Pt 7):1866–81. doi:10. 1093/brain/awp102. Etminan N, Vergouwen MD, Macdonald RL. Angiographic vasospasm versus cerebral infarction as outcome measures after aneurysmal subarachnoid hemorrhage. Acta Neurochir Suppl. 2013;115:33–40. doi:10.1007/978-3-7091-1192-5_8. Friedrich B, Muller F, Feiler S, Scholler K, Plesnila N. Experimental subarachnoid hemorrhage causes early and longlasting microarterial constriction and microthrombosis: an in-vivo microscopy study. J Cereb Blood Flow Metab. 2012;32(3):447–55. doi:10.1038/jcbfm.2011.154. Park KW, Metais C, Dai HB, Comunale ME, Sellke FW. Microvascular endothelial dysfunction and its mechanism in a rat model of subarachnoid hemorrhage. Anesth Analg. 2001;92(4): 990–6. Vergouwen MD, Vermeulen M, Coert BA, Stroes ES, Roos YB. Microthrombosis after aneurysmal subarachnoid hemorrhage: an additional explanation for delayed cerebral ischemia. J Cereb Blood Flow Metab. 2008;28(11):1761–70. doi:10.1038/jcbfm. 2008.74. Vergouwen MD, Vermeulen M, van Gijn J, Rinkel GJ, Wijdicks EF, Muizelaar JP, et al. Definition of delayed cerebral ischemia after aneurysmal subarachnoid hemorrhage as an outcome event in clinical trials and observational studies: proposal of a multidisciplinary research group. Stroke. 2010;41(10):2391–5. doi:10.1161/ STROKEAHA.110.589275. Yundt KD, Grubb Jr RL, Diringer MN, Powers WJ. Autoregulatory vasodilation of parenchymal vessels is impaired during cerebral vasospasm. J Cereb Blood Flow Metab. 1998;18(4):419–24. doi: 10.1097/00004647-199804000-00010. Brathwaite S, Macdonald RL. Current management of delayed cerebral ischemia: update from results of recent clinical trials. Transl Stroke Res. 2014;5(2):207–26. doi:10.1007/s12975-013-0316-8.

19.

Etminan N, Vergouwen MD, Ilodigwe D, Macdonald RL. Effect of pharmaceutical treatment on vasospasm, delayed cerebral ischemia, and clinical outcome in patients with aneurysmal subarachnoid hemorrhage: a systematic review and meta-analysis. J Cereb Blood Flow Metab. 2011;31(6):1443–51. doi:10.1038/jcbfm. 2011.7. 20. Macdonald RL, Higashida RT, Keller E, Mayer SA, Molyneux A, Raabe A, et al. Clazosentan, an endothelin receptor antagonist, in patients with aneurysmal subarachnoid haemorrhage undergoing surgical clipping: a randomised, double-blind, placebo-controlled phase 3 trial (CONSCIOUS-2). Lancet Neurol. 2011;10(7):618–25. 21. Vergouwen MD. Vasospasm versus delayed cerebral ischemia as an outcome event in clinical trials and observational studies. Neurocrit Care. 2011;15(2):308–11. doi:10.1007/s12028-011-9586-8. 22. Vergouwen MD, Etminan N, Ilodigwe D, Macdonald RL. Lower incidence of cerebral infarction correlates with improved functional outcome after aneurysmal subarachnoid hemorrhage. J Cereb Blood Flow Metab. 2011;31(7):1545–53. doi:10.1038/jcbfm. 2011.56. 23. Chen S, Ma Q, Krafft PR, Chen Y, Tang J, Zhang J, et al. P2X7 receptor antagonism inhibits p38 mitogen-activated protein kinase activation and ameliorates neuronal apoptosis after subarachnoid hemorrhage in rats. Crit Care Med. 2013;41(12):e466–74. doi:10. 1097/CCM.0b013e31829a8246. 24. Chou SH, Feske SK, Simmons SL, Konigsberg RG, Orzell SC, Marckmann A, et al. Elevated peripheral neutrophils and matrix metalloproteinase 9 as biomarkers of functional outcome following subarachnoid hemorrhage. Transl Stroke Res. 2011;2(4):600–7. doi:10.1007/s12975-011-0117-x. 25. Fujii M, Sherchan P, Soejima Y, Hasegawa Y, Flores J, Doycheva D, et al. Cannabinoid receptor type 2 agonist attenuates apoptosis by activation of phosphorylated CREB-Bcl-2 pathway after subarachnoid hemorrhage in rats. Exp Neurol. 2014;261:396–403. doi:10.1016/j.expneurol.2014.07.005. 26. Murakami K, Koide M, Dumont TM, Russell SR, Tranmer BI, Wellman GC. Subarachnoid hemorrhage induces gliosis and increased expression of the pro-inflammatory cytokine high mobility group box 1 protein. Transl Stroke Res. 2011;2(1):72–9. doi:10. 1007/s12975-010-0052-2. 27. Shao AW, Wu HJ, Chen S, Ammar AB, Zhang JM, Hong Y. Resveratrol attenuates early brain injury after subarachnoid hemorrhage through inhibition of NF-kappaB-dependent inflammatory/ MMP-9 pathway. CNS Neurosci Ther. 2014;20(2):182–5. doi:10. 1111/cns.12194. 28. Shiba M, Fujimoto M, Imanaka-Yoshida K, Yoshida T, Taki W, Suzuki H. Tenascin-C causes neuronal apoptosis after subarachnoid hemorrhage in rats. Transl Stroke Res. 2014;5(2):238–47. doi:10. 1007/s12975-014-0333-2. 29. Simard JM, Tosun C, Ivanova S, Kurland DB, Hong C, Radecki L, et al. Heparin reduces neuroinflammation and transsynaptic neuronal apoptosis in a model of subarachnoid hemorrhage. Transl Stroke Res. 2012;3 Suppl 1:155–65. doi:10.1007/s12975-0120166-9. 30. Yan J, Manaenko A, Chen S, Klebe D, Ma Q, Caner B, et al. Role of SCH79797 in maintaining vascular integrity in rat model of subarachnoid hemorrhage. Stroke. 2013;44(5):1410–7. doi:10.1161/ STROKEAHA.113.678474. 31. Etminan N, Macdonald RL, Davis C, Burton K, Steiger HJ, Hanggi D. Intrathecal application of the nimodipine slow-release microparticle system eg-1962 for prevention of delayed cerebral ischemia and improvement of outcome after aneurysmal subarachnoid hemorrhage. Acta Neurochir Suppl. 2015;120:281–6. doi:10.1007/9783-319-04981-6_47. 32. Kasuya H. Clinical trial of nicardipine prolonged-release implants for preventing cerebral vasospasm: multicenter cooperative study in

Transl. Stroke Res.

33.

34.

35.

36.

37.

38.

39.

40.

41.

42.

43.

44.

45.

46.

47.

48.

Tokyo. Acta Neurochir Suppl. 2011;110(Pt 2):165–7. doi:10.1007/ 978-3-7091-0356-2_30. Lei Q, Li S, Zheng R, Xu K, Li S. Endothelin-1 expression and alterations of cerebral microcirculation after experimental subarachnoid hemorrhage. Neuroradiology. 2015;57(1):63–70. doi:10.1007/ s00234-014-1435-y. Tso MK, Macdonald RL. Subarachnoid hemorrhage: a review of experimental studies on the microcirculation and the neurovascular unit. Transl Stroke Res. 2014;5(2):174–89. doi:10.1007/s12975014-0323-4. Buhler D, Schuller K, Plesnila N. Protocol for the induction of subarachnoid hemorrhage in mice by perforation of the Circle of Willis with an endovascular filament. Transl Stroke Res. 2014;5(6): 653–9. doi:10.1007/s12975-014-0366-6. Guresir E, Schuss P, Borger V, Vatter H. Experimental subarachnoid hemorrhage: double cisterna magna injection rat model-assessment of delayed pathological effects of cerebral vasospasm. Transl Stroke Res. 2015. doi:10.1007/s12975-015-0392-z. Kikkawa Y, Kurogi R, Sasaki T. The single and double blood injection rabbit subarachnoid hemorrhage model. Transl Stroke Res. 2015;6(1):88–97. doi:10.1007/s12975-014-0375-5. Mori K. Double cisterna magna blood injection model of experimental subarachnoid hemorrhage in dogs. Transl Stroke Res. 2014;5(6):647–52. doi:10.1007/s12975-014-0356-8. Pluta RM, Bacher J, Skopets B, Hoffmann V. A non-human primate model of aneurismal subarachnoid hemorrhage (SAH). Transl Stroke Res. 2014;5(6):681–91. doi:10.1007/s12975-014-0371-9. Baharoglu MI, Germans MR, Rinkel GJ, Algra A, Vermeulen M, van Gijn J, et al. Antifibrinolytic therapy for aneurysmal subarachnoid haemorrhage. Cochrane Database Syst Rev. 2013;8, CD001245. doi:10.1002/14651858.CD001245.pub2. Germans MR, Post R, Coert BA, Rinkel GJ, Vandertop WP, Verbaan D. Ultra-early tranexamic acid after subarachnoid hemorrhage (ULTRA): study protocol for a randomized controlled trial. Trials. 2013;14:143. doi:10.1186/1745-6215-14-143. Cremers CH, van der Schaaf IC, Wensink E, Greving JP, Rinkel GJ, Velthuis BK, et al. CT perfusion and delayed cerebral ischemia in aneurysmal subarachnoid hemorrhage: a systematic review and meta-analysis. J Cereb Blood Flow Metab. 2014;34(2):200–7. doi:10. 1038/jcbfm.2013.208. Rawal S, Barnett C, John-Baptiste A, Thein HH, Krings T, Rinkel GJ. Effectiveness of diagnostic strategies in suspected delayed cerebral ischemia: a decision analysis. Stroke. 2015;46(1):77–83. doi: 10.1161/STROKEAHA.114.005916. Washington CW, Zipfel GJ. Detection and monitoring of vasospasm and delayed cerebral ischemia: a review and assessment of the literature. Neurocrit Care. 2011;15(2):312–7. doi:10.1007/ s12028-011-9594-8. Etminan N, Beseoglu K, Heiroth HJ, Turowski B, Steiger HJ, Hanggi D. Early perfusion computerized tomography imaging as a radiographic surrogate for delayed cerebral ischemia and functional outcome after subarachnoid hemorrhage. Stroke. 2013;44(5): 1260–6. doi:10.1161/STROKEAHA.111.675975. Dhar R, Scalfani MT, Zazulia AR, Videen TO, Derdeyn CP, Diringer MN. Comparison of induced hypertension, fluid bolus, and blood transfusion to augment cerebral oxygen delivery after subarachnoid hemorrhage. J Neurosurg. 2012;116(3):648–56. doi: 10.3171/2011.9.JNS11691. Treggiari MM, Deem S. Which H is the most important in triple-H therapy for cerebral vasospasm? Curr Opin Crit Care. 2009;15(2): 83–6. doi:10.1097/MCC.0b013e32832922d1. Treggiari MM, Participants in the International Multi-disciplinary Consensus Conference on the Critical Care Management of Subarachnoid H. Hemodynamic management of subarachnoid

hemorrhage. Neurocrit Care. 2011;15(2):329–35. doi:10.1007/ s12028-011-9589-5. 49. Abruzzo T, Moran C, Blackham KA, Eskey CJ, Lev R, Meyers P, et al. Invasive interventional management of post-hemorrhagic cerebral vasospasm in patients with aneurysmal subarachnoid hemorrhage. J Neurointerv Surg. 2012;4(3):169–77. doi:10.1136/ neurintsurg-2011-010248. 50. Dorhout Mees SM, Rinkel GJ, Feigin VL, Algra A, van den Bergh WM, Vermeulen M, et al. Calcium antagonists for aneurysmal subarachnoid haemorrhage. Cochrane Database Syst Rev. 2007;3, CD000277. doi:10.1002/14651858.CD000277.pub3. 51. Kirkpatrick PJ, Turner CL, Smith C, Hutchinson PJ, Murray GD, Collaborators S. Simvastatin in aneurysmal subarachnoid haemorrhage (STASH): a multicentre randomised phase 3 trial. Lancet Neurol. 2014;13(7):666–75. doi:10.1016/S1474-4422(14)70084-5. 52. Wong GK, Chan DY, Siu DY, Zee BC, Poon WS, Chan MT, et al. High-dose simvastatin for aneurysmal subarachnoid hemorrhage: multicenter randomized controlled double-blinded clinical trial. Stroke. 2015;46(2):382–8. doi:10.1161/STROKEAHA.114. 007006. 53. Kreiter KT, Mayer SA, Howard G, Knappertz V, Ilodigwe D, Sloan MA, et al. Sample size estimates for clinical trials of vasospasm in subarachnoid hemorrhage. Stroke. 2009;40(7):2362–7. doi:10. 1161/STROKEAHA.109.547331. 54. Jaja BN, Attalla D, Macdonald RL, Schweizer TA, Cusimano MD, Etminan N, et al. The Subarachnoid Hemorrhage International Trialists (SAHIT) Repository: advancing clinical research in subarachnoid hemorrhage. Neurocrit Care. 2014;21(3):551–9. doi:10. 1007/s12028-014-9990-y. 55. Macdonald RL, Cusimano MD, Etminan N, Hanggi D, Hasan D, Ilodigwe D, et al. Subarachnoid Hemorrhage International Trialists data repository (SAHIT). World Neurosurg. 2013;79(3–4):418–22. doi:10.1016/j.wneu.2013.01.006. 56. Macdonald RL, Jaja B, Cusimano MD, Etminan N, Hanggi D, Hasan D, et al. SAHIT Investigators—on the outcome of some subarachnoid hemorrhage clinical trials. Transl Stroke Res. 2013;4(3):286–96. doi:10.1007/s12975-012-0242-1. 57. Al-Khindi T, Macdonald RL, Schweizer TA. Cognitive and functional outcome after aneurysmal subarachnoid hemorrhage. Stroke. 2010;41(8):e519–36. doi:10.1161/STROKEAHA.110.581975. 58. Sheldon S, Macdonald RL, Cusimano M, Spears J, Schweizer TA. Long-term consequences of subarachnoid hemorrhage: examining working memory. J Neurol Sci. 2013;332(1–2):145–7. doi:10. 1016/j.jns.2013.06.021. 59. Sheldon S, Macdonald RL, Schweizer TA. Free recall memory performance after aneurysmal subarachnoid hemorrhage. J Int Neuropsychol Soc : JINS. 2012;18(2):334–42. doi:10.1017/ S1355617711001780. 60. Hanggi D, Etminan N, Macdonald RL, Steiger HJ, Mayer SA, Aldrich F, et al. NEWTON: Nimodipine Microparticles to Enhance Recovery While Reducing Toxicity After Subarachnoid Hemorrhage. Neurocrit Care. 2015. doi:10. 1007/s12028-015-0112-2. 61. Rasmussen R, Juhler M, Wetterslev J. Effects of continuous prostacyclin infusion on regional blood flow and cerebral vasospasm following subarachnoid haemorrhage: statistical analysis plan for a randomized controlled trial. Trials. 2014;15:228. doi:10.1186/ 1745-6215-15-228. 62. Westermaier T, Stetter C, Vince GH, Pham M, Tejon JP, Eriskat J, et al. Prophylactic intravenous magnesium sulfate for treatment of aneurysmal subarachnoid hemorrhage: a randomized, placebo-controlled, clinical study. Crit Care Med. 2010;38(5):1284–90. doi:10. 1097/CCM.0b013e3181d9da1e.

Aneurysmal subarachnoid hemorrhage--status quo and perspective.

Aneurysmal subarachnoid hemorrhage--status quo and perspective. - PDF Download Free
180KB Sizes 0 Downloads 13 Views