Surgical decompression for space-occupying hemispheric infarction Jeannette Hofmeijer ISBN: Cover: Lay out: Printed by: 978-90-393-4487-3 Roy Sanders (based on Wieslaw Rosocha: Shakespeare’s HAMLET, Polish theatre, 1996) Roy Sanders Gildeprint drukkerijen BV Surgical decompression for space-occupying hemispheric infarction Chirurgische decompressie voor het ruimte-innemende supra-tentoriële herseninfarct (met een samenvatting in het Nederlands) Proefschrift ter verkrijging van de graad van doctor aan de Universiteit Utrecht op gezag van de rector magnificus, prof.dr. W.H. Gispen, ingevolge het besluit van het college voor promoties in het openbaar te verdedigen op donderdag 19 april 2007 des middags te 12.45 uur door Jeannette Hofmeijer geboren op 20 december 1971 te Ermelo Promotor: Prof. dr. L.J. Kappelle Co-promotor: Dr. H.B. van der Worp Research described in this thesis was supported by a grant of the Netherlands Heart Foundation (grant number 2002B138). Financial support by the Netherlands Heart Foundation for the publication of this thesis is gratefully acknowledged. Financial support for the publication of this thesis by Boehringer Ingelheim is gratefully acknowledged. Everything is what it is, because it got that way. Alles is wat het is, omdat het zo geworden is. (‘d Arcey W. Thompson. On growth and form. 1917.) Aan mijn vader Contents Chapter 1 General introduction 9 Part I. Experimental studies Chapter 2 Treatment of space-occupying middle cerebral artery infarction 17 Chapter 3 The time course of ischemic damage and perfusion in a rat model of space-occupying cerebral infarction 41 Chapter 4 Perfusion MRI by flow-sensitive alternating inversion recovery and dynamic susceptibility contrast in rats with permanent middle cerebral artery occlusion 57 Chapter 5 Delayed decompressive surgery improves peri-infarct perfusion in rats with space-occupying cerebral infarction 67 Part II. Clinical studies Chapter 6 Predictors of life-threatening brain edema in middle cerebral artery infarction 83 Chapter 7 A new algorithm for sequential allocation of two treatments in small clinical trials 99 Chapter 8 Hemicraniectomy After Middle cerebral artery infarctionwithLife-threateningEdemaTrial(HAMLET). Protocol for a randomized controlled trial of decompressive surgery in space-occupying hemispheric infarction 111 Chapter 9 Appreciation of the informed consent procedure in a randomized trial of decompressive surgery for space-occupying hemispheric infarction 123 Chapter 10 Long-term cognitive outcome after decompressive surgery for space-occupying hemispheric infarction 133 Chapter 11 Early decompressive surgery in space-occupying hemispheric infarction: a pooled analysis of three randomized controlled trials 143 Chapter 12 General discussion 159 Appendix Summary Samenvatting Dankwoord Curriculum vitae Publications 169 171 175 181 185 187 Introduction Chapter 1 General introduction 9 Chapter 1 I n 1999 a 32 year old woman was admitted because of weakness of the left arm and left leg. She had no medical history, worked as a school teacher, and had two young children. On neurological examination she had a normal consciousness but a severe left-sided hemiparesis. CT showed infarction of the right anterior and middle cerebral artery (MCA) territory and a dissection of the right internal carotid artery (Figure 1). Twenty-four hours after the onset of symptoms she deteriorated with a decrease in consciousness and dilatation of the right pupil. A CT scan revealed massive space-occupying edema with tissue shift across the midline (Figure 2). Decompressive surgery was considered, but not performed, because of uncertainty about the benefit. Her neurological condition deteriorated, despite medical therapy on the stroke unit and she died as a result of transtentorial herniation 48 hours after the onset of symptoms. Preface Large cerebral infarcts may be associated with space-occupying edema, that may lead to transtentorial or uncal herniation.1 Fatal space-occupying brain edema occurs in 1 to 5% of all patients with a supratentorial infarct.2,3 Case fatality rates of up to 80% have been reported, despite maximal medical therapy on an intensive care unit.1,4 Different treatment modalities have been suggested for patients with supratentorial infarcts who deteriorate as a result of edema formation. None of these therapies has been proven to improve clinical outcome.5 According to the guidelines of the American Heart Association, these patients may be treated with osmotic agents and hyperventilation.6 However, several reports suggest that these measures are ineffective1,4,7 or even detrimental.8 Because of the limitations of medical therapies, there have been proposals for decompressive surgery in patients with neurological deterioration due to large hemispheric infarction and edema (Figure 3). The rationale of this therapy is to revert brain tissue shifts and to normalize intracranial pressure, thereby preserving cerebral blood flow and preventing secondary damage.9 Decompressive surgery is not a new modality for the treatment of patients with an intracranial mass.10 The technique is relatively simple and consists of a large hemicraniectomy and a duraplasty. It can be performed in every neurosurgical center. Case reports and non-randomized patient series have suggested a substantial reduction of mortality after decompressive surgery without an increase in the number of severely disabled survivors.11-14 However, information on functional outcome and quality of life of the surviving patients is insufficient.13,14 The work presented in this thesis consists of the preparation and preliminary results of a randomized clinical trial to study the effect of decompressive surgery on functional outcome in patients with hemispheric infarction, who deteriorate as a result of brain edema formation. 10 Introduction Figure 1 CT scan of a 32 year old patient with a large infarct in the territory of the right anterior and middle cerebral arteries one hour after the onset of symptoms. Figure 2 CT scan of the same patient 24 hours after the onset of symptoms, showing space-occupying edema and midline shift. Figure 3 CT scan of a 44 year old patient with a large infarct in the territory of the left anterior and middle cerebral arteries after decompressive surgery. 11 Chapter 1 Outline of the thesis In chapter 2 the existing evidence of efficacy of treatment modalities for spaceoccupying hemispheric infarction is summarized. In chapter 3 a rat model of spaceoccupying cerebral infarction is presented. By means of this model the time course of ischemic damage and perfusion as measured by magnetic resonance imaging (MRI) are evaluated (chapter 3), brain perfusion is studied and non-invasive and invasive MR perfusion measurements are compared (chapter 4), and the effects of decompressive surgery on tissue damage and perfusion are investigated (chapter 5). In chapter 6 a meta-analysis on predictors of fatal edema formation is presented. In chapter 7 an alternative way of randomizing patients is described, useful for relatively small clinical trials. Chapter 8 contains the study protocol of the ongoing multi-center randomized Hemicraniectomy After MCA infarction with Life-threatening Edema Trial (HAMLET). The results of studies on recall and appreciation of the informed consent procedure in HAMLET (chapter 9) and of cognitive outcome of a cohort of patients after space-occupying MCA infarction and hemicraniectomy (chapter 10) are presented. Chapter 11 brings the results of a pooled analysis of individual patient data of three European trials on decompressive surgery after space-occupying hemispheric infarction. In chapter 12 the implications of the studies described in this thesis on patient care and future research are discussed. References 1. 2. 3. 4. 5. 6. 7. 8. 9. 12 Hacke W, Schwab S, Horn M, Spranger M, De Georgia M, von Kummer R. ‘Malignant’ middle cerebral artery territory infarction: clinical course and prognostic signs. Arch Neurol 1996;53:309-315. Silver FL, Norris JW, Lewis AJ, Hachinski VC. Early mortality following stroke: a prospective review. Stroke 1984;15:492-496. Heinsius T, Bogousslavsky J, Van Melle G. Large infarcts in the middle cerebral artery territory. Etiology and outcome patterns. Neurology 1998;50:341-350. Wijdicks EF, Diringer MN. Middle cerebral artery territory infarction and early brain swelling: progression and effect of age on outcome. Mayo Clin Proc 1998;73:829-836. Hofmeijer J, van der Worp HB, Kappelle LJ. Treatment of space-occupying cerebral infarction. Crit Care Med 2003;31:617-625. Adams Jr HP., Adams RJ, Brott T. Guidelines for the early management of patients with ischemic stroke: A scientific statement from the Stroke Council of the American Stroke Association. Stroke 2003;34:10561083. Schwab S, Spranger M, Schwarz S, Hacke W. Barbiturate coma in severe hemispheric stroke: useful or obsolete? Neurology 1997;48:1608-1613. Muizelaar JP, Marmarou A, Ward JD. Adverse effects of prolonged hyperventilation in patients with severe head injury: a randomized clinical trial. J Neurosurg 1991;75:731-739. Schwab S, Rieke K, Aschoff A, Albert F, von Kummer R, Hacke W. Hemicraniotomy in space-occupying hemispheric infarction: useful early intervention or desparate activism? 1996;6:325-329. Introduction 10. 11. 12. 13. 14. Kerr FW. Radical decompression and dural grafting in severe cerebral edema. Mayo Clin Proc 1968;43:852-864. Koh MS, Goh KY, Tung MY, Chan C. Is decompressive craniectomy for acute cerebral infarction of any benefit? Surg Neurol 2000;53:225-230. Carter BS, Ogilvy CS, Candia GJ, Rosas HD, Buonanno F. One-year outcome after decompressive surgery for massive nondominant hemispheric infarction. Neurosurgery 1997;40:1168-1175. Rieke K, Schwab S, Krieger D. Decompressive surgery in space-occupying hemispheric infarction: results of an open, prospective trial. Crit Care Med 1995;23:1576-1587. Schwab S, Steiner T, Aschoff A. Early hemicraniectomy in patients with complete middle cerebral artery infarction. Stroke 1998;29:1888-1893. 13 Chapter 1 14 Treatment of space-occupying MCA infarction Part I Experimental studies 15 Treatment of space-occupying MCA infarction Chapter 2 Treatment of space-occupying middle cerebral artery infarction Jeannette Hofmeijer, H. Bart van der Worp, and L. Jaap Kappelle Based on Critical Care Medicine 2003;31:617-625 17 Chapter 2 Summary Patients with a hemispheric infarct and massive edema have a poor prognosis. The case fatality rate is approximately 80% and most survivors are left severely disabled. Various treatment strategies have been proposed to limit brain tissue shifts and to reduce intracranial pressure, but their use is controversial. In this chapter the results of a systematic review of the literature addressing the evidence of efficacy of these treatments is presented. Literature searches were performed in Medline and PubMed. Studies were included if they were published in English between Januari 1966 and August 2006 and addressed the effect of osmotherapy, hyperventilation, barbiturates, steroids, hypothermia, or decompressive surgery in supratentorial infarction with edema in animals or humans. Animal studies of medical treatment strategies in focal cerebral ischemia produced conflicting results. If any, experimental support for these strategies is derived from studies with animal models of moderately severe focal ischemia instead of severe space-occupying infarction. None of the treatment options has improved outcome in randomized clinical trials. Two large non-randomized studies of decompressive surgery yielded promising results in terms of reduction of mortality and improvement of functional outcome. There is no treatment modality of proven efficacy for patients with space-occupying hemispheric infarction. Decompressive surgery might be the most effective therapeutic option. For decisive answers randomized controlled clinical trials are needed. 18 Treatment of space-occupying MCA infarction L arge cerebral infarcts are commonly associated with variable degrees of brain edema. In severe cases this may lead to transtentorial or uncal herniation. Fatal space-occupying brain edema occurs in 1-5% of all patients with a supratentorial infarct.1 Transtentorial herniation accounts for up to 78% of deaths during the first week after supratentorial infarction and up to 27% of deaths during the first 30 days.1,2 In younger patients with ischemic stroke, herniation is the cause of about half of the deaths in the first month.3 In recent prospective intensive care-based series the case fatality rate of space-occupying cerebral infarcts was about 80%, despite maximal conservative therapy.4,5 Various treatment strategies have been proposed to limit brain tissue shifts and to reduce intracranial pressure, such as osmotic therapy, hyperventilation, and sedation with barbiturates.6-8 In the guidelines of the American Heart Association osmotherapy and hyperventilation are recommended for patients whose condition is deteriorating secondary to increased intracranial pressure or brain herniation.6 These treatment options are considered standard care by experts in various stroke centers worldwide. However, no trials have addressed the efficacy of these therapies to improve clinical outcome,9 and several reports suggest that these are ineffective4,5,10 or even detrimental.11,12 In this chapter the evidence of efficacy of treatments that have been proposed to improve outcome after space-occupying hemispheric infarction will be reviewed. Methods Literature searches were carried out in Medline and PubMed, using a combination of keywords covering brain infarction, brain edema, and the different options of intervention. Keywords related to brain infarction were stroke, cerebral infarction, and cerebral ischemia, and the alternatives presented in the Thesaurus of Medline. Keywords related to edema were brain edema, brain swelling, and their alternatives. Furthermore, relevant papers were checked for references. Studies were included if they were published in English between Januari 1966 and August 2006 and addressed treatment of edema in supratentorial focal cerebral ischemia in animals (Table 1) or humans (Table 2). Treatment modalities that had only been used in animals, but not in humans, were excluded. 19 20 Hoff, 1982 Albright, 1984 Millson, 1981 Selman, 1982b Kobayashi, 1995 Kotwica, 1991a Selman, 1981b Kobayashi, 1994 Selman, 1982b Selman, 1981b Corkill, 1978 Karibe, 1995 Harbaugh, 1979 Tuor, 1999 Slivka, 2001c Moseley, 1975 Albright, 1984 Tuor, 1993 de Courten, 1994 Hoff, 1975 Little, 1980 Chumas, 1993 Barks, 1991 Braughler, 1986 Little, 1979 Tosaki, 1985 Steroids Smith, 1974 Barbiturates Hoff, 1973 Paczynski, 2000 Albright, 1984 Furosemide Little, 1978 Suzuki, 1980 Suzuki, 1980 Hypertonic Saline Little, 1978 Popovic, 1978 Dietis, 1986 Dodson, 1975 Karibe, 1995 Paczynski, 2000 Meyer, 1972 Ehteshami, 1988 Paczynski, 1997 Glycerol Mannitol Table 1 Summary of experimental studies on treatment of hemispheric infarction. ICP lowering Reduced ischemic damage Reduced edema Treatment result Chapter 2 a Steroids Donley, 1973 Barbiturates Hoff, 1982 Increased volume ratios/shift Increased ICP indicates rebound phenomenon; b, with reperfusion and if therapy was initiated within 30 minutes after ischemia; c, only after reperfusion Paczynski, 2000 Paczynski, 1997 Koide, 1986 Increased edema Increased infarct volume Kaufmann, 1992 Sapolsky, 1985 Accumulation in infarcted tissue No effect on histopathology Treatment result Kaufmann, 1992 Oktem, 2000 Bhardwaj, 2000 de la Torre, 1976 Furosemide Gueniau, 1997 Bhardwaj, 2000 Hypertonic Saline Lee, 1974 Glycerol Ogilvy, 1996 Koc, 1994 Mannitol Table 1 – Continued – Treatment of space-occupying MCA infarction 21 22 a Patten, 1972 Steroids a indicates temporary effect with reduction of cerebral perfusion pressure Norris, 1976 a’Rogvi, 2000 Ogun, 2001 Norris, 1986 Santambrogio, 1978 Mulley, 1978 Kaste, 1976 Yu, 1993 Santambrogio, 1978 Bauer, 1973 Larsson, 1976 Candalise, 1975 Bayer, 1987 Fawer, 1978 Rockoff, 1979 Schwab, 1997 Barbiturates ICP lowering Treatment result No effect on outcome Improved outcome Increased volume ratios Furosemide Videen, 2001 Schwarz, 1998 Hypertonic Saline No effect tissue shift Mathew, 1972 Glycerol Manno, 1999 Schwarz, 1998 Mannitol Table 2 Summary of clinical studies on treatment of hemispheric infarction. Chapter 2 Treatment of space-occupying MCA infarction Results Osmotherapy Mannitol Osmotic agents, such as mannitol, are presumed to draw water from interstitial and intracellular spaces into the intravascular compartment by creating an osmotic pressure gradient over the semi-permeable blood-brain barrier.13 In addition to its osmotic capacity, reported effects of mannitol include reduction of blood viscosity and improvement of microvascular cerebral blood flow,14-17 vasoconstriction with a reduction in cerebral blood volume,18,19 and scavenging of free radicals.20 In various animal studies, mannitol, administered before or within 24 hours after the onset of transient or permanent focal cerebral ischemia reduced edema formation or ischemic damage.21-31 In a recent study, a trend towards a dose-dependent decrease in the water content of the ischemic middle cerebral artery (MCA) cortex and infarcted hemisphere was found. However, relatively more dehydration of the non-infarcted hemisphere than of the hemisphere with the infarction was seen in rats receiving a high dose (2.5mg / kg) than in rats receiving a low dose (0.5mg / kg) of mannitol, suggesting that more severe hyperosmolar dehydration may increase pressure gradients and aggravate tissue shifts.30 In a subsequent study, high doses of mannitol (1.5g / kg every five hours) caused paradoxical increases in the water content of the infarcted hemisphere, in the infarcted / non-infarcted hemisphere volume ratios, and in midline shift. In contrast, low doses of mannitol had significant positive effects on these variables.31 Other experimental studies failed to show a statistically significant positive effect of mannitol therapy on infarct volume32-35 or cerebral edema.36 The lack of efficacy might be the result of continuous infusion instead of bolus administration35 or of administration before rather than after the ischemic insult, so that the osmotic gradient had already disappeared before the occurrence of edema.32 Other proposed reasons for the lack of effect include relatively late administration36 or administration of too low doses of mannitol (0.2g / kg).33 Several authors have suggested that administration of hypertonic agents in the presence of cerebral ischemic injury may be detrimental.12,35 To create an osmotic gradient, an intact blood-brain barrier is needed. As the blood-brain barrier is compromised during cerebral ischemia, it is presumed that water will be extracted from healthy but not from ischemic brain tissue,37 resulting in a worsening of tissue shifts. In cats with cortical cold injury, accumulation of mannitol in damaged brain tissue has been reported after 5 doses of 0.33 g/kg. In the edematous white matter the mannitol concentration exceeded the plasma concentration by a ratio of 2.69:1. This caused a reversal of the osmotic gradient and aggravation of cerebral edema.12 In other 23 Chapter 2 animal studies, rebound phenomena have been observed.37 These have been attributed to the longer elimination half-life of mannitol from cerebro-spinal fluid (CSF) than from blood, with a consequent temporary reversal of the serum / CSF concentration gradient during elimination.38,39 In a clinical study in 9 patients with recent ischemic stroke, single doses of 40g mannitol were effective in temporarily reducing elevated ICP to more than 10% below the baseline value in 10 of 14 episodes. The maximum effect occurred at the end of infusion and the effect was visible over 4 hours.8 In a recent series of seven patients with edema and midline shift due to hemispheric infarction, successive magnetic resonance imaging (MRI) before, during, and after the administration of a bolus of mannitol (1,5g / kg) did not reveal any change in midline shifts, nor did the neurological status of the patients change. Although these findings dispel some of the concerns of increases in mass effect after administration of mannitol, the clinical implications concerning either beneficial or harmfull effects are limited, since the study did not address the effects of multiple dosing.40 Furthermore, subsequent analyses of hemispheric volumes of these patients revealed that there was a slight reduction in brain volume after mannitol treatment that was restricted to the non-infarcted hemisphere.41 No randomized clinical trial has addressed the effect of mannitol on outcome in patients with space-occupying hemispheric infarction. One early prospective42 and one retrospective43 clinical study failed to show a significant benefit on outcome in patients with acute stroke. However, these studies were not CT based and cases of cerebral hemorrhage could therefore have been included inadvertently. In addition, clinically different infarct subtypes were included, and treatment was not primarily aimed at reducing edema formation in large infarcts. In a systematic (Cochrane) review outcome analyses could not be performed due to lack of appropriate trials.44 Glycerol Glycerol has been reported to improve cerebral blood flow,45,46 and to have edemareducing as well as neuroprotective properties.47 Only few experimental studies have addressed the effect of glycerol in cerebral infarction. In rat models of focal cerebral ischemia the compound reduced edema.48-51 A single bolus decreased ICP52 or T2 hyperintensity on MRI53 rapidly in patients with MCA infarction, but accumulated in infarcted tissue.52 Glycerol has been tested in several randomized and non-randomized clinical trials of acute stroke, but none of these specifically addressed its effect on space-occupying hemispheric infarction. A systematic (Cochrane) review of these trials suggests a favorable effect of glycerol treatment on short term survival, but no long term efficacy. The lack of proven benefit on long term survival does not support the routine use of glycerol in patients with acute ischemic stroke.54 24 Treatment of space-occupying MCA infarction Hypertonic saline Sodium chloride is actively excluded from an intact blood-brain barrier, which theoretically makes it a more potent osmotic agent than mannitol.55 In recent studies of transient focal cerebral ischemia in rats, edema in both the affected and the unaffected hemisphere decreased after continuous hypertonic (7.5%) saline infusion started 6 or 24 hours after induction of ischemia respectively.56,57 In a comparable study hypertonic saline increased rather than decreased infarct volume. Water content in the contralateral (non-injured) hemisphere was significantly less in the hypertonic salinetreated group than in the control group at 22 hours of reperfusion, whereas there was no difference in water content of the injured hemisphere between the two groups.35 In patients with space-occupying hemispheric infarction or putaminal hemorrhage with perifocal edema, single doses of hypertonic saline temporarily reduced elevated ICP.8 A temporary reduction of elevated ICP, leading to an increased cerebral perfusion pressure, was also seen in eight patients treated with 75ml 10% saline after conventional therapy with mannitol had failed. However, the group of patients described in this study was heterogeneous; six had space-occupying hemispheric infarction and two had supra-tentorial hemorrhage with edema.58 In addition, patients received different combinations of other ICP lowering therapies (two with decompressive surgery, four with hypothermia, and two with CSF drainage via an intraventricular catheter), which makes the results difficult to interpret. No clinical trials have addressed the effect of hypertonic saline on functional outcome. Furosemide Loop diuretics such as furosemide may decrease ICP by decreasing total body water and increasing blood osmolality, and thereby removing water from the edematous brain.59,60 In two studies of experimental brain edema induced by cortical freezing in rabbits, furosemide significantly decreased ICP.61,62 In rats with transient focal cerebral ischemia, furosemide (0.5mg / kg 5 hourly) reduced body weight, but had no significant effect on the water content of the affected hemisphere.31 There have been no controlled clinical studies testing the effect of loop diuretics on outcome after ischemic stroke. Barbiturates Barbiturates may have neuroprotective properties by reducing the cerebral metabolic rate63-65 and by acting as free radical scavengers.66,67 Reduction of the cerebral metabolic rate and the subsequent lowering of cerebral blood flow and volume could theoretically reduce edema formation and lower ICP. Barbiturate treatment ameliorated the clinical course and reduced lesion size in 25 Chapter 2 some animal studies of focal cerebral ischemia68-74 but had no effect on edema and ICP in experimental space-occupying cerebral infarction.74-76 In baboons with permanent MCA occlusion fatal ICP elevation occurred even more often after barbiturate treatment than in controls. On the other hand, barbiturates reduced mortality if ischemia was transient and if treatment was initiated within 30 minutes after the onset of ischemia, before edema formation had occurred.73,77 Case studies suggested that barbiturate treatment may be effective to reduce intracranial hypertension caused by traumatic brain injury,78 cerebral ischemia during aneurysm surgery,79 or severe pre-eclampsia.80 However, in most clinical reports the effect of barbiturates on brain swelling secondary to infarction was disappointing.81,82 In the only prospective, but uncontrolled, clinical study on this subject the usefulness of barbiturate coma in reducing elevated ICP after MCA infarction was limited.10 Barbiturate coma was induced with thiopental infusion in 60 patients with critically increased ICP secondary to large hemispheric infarction, after failure of osmotherapy and mild hyperventilation. Although doses were high enough to achieve a burstsuppression pattern on the EEG, and ICP was significantly lowered in the early phases after initiation of therapy, long term control of ICP could not be achieved. Moreover, cerebral perfusion pressure decreased during the course of treatment. Only five of the 60 patients treated with thiopental survived; all other patients died from transtentorial herniation. Randomized trials are lacking. Steroids In very high doses, steroids have been claimed to have neuroprotective properties in ischemic stroke.83 In addition, corticosteroids reduce vasogenic cerebral edema in patients with brain tumors.84 There is no evidence from experimental studies that steroids reduce edema in cerebral infarction.85-88 This may be explained by the fact that edema in ischemic stroke consists of both a vasogenic and a cytotoxic component.89 Dexamethasone improved outcome after acute stroke in a single placebocontrolled clinical trial,90 whereas in other clinical studies no favorable effects of dexamethasone42,91-96 or prednisolone97 treatment on clinical outcome were found. Dexamethason increased short-term mortality in older patients with ischemic stroke.98 A meta-analysis of randomized trials comparing corticosteroid treatment with placebo in patients with acute ischemic stroke did not show a positive treatment effect on functional outcome.99 There are no trials addressing the efficacy of steroids to reduce edema formation in space-occupying cerebral infarction. Hyperventilation Hyperventilation lowers ICP by inducing serum and CSF alkalosis and vasoconstriction.100 However, the effect of hyperventilation may diminish within hours.101 26 Treatment of space-occupying MCA infarction Moreover, rebound vasodilatation with increases of ICP may occur if normoventilation is resumed.59 This may even induce a steal phenomenon if vasodilatation is more profound in healthy than in ischemic brain tissue.102 Several clinical studies suggested that prolonged hyperventilation induces cerebral ischemia103,104 and worsens clinical outcome in patients with traumatic brain injury.11 In primate models of focal brain ischemia, hypocapnia initiated after induction of ischemia did not alter mortality, degree of neurological deficit, or volume of the infarct.105,106 Clinical studies in the early 1970’s addressing the effect of normocapnic (40 mmHg) and hypocapnic (20-25 mmHg) hyperventilation in stroke found no beneficial treatment effect on patient outcome.107,108 In these studies hyperventilation was continued for 7274 hours. More recent clinical trials are lacking. Hypothermia Hypothermia is presumed to reduce cerebral ischemic damage by means of reducing brain metabolism,109,110 preservation of the blood brain barrier,111 a reduction in the inflammatory respons,112 and a reduced neurotransmitter release.113-115 In a variety of animal studies hypothermia reduced infarct size after focal cerebral ischemia as mono-therapy112,116-129 or additionally, next to neuroprotective agents.130132 In experimental studies of hypothermia in transient focal133,134 or global135 cerebral ischemia, a reduction of edema development during reperfusion was found. In only one early study of acute ischemic stroke in primates, hypothermia (29°C) had a detrimental effect: all treated animals had infarction with massive edema and died.106 Two non-randomized studies in patients with severe space-occupying edema after MCA infarction suggested that moderate hypothermia (32°C -34°C) on an ICU could help to control critically elevated ICP values and improve clinical outcome. Hypothermia was associated with several side effects, of which thrombocytopenia, bradycardia and pneumonia were most frequently encountered. Most deaths occurred during rewarming as a result of excessive ICP rise and herniation.136,137 A shorter rewarming period was associated with a more pronounced rise of ICP.137 Rebound ICP rise could possibly be prevented by slow and controlled, instead of passive rewarming.138 In a recent non-randomized open pilot study of hypothermia in severe MCA infarction, cooling to 32 ± 1°C appeared to be safe. No significant improvement of functional outcome in the hypothermia treated group was seen, but sample sizes were small and outcome trends favored hypothermia.139 Randomized trials have not been performed. Surgical decompression Because of the limitations of medical therapies, there have been proposals for decompressive surgery in patients with hemispheric infarction and neurological 27 Chapter 2 deterioration caused by space-occupying edema. This therapy is presumed to revert brain tissue shifts, to normalize intracranial pressure, and to preserve cerebral blood flow, thus preventing secondary brain damage. The technique of decompressive surgery is relatively simple and consists of a large hemicraniectomy and a duraplasty.140 Animal studies have shown that this intervention reduces mortality and improves functional and histological outcome.141-144 Case reports and small retrospective or non-controlled studies suggested that hemicraniectomy lowers mortality without increasing the number of severely disabled survivors.145-153 This finding has been confirmed in two prospective series, in which patients younger than 70 years with clinical and CT evidence of acute severe MCA infarction were included. In the first series decompression was performed in 32 patients after clinical deterioration consisting of a progressive decrease in consciousness. Mortality was reduced from 79% in controls to 34% in surgicallytreated patients, and poor functional outcome from 95% to 50%. The mean interval between the onset of symptoms and surgery was 39 hours.154 In a subsequent study, in which hemicraniectomy was performed in 31 patients within 24 hours after the onset of symptoms, mortality was reduced to 16%, without an increase in the number of severely disabled survivors.155 Complications of the operative procedure were generally not serious and had no effect on patient outcome. Parenchymal bleeding occurred more often with smaller bone resections.156 In another small prospective series of patients (n=19), hemicraniectomy reduced mortality and improved short term clinical outcome (Glasgow Outcome Scale at three months) as compared to a non-randomized control group (n=15).157 Other reports suggest that decompressive surgery is less effective in elderly patients158 and that substantial recovery extends into the second half year and thereafter.159 In recent non-controlled patient series, a better outcome was observed in younger patients and associated with intervention as soon as possible after160,161 or before162 the onset of clinical signs of herniation. The results of two relatively large prospective studies154,155 suggest a substantial benefit of decompressive surgery as compared with medical therapy alone. However, groups were not constituted by random selection. Control groups consisted of patients with a significantly higher age, more co-morbidity and more frequent lesions in the dominant hemisphere than those in the surgical groups. In addition, information on functional outcome was insufficient.154,155 Randomized trials have not been performed.163 Discussion None of the therapeutic strategies proposed to control cerebral edema formation and to reduce tissue shifts and raised ICP after space-occupying hemispheric infarction is supported by adequate evidence of efficacy from experimental studies or clinical trials. If any, experimental support for these strategies is derived from studies with 28 Treatment of space-occupying MCA infarction animal models of moderately severe focal cerebral ischemia, not adequately reflecting the syndrome of space-occupying infarction in man. Several studies suggest that the beneficial effects of treatment with mannitol,24,164-166 hypothermia,167,168 or barbiturates73 demonstrated in transient or moderate focal cerebral ischemia may be absent in cases of permanent or more severe infarcts. In rats, edema formation after cerebral infarction occurs earlier than in man. On histopathological examination of rat brains at 6, 24, and 72 hours, and at seven days after the onset of transient focal cerebral ischemia brain water content peaked at 24 hours.169 In patients, clinical deterioration from serious edema formation usually occurs between the second and the fifth day after stroke onset.4,170-172 This difference in the timing of edema formation may have consequences for the extrapolation of the results of rat studies into the clinic. Most treatments are based on the perception that a raised intracranial pressure is the dominant cause of neurologic deterioration. However, displacement of brain tissue rather than increased ICP is probably the most likely cause of the initial decrease in consciousness and further neurological deterioration.170 One study, in which ICP was monitored in 48 patients with clinical signs of increased ICP caused by large hemispheric infarction, showed that ICP measurements were not helpful in guiding long-term treatment.173 Reducing ICP with osmotic agents or hyperventilation might even be harmful, because the reduction in volume of the contralateral hemisphere, where the blood brain barrier and cerebral autoregulation are still intact, might be more pronounced than that of the infarcted hemisphere, thereby increasing brain tissue shifts.170 Moreover, osmotic agents like mannitol and glycerol may accumulate in the affected tissue, thereby reversing the osmotic gradient between tissue and plasma, leading to an exacerbation of edema.12 Therefore, the outcomes of osmotic treatment may be largely dependent on the timing and the duration of treatment.174 According to the guidelines of the American Heart Association, patients with space-occupying cerebral infarction whose condition is deteriorating secondary to edema formation should be treated with osmotic agents and hyperventilation.6 Other experts recommend decompressive surgery and hypothermia for the treatment of these patients. They suggest that early intervention generates better results in terms of mortality and functional recovery of survivors, and that treatment should probably be started even before clinical deterioration in patients with massive infarction.175 There is no unequivocal evidence to support either opinion. It remains unclear which patients should be candidates for intensive anti-edema treatment. Several parameters have been studied as possible predictors of the development of fatal brain edema. An increased risk was found to be associated with clinical conditions such as a high NIH Stroke Scale score on admission, early nausea and vomiting, hypertension, and heart failure, but the predictive value of the different conditions was weak.176,177 Radiological predictors of fatal brain edema include CT 29 Chapter 2 hypodensity of 50% or more of the MCA territory176,177 and lesion volume on diffusion weighted MRI (DWI) exceeding 145cm3.178 Although DWI has high sensitivity and specificity rates when performed within 14 hours after stroke onset, the sensitivity of this parameter may be considerably lower in earlier phases of the infarct.179 In these very early phases other parameters, such as a complete MCA territory perfusion deficit or MCA occlusion on MR angiography may be more sensitive predictors of the development of malignant infarction.180,181 An unambiguous decision based on one or on a combination of these parameters can not yet be made. It remains unclear whether patients with severe aphasia should be treated as aggressively as patients without. Despite severe language disturbances, quality of life in these patients is not necessarily worse than in other patients.182 Therefore patients with aphasia should not be excluded from trials testing treatment strategies in spaceoccupying infarction. In animal studies hypothermia reduced infarct size very consistently. Furthermore, non-randomized studies in patients with severe space-occupying edema after MCA infarction suggested that moderate hypothermia (32°C -34°C) can help to control critically elevated ICP values and to improve clinical outcome.136-138 Thus, hypothermia deserves further research as a measure to prevent and treat massive edema formation. Also surgical decompression is a promising treatment option, given the suggested large reductions in mortality.154,155 Before implementation of the different proposed strategies as standard treatment modalities, data from randomized controlled clinical trials are needed. Multi-center randomized trials of decompressive surgery for space-occupying hemispheric infarction are on their way.183 References 1. 2. 3. 4. 5. 6. 7. 30 Silver FL, Norris JW, Lewis AJ, Hachinski VC. Early mortality following stroke: a prospective review. Stroke 1984;15:492-496. Heinsius T, Bogousslavsky J, Van Melle G. Large infarcts in the middle cerebral artery territory. Etiology and outcome patterns. Neurology 1998;50:341-350. Biller J, Adams-HP J, Bruno A, Love BB, Marsh EE. Mortality in acute cerebral infarction in young adults--a ten-year experience. Angiology 1991;42:224-230. Hacke W, Schwab S, Horn M, Spranger M, De Georgia M, von Kummer R. ‘Malignant’ middle cerebral artery territory infarction: clinical course and prognostic signs. Arch Neurol 1996;53:309-315. Wijdicks EF, Diringer MN. Middle cerebral artery territory infarction and early brain swelling: progression and effect of age on outcome. Mayo Clin Proc 1998;73:829-836. Adams HP, Jr., Adams RJ, Brott T. Guidelines for the early management of patients with ischemic stroke: A scientific statement from the Stroke Council of the American Stroke Association. Stroke 2003;34:10561083. Wijdicks EF. Management of massive hemispheric cerebral infarct: is there a ray of hope? Mayo Clin Proc 2000;75:945-952. Treatment of space-occupying MCA infarction 8. 9. 10. 11. 12. 13. 14. 15. 16. 17. 18. 19. 20. 21. 22. 23. 24. 25. 26. 27. 28. Schwarz S, Schwab S, Bertram M, Aschoff A, Hacke W. Effects of hypertonic saline hydroxyethyl starch solution and mannitol in patients with increased intracranial pressure after stroke. Stroke 1998;29:15501555. van der Worp HB, Kappelle LJ. Complications of acute ischaemic stroke. Cerebrovasc Dis 1998;8:124132. Schwab S, Spranger M, Schwarz S, Hacke W. Barbiturate coma in severe hemispheric stroke: useful or obsolete? Neurology 1997;48:1608-1613. Muizelaar JP, Marmarou A, Ward JD. Adverse effects of prolonged hyperventilation in patients with severe head injury: a randomized clinical trial. J Neurosurg 1991;75:731-739. Kaufmann AM, Cardoso ER. Aggravation of vasogenic cerebral edema by multiple-dose mannitol. J Neurosurg 1992;77:584-589. Schell RM, Applegate RL, Cole DJ. Salt, starch, and water on the brain. J Neurosurg Anesthesiol 1996;8:178-182. Burke AM, Quest DO, Chien S, Cerri C. The effects of mannitol on blood viscosity. J Neurosurg 1981;55:550-553. Jafar JJ, Johns LM, Mullan SF. The effect of mannitol on cerebral blood flow. J Neurosurg 1986;64:754759. Andrews RJ, Bringas JR, Muto RP. Effects of mannitol on cerebral blood flow, blood pressure, blood viscosity, hematocrit, sodium, and potassium. Surg Neurol 1993;39:218-222. Shirane R, Weinstein PR. Effect of mannitol on local cerebral blood flow after temporary complete cerebral ischemia in rats. J Neurosurg 1992;76:486-492. Muizelaar JP, Wei EP, Kontos HA, Becker DP. Mannitol causes compensatory cerebral vasoconstriction and vasodilation in response to blood viscosity changes. J Neurosurg 1983;59:822-828. Rosner MJ, Coley I. Cerebral perfusion pressure: a hemodynamic mechanism of mannitol and the postmannitol hemogram. Neurosurgery 1987;21:147-156. Suzuki J, Imaizumi S, Kayama T, Yoshimoto T. Chemiluminescence in hypoxic brain--the second report: cerebral protective effect of mannitol, vitamin E and glucocorticoid. Stroke 1985;16:695-700. Little JR. Modification of acute focal ischemia by treatment with mannitol and high-dose dexamethasone. J Neurosurg 1978;49:517-524. Little JR. Modification of acute focal ischemia by treatment with mannitol. Stroke 1978;9:4-9. Little JR. Treatment of acute focal cerebral ischemia with intermittent, low dose mannitol. Neurosurgery 1979;5:687-691. Little JR. Morphological changes in acute focal ischemia: response to osmotherapy. Adv Neurol 1980;28:443-457. Suzuki J, Tanaka S, Yoshimoto T, Seki H. Recirculation in the acute period of cerebral infarction: experimental research on brain swelling and its suppression by using mannitol or glycerol. Acta Neurochir Wien 1980;54:219-231. Ehteshami S, Aspey BS, Hurst CM, McCoy AL, Harrison MJ. The combined effects of hypertension, hemodilution, and osmotherapy on the metabolic sequelae of acute experimental cerebral ischemia. Metab Brain Dis 1988;3:235-244. Karibe H, Zarow GJ, Weinstein PR. Use of mild intraischemic hypothermia versus mannitol to reduce infarct size after temporary middle cerebral artery occlusion in rats. J Neurosurg 1995;83:93-98. Kobayashi H, Ide H, Kabuto M, Handa Y, Kubota T, Ishii Y. Effect of mannitol on focal cerebral ischemia evaluated by somatosensory-evoked potentials and magnetic resonance imaging. Surg Neurol 31 Chapter 2 29. 30. 31. 32. 33. 34. 35. 36. 37. 38. 39. 40. 41. 42. 43. 44. 45. 46. 47. 48. 32 1995;44:55-61. Kobayashi H, Ide H, Kodera T. Effect of mannitol on focal cerebral ischemia evaluated by magnetic resonance imaging. Acta Neurochir Suppl Wien 1994;60:228-230. Paczynski RP, He YY, Diringer MN, Hsu CY. Multiple-dose mannitol reduces brain water content in a rat model of cortical infarction. Stroke 1997;28:1437-1443. Paczynski RP, Venkatesan R, Diringer MN, He YY, Hsu CY, Lin W. Effects of fluid management on edema volume and midline shift in a rat model of ischemic stroke. Stroke 2000;31:1702-1708. Ogilvy CS, Chu D, Kaplan S. Mild hypothermia, hypertension, and mannitol are protective against infarction during experimental intracranial temporary vessel occlusion. Neurosurgery 1996;38:12021209. Oktem IS, Menku A, Akdemir H, Kontas O, Kurtsoy A, Koc RK. Therapeutic effect of tirilazad mesylate (U-74006F), mannitol, and their combination on experimental ischemia. Res Exp Med Berl 2000;199:231242. Koc RK, Akdemir H, Kandemir O, Pasaoglu H, Oktem IS, Pasaoglu A. The therapeutic value of naloxone and mannitol in experimental focal cerebral ischemia. Neurological outcome, histopathological findings, and tissue concentrations of Na+, K+ and water. Res Exp Med Berl 1994;194:277-285. Bhardwaj A, Harukuni I, Murphy SJ. Hypertonic saline worsens infarct volume after transient focal ischemia in rats. Stroke 2000;31:1694-1701. Gueniau C, Oberlander C. The kappa opioid agonist niravoline decreases brain edema in the mouse middle cerebral artery occlusion model of stroke. J Pharmacol Exp Ther 1997;282:1-6. Kotwica Z, Persson L. Effect of mannitol on intracranial pressure in focal cerebral ischemia. An experimental study in a rat. Mater Med Pol 1991;23:280-284. Nau R, Prins FJ, Kolenda H, Prange HW. Temporary reversal of serum to cerebrospinal fluid glycerol concentration gradient after intravenous infusion of glycerol. Eur J Clin Pharmacol 1992;42:181-185. Nau R, Desel H, Lassek C. Slow elimination of mannitol from human cerebrospinal fluid. Eur J Clin Pharmacol 1997;53:271-274. Manno EM, Adams RE, Derdeyn CP, Powers WJ, Diringer MN. The effects of mannitol on cerebral edema after large hemispheric cerebral infarct. Neurology 1999;52:583-587. Videen TO, Zazulia AR, Manno EM, et al. Mannitol bolus preferentially shrinks non-infarcted brain in patients with ischemic stroke. Neurology 2001;11:2120-2122. Santambrogio S, Martinotti R, Sardella F, Porro F, Randazzo A. Is there a real treatment for stroke? Clinical and statistical comparison of different treatments in 300 patients. Stroke 1978;9:130-132. Candelise L, Colombo A, Spinnler H. Therapy against brain swelling in stroke patients. A retrospective clinical study on 227 patients. Stroke 1975;6:353-356. Bereczki D, Liu M, do PG, Fekete I. Mannitol for acute stroke. Cochrane Database Syst Rev 2002 Jan;CD001153. Antonini FM, Bertini G, Fumagalli C. Effects of intravenous infusion of glycerol on regional cerebral blood flow in cerebral infarction. Gerontology 1977;23:376-380. Ott EO, Mathew NT, Meyer JS. Redistribution of regional cerebral blood flow after glycerol infusion in acute cerebral infarction. Neurology 1974;24:1117-1126. Meyer JS, Itoh Y, Okamoto S. Circulatory and metabolic effects of glycerol infusion in patients with recent cerebral infarction. Circulation 1975;51:701-712. Dodson RF, Tagashira Y, Wai-Fong CL. The effects of glycerol on cerebral ultrastructure following experimentally induced cerebral ischemia. J Neurol Sci 1975;26:235-243. Treatment of space-occupying MCA infarction 49. 50. 51. 52. 53. 54. 55. 56. 57. 58. 59. 60. 61. 62. 63. 64. 65. 66. 67. Popovic P, Popovic V, Schaffer R, Sutton CH. Treatment of experimental cerebral infarction in rats with levodopa or with glycerol. J Neurosurg 1978;48:962-969. Dietis A, Ehteshami S, Harrison MJ, Perinpanayagam NI. The effect of isovolaemic haemodilution and intravenous glycerol on the sequelae of middle cerebral artery occlusion in the rat. J Neurol Neurosurg Psychiatry 1986;49:428-430. Meyer JS, Teraura T, Marx P, Hashi K, Sakamoto K. Brain swelling due to experimental cerebral infarction. Changes in vasomotor capacitance and effects of intravenous glycerol. Brain 1972;95:833852. Berger C, Sakowitz OW, Kiening KL, Schwab S. Neurochemical monitoring of glycerol therapy in patients with ischemic brain edema. Stroke 2005;36:e4-e6. Sakamaki M, Igarashi H, Nishiyama Y. Effect of glycerol on ischemic cerebral edema assessed by magnetic resonance imaging. J Neurol Sci 2003;15:69-74. Righetti E, Celani MG, Cantisani T, Sterzi R, Boysen G, Ricci S. Glycerol for acute stroke. Cochrane Database Syst Rev 2004;CD000096. Zornow MH. Hypertonic saline as a safe and efficacious treatment of intracranial hypertension. J Neurosurg Anesthesiol 1996;8:175-177. Toung TJ, Chang Y, Lin J, Bhardwaj A. Increases in lung and brain water following experimental stroke: effect of mannitol and hypertonic saline. Crit Care Med 2005;33:203-208. Toung TJ, Hurn PD, Traystman RJ, Bhardwaj A. Global brain water increases after experimental focal cerebral ischemia: effect of hypertonic saline. Crit Care Med 2002;30:644-649. Schwarz S, Georgiadis D, Aschoff A, Schwab S. Effects of hypertonic (10%) saline in patients with raised intracranial pressure after stroke. Stroke 2002;33:136-140. Hacke W, Schwab S, De Georgia M. Intensive Care of Acute Ischemic Stroke. Cerebrovasc Dis 1994;4:385-392. Woster PS, LeBlanc KL. Management of elevated intracranial pressure. Clin Pharm 1990;9:762-772. Harbaugh RD, James HE, Marshall LF, Shapiro HM, Laurin R. Acute therapeutic modalities for experimental vasogenic edema. Neurosurgery 1979;5:656-665. Millson C, James HE, Shapiro HM, Laurin R. Intracranial hypertension and brain oedema in albino rabbits. Part 2: Effects of acute therapy with diuretics. Acta Neurochir (Wien) 1981;56:167-181. Nordstrom CH, Messeter K, Sundbarg G, Schalen W, Werner M, Ryding E. Cerebral blood flow, vasoreactivity, and oxygen consumption during barbiturate therapy in severe traumatic brain lesions. J Neurosurg 1988;68:424-431. Kassell NF, Hitchon PW, Gerk MK, Sokoll MD, Hill TR. Alterations in cerebral blood flow, oxygen metabolism, and electrical activity produced by high dose sodium thiopental. Neurosurgery 1980;7:598603. Ochiai C, Asano T, Takakura K, Fukuda T, Horizoe H, Morimoto Y. Mechanisms of cerebral protection by pentobarbital and nizofenone correlated with the course of local cerebral blood flow changes. Stroke 1982;13:788-796. Smith DS, Rehncrona S, Siesjo BK. Inhibitory effects of different barbiturates on lipid peroxidation in brain tissue in vitro: comparison with the effects of promethazine and chlorpromazine. Anesthesiology 1980;53:186-194. Smith DS, Rehncrona S, Siesjo BK. Barbiturates as protective agents in brain ischemia and as free radical scavengers in vitro. Acta Physiol Scand Suppl 1980;492:129-134. 33 Chapter 2 68. 69. 70. 71. 72. 73. 74. 75. 76. 77. 78. 79. 80. 81. 82. 83. 84. 85. 86. 87. 88. 89. 90. 34 Hoff J, Smith A, Nielsen S, Larson P. Effects of barbiturate and halothane anesthesia on focal cerebral infarction in the dog. Surg Forum 1973;24:449-451. Smith AL, Hoff JT, Nielsen SL, Larson CP. Barbiturate protection in acute focal cerebral ischemia. Stroke 1974;5:1-7. Hoff JT, Smith AL, Hankinson HL, Nielsen SL. Barbiturate protection from cerebral infarction in primates. Stroke 1975;6:28-33. Moseley JI, Laurent JP, Molinari GF. Barbiturate attenuation of the clinical course and pathologic lesions in a primate stroke model. Neurology 1975;25:870-874. Corkill G, Sivalingam S, Reitan JA, Gilroy BA, Helphrey MG. Dose dependency of the post-insult protective effect of pentobarbital in the canine experimental stroke model. Stroke 1978;9:10-12. Selman WR, Spetzler RF, Roessmann UR, Rosenblatt JI, Crumrine RC. Barbiturate-induced coma therapy for focal cerebral ischemia. Effect after temporary and permanent MCA occlusion. J Neurosurg 1981;55:220-226. Hoff JT, Nishimura M, Newfield P. Pentobarbital protection from cerebral infarction without suppression of edema. Stroke 1982;13:623-628. Pappas TN, Mironovich RO. Barbiturate-induced coma to protect against cerebral ischemia and increased intracranial pressure. Am J Hosp Pharm 1981;38:494-498. Shapiro HM. Barbiturates in brain ischaemia. Br J Anaesth 1985;57:82-95. Selman WR, Spetzler RF, Roski RA, Roessmann U, Crumrine R, Macko R. Barbiturate coma in focal cerebral ischemia. Relationship of protection to timing of therapy. J Neurosurg 1982;56:685-690. Woodhurst WB, Robertson WD, Thompson GB. Carotid injury due to intraoral trauma: case report and review of the literature. Neurosurgery 1980;6:559-563. Belopavlovic M, Buchthal A. Barbiturate therapy in the management of cerebral ischaemia. Anaesthesia 1980;35:271-278. Caseby NG. Postpartum stroke successfully treated with high-dose pentobarbitone therapy: a case report. Can Anaesth Soc J 1983;30:77-83. Woodcock J, Ropper AH, Kennedy SK. High dose barbiturates in non-traumatic brain swelling: ICP reduction and effect on outcome. Stroke 1982;13:785-787. Rockoff MA, Marshall LF, Shapiro HM. High-dose barbiturate therapy in humans: a clinical review of 60 patients. Ann Neurol 1979;6:194-199. Hall ED. The neuroprotective pharmacology of methylprednisolone. J Neurosurg 1992;76:13-22. French L. The use of steroids in the treatment of cerebral oedema. Bull N Y Acad Med 1966;42:301311. Donley RF, Sundt-TM J. The effect of dexamethasone on the edema of focal cerebral ischemia. Stroke 1973;4:148-155. Lee MC, Mastri AR, Waltz AG, Loewenson RB. Ineffectiveness of dexamethasone for treatment of experimental cerebral infarction. Stroke 1974;5:216-218. de-la-Torre JC, Surgeon JW. Dexamethasone and DMSO in experimental transorbital cerebral infarction. Stroke 1976;7:577-583. Slivka AP, Murphy EJ. High-dose methylprednisolone treatment in experimental focal cerebral ischemia. Exp Neurol 2001;167:166-172. Klatzo I. Neuropathological aspects of brain edema. J Neuropathol Exp Neurol 1967;26:1-14. Patten BM, Mendell J, Bruun B, Curtin W, Carter S. Double-blind study of the effects of dexamethasone on acute stroke. Neurology 1972;22:377-383. Treatment of space-occupying MCA infarction 91. 92. 93. 94. 95. 96. 97. 98. 99. 100. 101. 102. 103. 104. 105. 106. 107. 108. 109. 110. 111. 112. Bauer RB, Tellez H. Dexamethasone as treatment in cerebrovascular disease. 2. A controlled study in acute cerebral infarction. Stroke 1973;4:547-555. Kaste M, Fogelholm R, Waltimo O. Combined dexamethasone and low-molecular-weight dextran in acute brain infarction: double-blind study. Br Med J 1976 11;2:1409-1410. Norris JW. Steroid therapy in acute cerebral infarction. Arch Neurol 1976;33:69-71. Mulley G, Wilcox RG, Mitchell JR. Dexamethasone in acute stroke. Br Med J 1978;7:994-996. Norris JW, Hachinski VC. High dose steroid treatment in cerebral infarction. Br Med J Clin Res Ed 1986;4:21-23. Ogun SA, Odusote KA. Effectiveness of high dose dexamethasone in the treatment of acute stroke. West Afr J Med 2001;20:1-6. Hetzel BS, Lander H, Robson H. Immediate treatment of apoplexy. BMJ 1957;1:1122. Zuliani G, Cherubini A, Atti AR. Prescription of anti-oedema agents and short-term mortality in older patients with acute ischaemic stroke. Drugs Aging 2004;21:273-278. Qizilbash N, Lewington SL, Lopez-Arrieta JM. Corticosteroids for acute ischaemic stroke. Cochrane Database Syst Rev 2002;CD000064. Marshall LF, Smith RW, Shapiro HM. The outcome with aggressive treatment in severe head injuries. Part I: the significance of intracranial pressure monitoring. J Neurosurg 1979;50:20-25. Muizelaar JP, van der Poel HG, Li ZC, Kontos HA, Levasseur JE. Pial arteriolar vessel diameter and CO2 reactivity during prolonged hyperventilation in the rabbit. J Neurosurg 1988;69:923-927. Nariai T, Senda M, Ishii K. Posthyperventilatory steal response in chronic cerebral hemodynamic stress: a positron emission tomography study. Stroke 1998;29:1281-1292. Yundt KD, Diringer MN. The use of hyperventilation and its impact on cerebral ischemia in the treatment of traumatic brain injury. Crit Care Clin 1997;13:163-184. Stringer WA, Hasso AN, Thompson JR, Hinshaw DB, Jordan KG. Hyperventilation-induced cerebral ischemia in patients with acute brain lesions: demonstration by xenon-enhanced CT. AJNR 1993;14:475484. Soloway M, Moriarty G, Fraser JG, White RJ. Effect of delayed hyperventilation on experimental cerebral infarction. Neurology 1971;21:479-485. Michenfelder JD, Milde JH. Failure of prolonged hypocapnia, hypothermia, or hypertension to favorably alter acute stroke in primates. Stroke 1977;8:87-91. Christensen MS, Paulson OB, Olesen J. Cerebral apoplexy (stroke) treated with or without prolonged artificial hyperventilation. 1. Cerebral circulation, clinical course, and cause of death. Stroke 1973;4:568-631. Simard D, Paulson OB. Artifical hyperventilation in stroke. Trans Am Neurol Assoc 1973;98:309310. Chopp M, Knight R, Tidwell CD, Helpern JA, Brown E, Welch KM. The metabolic effects of mild hypothermia on global cerebral ischemia and recirculation in the cat: comparison to normothermia and hyperthermia. J Cereb Blood Flow Metab 1989;9:141-148. Berntman L, Welsh FA, Harp JR. Cerebral protective effect of low-grade hypothermia. Anesthesiology 1981;55:495-498. Dietrich WD, Busto R, Halley M, Valdes I. The importance of brain temperature in alterations of the blood-brain barrier following cerebral ischemia. J Neuropathol Exp Neurol 1990;49:486-497. Toyoda T, Suzuki S, Kassell NF, Lee KS. Intraischemic hypothermia attenuates neutrophil infiltration in the rat neocortex after focal ischemia-reperfusion injury. Neurosurgery 1996;39:1200-1205. 35 Chapter 2 113. Busto R, Globus MY, Dietrich WD, Martinez E, Valdes I, Ginsberg MD. Effect of mild hypothermia on ischemia-induced release of neurotransmitters and free fatty acids in rat brain. Stroke 1989;20:904910. 114. Huang FP, Zhou LF, Yang GY. Effects of mild hypothermia on the release of regional glutamate and glycine during extended transient focal cerebral ischemia in rats. Neurochem Res 1998;23:991-996. 115. Winfree CJ, Baker CJ, Connolly-ES J, Fiore AJ, Solomon RA. Mild hypothermia reduces penumbral glutamate levels in the rat permanent focal cerebral ischemia model. Neurosurgery 1996;38:12161222. 116. Goto Y, Kassell NF, Hiramatsu K, Soleau SW, Lee KS. Effects of intraischemic hypothermia on cerebral damage in a model of reversible focal ischemia. Neurosurgery 1993;32:980-984. 117. Jiang Q, Chopp M, Zhang ZG. The effect of hypothermia on transient focal ischemia in rat brain evaluated by diffusion- and perfusion-weighted NMR imaging. J Cereb Blood Flow Metab 1994;14:732741. 118. Karibe H, Chen J, Zarow GJ, Graham SH, Weinstein PR. Delayed induction of mild hypothermia to reduce infarct volume after temporary middle cerebral artery occlusion in rats. J Neurosurg 1994;80:112-119. 119. Moyer DJ, Welsh FA, Zager EL. Spontaneous cerebral hypothermia diminishes focal infarction in rat brain. Stroke 1992;23:1812-1816. 120. Kawai N, Okauchi M, Morisaki K, Nagao S. Effects of delayed intraischemic and postischemic hypothermia on a focal model of transient cerebral ischemia in rats. Stroke 2000;31:1982-1989. 121. Yanamoto H, Hong SC, Soleau S, Kassell NF, Lee KS. Mild postischemic hypothermia limits cerebral injury following transient focal ischemia in rat neocortex. Brain Res 1996;29:207-211. 122. Yanamoto H, Nagata I, Niitsu Y, et al. Prolonged mild hypothermia therapy protects the brain against permanent focal ischemia. Stroke 2001;32:232-239. 123. Markarian GZ, Lee JH, Stein DJ, Hong SC. Mild hypothermia: therapeutic window after experimental cerebral ischemia. Neurosurgery 1996;38:542-550. 124. Zhang ZG, Chopp M, Chen H. Duration dependent post-ischemic hypothermia alleviates cortical damage after transient middle cerebral artery occlusion in the rat. J Neurol Sci 1993;117:240-244. 125. Zhang RL, Chopp M, Chen H, Garcia JH, Zhang ZG. Postischemic (1 hour) hypothermia significantly reduces ischemic cell damage in rats subjected to 2 hours of middle cerebral artery occlusion. Stroke 1993;24:1235-1240. 126. Huh PW, Belayev L, Zhao W, Koch S, Busto R, Ginsberg MD. Comparative neuroprotective efficacy of prolonged moderate intraischemic and postischemic hypothermia in focal cerebral ischemia. J Neurosurg 2000;92:91-99. 127. Kader A, Brisman MH, Maraire N, Huh JT, Solomon RA. The effect of mild hypothermia on permanent focal ischemia in the rat. Neurosurgery 1992;31:1056-1060. 128. Baker CJ, Onesti ST, Solomon RA. Reduction by delayed hypothermia of cerebral infarction following middle cerebral artery occlusion in the rat: a time-course study. J Neurosurg 1992;77:438-444. 129. Maier CM, Sun GH, Kunis D, Yenari MA, Steinberg GK. Delayed induction and long-term effects of mild hypothermia in a focal model of transient cerebral ischemia: neurological outcome and infarct size. J Neurosurg 2001;94:90-96. 130. Kamiya T, Nito C, Ueda M. Mild hypothermia enhances the neuroprotective effects of a selective thrombin inhibitor following transient focal ischemia in rats. Acta Neurochir Suppl 2003;86:195198. 36 Treatment of space-occupying MCA infarction 131. Nito C, Kamiya T, Ueda M, Arii T, Katayama Y. Mild hypothermia enhances the neuroprotective effects of FK506 and expands its therapeutic window following transient focal ischemia in rats. Brain Res 2004;22:179-185. 132. Nito C, Kamiya T, Amemiya S, Katoh K, Katayama Y. The neuroprotective effect of a free radical scavenger and mild hypothermia following transient focal ischemia in rats. Acta Neurochir Suppl 2003;86:199-203. 133. Karibe H, Zarow GJ, Graham SH, Weinstein PR. Mild intraischemic hypothermia reduces postischemic hyperperfusion, delayed postischemic hypoperfusion, blood-brain barrier disruption, brain edema, and neuronal damage volume after temporary focal cerebral ischemia in rats. J Cereb Blood Flow Metab 1994;14:620-627. 134. Kollmar R, Schabitz WR, Heiland S. Neuroprotective effect of delayed moderate hypothermia after focal cerebral ischemia: an MRI study. Stroke 2002;33:1899-1904. 135. Dempsey RJ, Combs DJ, Maley ME, Cowen DE, Roy MW, Donaldson DL. Moderate hypothermia reduces postischemic edema development and leukotriene production. Neurosurgery 1987;21:177181. 136. Schwab S, Schwarz S, Spranger M, Keller E, Bertram M, Hacke W. Moderate hypothermia in the treatment of patients with severe middle cerebral artery infarction. Stroke 1998;29:2461-2466. 137. Schwab S, Georgiadis D, Berrouschot J, Schellinger PD, Graffagnino C, Mayer SA. Feasibility and safety of moderate hypothermia after massive hemispheric infarction. Stroke 2001;32:2033-2035. 138. Steiner T, Friede T, Aschoff A, Schellinger PD, Schwab S, Hacke W. Effect and feasibility of controlled rewarming after moderate hypothermia in stroke patients with malignant infarction of the middle cerebral artery. Stroke 2001;32:2833-2835. 139. Krieger DW, De Georgia M, Abou-Chebl A, Andrefsky JC, Sila CA, Katzan IL, Mayberg MR, Furlan AJ. Cooling for acute ischemic brain damage (COOL AID). An open pilot study of induced hypothermia in acute ischemic stroke. Stroke 2001;32:1847-1854. 140. Schwab S, Rieke K, Aschoff A, Albert F, von Kummer R, Hacke W. Hemicraniotomy in space-occupying hemispheric infarction: useful early intervention or desparate activism? Cerebrovasc Dis 1996;6:325329. 141. Doerfler A, Forsting M, Reith W. Decompressive craniectomy in a rat model of “malignant” cerebral hemispheric stroke: experimental support for an aggressive therapeutic approach. J Neurosurg 1996;85:853-859. 142. Forsting M, Reith W, Schabitz WR. Decompressive craniectomy for cerebral infarction. An experimental study in rats. Stroke 1995;26:259-264. 143. Engelhorn T, Doerfler A, Kastrup A. Decompressive craniectomy, reperfusion, or a combination for early treatment of acute “malignant” cerebral hemispheric stroke in rats? Potential mechanisms studied by MRI. Stroke 1999;30:1456-1463. 144. Engelhorn T, von Kummer R, Reith W, Forsting M, Doerfler A. What is effective in malignant middle cerebral artery infarction: reperfusion, craniectomy, or both? An experimental study in rats. Stroke 2002;33:617-622. 145. Rengachary SS, Batnitzky S, Morantz RA, Arjunan K, Jeffries B. Hemicraniectomy for acute massive cerebral infarction. Neurosurgery 1981;8:321-328. 146. Robertson SC, Lennarson P, Hasan DM, Traynelis VC. Clinical course and surgical management of massive cerebral infarction. Neurosurgery 2004;55:55-61. 147. Reddy AK, Saradhi V, Panigrahi M, Rao TN, Tripathi P, Meena AK. Decompressive craniectomy for 37 Chapter 2 stroke : indications and results. Neurol India 2002;50 Suppl:S66-S69. 148. Walz B, Zimmermann C, Bottger S, Haberl RL. Prognosis of patients after hemicraniectomy in malignant middle cerebral artery infarction. J Neurol 2002;249:1183-1190. 149. Carter BS, Ogilvy CS, Candia GJ, Rosas HD, Buonanno F. One-year outcome after decompressive surgery for massive nondominant hemispheric infarction. Neurosurgery 1997;40:1168-1175. 150. Delashaw JB, Broaddus WC, Kassell NF. Treatment of right hemispheric cerebral infarction by hemicraniectomy. Stroke 1990;21:874-881. 151. Kondziolka D, Fazl M. Functional recovery after decompressive craniectomy for cerebral infarction. Neurosurgery 1988;23:143-147. 152. Kalia KK, Yonas H. An aggressive approach to massive middle cerebral artery infarction. Arch Neurol 1993;50:1293-1297. 153. Koh MS, Goh KY, Tung MY, Chan C. Is decompressive craniectomy for acute cerebral infarction of any benefit? Surg Neurol 2000;53:225-230. 154. Rieke K, Schwab S, Krieger D. Decompressive surgery in space-occupying hemispheric infarction: results of an open, prospective trial. Crit Care Med 1995;23:1576-1587. 155. Schwab S, Steiner T, Aschoff A. Early hemicraniectomy in patients with complete middle cerebral artery infarction. Stroke 1998;29:1888-1893. 156. Wagner S, Schnippering H, Aschoff A, Koziol JA, Schwab S, Steiner T. Suboptimum hemicraniectomy as a cause of additional cerebral lesions in patients with malignant infarction of the middle cerebral artery. J Neurosurg 2001;94:693-696. 157. Mori K, Aoki A, Yamamoto T, Horinaka N, Maeda M. Aggressive decompressive surgery in patients with massive hemispheric embolic cerebral infarction associated with severe brain swelling. Acta Neurochir (Wien) 2002;143:483-491. 158. Holtkamp M, Buchheim K, Unterberg A. Hemicraniectomy in elderly patients with space occupying media infarction: improved survival but poor functional outcome. J Neurol Neurosurg Psychiatry 2001;70:226-228. 159. Manai R, Srour A, Crozier S, Vandamme X, Samson Y, Cornu P, Rancurel G. Long-term functional outcome of hemicraniectomy in middle cerebral artery malignant infarcts [abstract]. J Neurol 2001; 248 (suppl 2):121-122. 160. Kilincer C, Asil T, Utku U. Factors affecting the outcome of decompressive craniectomy for large hemispheric infarctions: a prospective cohort study. Acta Neurochir (Wien ) 2005;147:587-594. 161. Kastrau F, Wolter M, Huber W, Block F. Recovery from aphasia after hemicraniectomy for infarction of the speech-dominant hemisphere. Stroke 2005;36:825-829. 162. Mori K, Nakao Y, Yamamoto T, Maeda M. Early external decompressive craniectomy with duroplasty improves functional recovery in patients with massive hemispheric embolic infarction: timing and indication of decompressive surgery for malignant cerebral infarction. Surg Neurol 2004;62:420-429. 163. Morley NC, Berge E, Cruz-Flores S, Whittle IR. Surgical decompression for cerebral oedema in acute ischaemic stroke. Cochrane Database Syst Rev 2002;CD003435. 164. Meyer FB, Anderson RE, Sundt-TM J, Yaksh TL. Treatment of experimental focal cerebral ischemia with mannitol. Assessment by intracellular brain pH, cortical blood flow, and electroencephalography. J Neurosurg 1987;66:109-115. 165. Seki H, Yoshimoto T, Ogawa A, Suzuki J. Effect of mannitol on rCBF in canine thalamic ischemia--an experimental study. Stroke 1983;14:46-50. 166. Suzuki J, Tanaka S, Yoshimoto T. Recirculation in the acute period of cerebral infarction: brain swelling 38 Treatment of space-occupying MCA infarction and its suppression using mannitol. Surg Neurol 1980;14:467-472. 167. Ridenour TR, Warner DS, Todd MM, McAllister AC. Mild hypothermia reduces infarct size resulting from temporary but not permanent focal ischemia in rats. Stroke 1992;23:733-738. 168. Morikawa E, Ginsberg MD, Dietrich WD, et al. The significance of brain temperature in focal cerebral ischemia: histopathological consequences of middle cerebral artery occlusion in the rat. J Cereb Blood Flow Metab 1992;12:380-389. 169. Lin TN, He YY, Wu G, Khan M, Hsu CY. Effect of brain edema on infarct volume in a focal cerebral ischemia model in rats. Stroke 1993;24:117-121. 170. Frank JI. Large hemispheric infarction, deterioration, and intracranial pressure. Neurology 1995;45:1286-1290. 171. Ropper AH, Shafran B. Brain edema after stroke. Clinical syndrome and intracranial pressure. Arch Neurol 1984;41:26-29. 172. Wardlaw J, Dennis M, Lindley R, Warlow C, Sandercock P, Sellar R. Does early reperfusion of a cerebral infarct influence cerebral infarct swelling in the acute stage or the final clinical outcome? Cerebrovasc Dis 1993;3:86-93. 173. Schwab S, Aschoff A, Spranger M, Albert F, Hacke W. The value of intracranial pressure monitoring in acute hemispheric stroke. Neurology 1996;47:393-398. 174. Ziai WC, Mirski MA, Bhardwaj A. Use of hypertonic saline in ischemic stroke. Stroke 2002;33:11661167. 175. Steiner T, Ringleb P, Hacke W. Treatment options for large hemispheric stroke. Neurology 2001;57: S61-S68. 176. Kasner SE, Demchuk AM, Berrouschot J. Predictors of fatal brain edema in massive hemispheric ischemic stroke. Stroke 2001;32:2117-2123. 177. Krieger DW, Demchuk AM, Kasner SE, Jauss M, Hantson L. Early clinical and radiological predictors of fatal brain swelling in ischemic stroke. Stroke 1999;30:287-292. 178. Oppenheim C, Samson Y, Manai R. Prediction of malignant middle cerebral artery infarction by diffusion-weighted imaging. Stroke 2000;31:2175-2181. 179. Neumann-Haefelin T, Sitzer M, du MdR, Lanfermann H. Prediction of malignant MCA infarction with DWI: pitfalls in hyperacute stroke. Stroke 2001;32:580-583. 180. Barber PA, Davis SM, Darby DG. Absent middle cerebral artery flow predicts the presence and evolution of the ischemic penumbra. Neurology 1999;52:1125-1132. 181. Neumann-Haefelin T, Moseley ME, Albers GW. New magnetic resonance imaging methods for cerebrovascular disease: emerging clinical applications. Ann Neurol 2000;47:559-570. 182. de Haan RJ, Limburg M, Van der Meulen JH, Jacobs HM, Aaronson NK. Quality of life after stroke. Impact of stroke type and lesion location. Stroke 1995;26:402-408. 183. Hofmeijer J, Amelink GJ, Algra A, van Gijn J, Macleod MR, Kappelle LJ, van der Worp HB; the HAMLET investigators. Hemicraniectomy After Middle cerebral artery infarction with Lifethreatening Edema Trial (HAMLET). Protocol for a randomised controlled trial of decompressive surgery in space-occupying hemispheric infarction. Trials 2006;7:29. 39 Chapter 2 40 Rat model of space-occupying infarction Chapter 3 The time course of ischemic damage and perfusion in a rat model of space-occupying cerebral infarction Jeannette Hofmeijer, Wouter B. Veldhuis, Janneke Schepers, Klaas Nicolay, L. Jaap Kappelle, Peter R. Bär, and H. Bart van der Worp Based on Brain Research 2004;1013:74-82 41 Chapter 3 Summary In this chapter a rat model of space-occupying hemispheric infarction is described. For adequate timing of therapy in future experiments, the development of tissue damage, edema formation, and perfusion over time were studied with different MRI techniques. Permanent middle cerebral artery (MCA) occlusion was performed in 32 Fisher344 rats. Forty-six MRI experiments including diffusion weighted (DWI), T2-weighted (T2W), flow-sensitive alternating inversion recovery (FAIR) perfusion-weighted, and T1-weighted (T1W) imaging before and after gadolinium were performed at 1, 3, 8, 16, 24, and 48 hours of ischemia. MCA occlusion consistently led to infarction of the complete MCA territory. Mortality was 75% at 48 hours after occlusion. Lesion volumes as derived from apparent diffusion coefficient- and T2-maps increased to maximum values of 400 ± 48mm3 at 24 hours and 420 ± 54mm3 at 48 hours of ischemia. Midline shift peaked at 24 hours. The area with diffusion-perfusion deficit decreased to a minimum at 24 hours after the onset of ischemia. Perfusion of the contralateral hemisphere dropped at the same time point. Leakage of gadolinium through the blood-brain barrier occurred within 3 hours of ischemia within the entire infarct. Permanent intraluminal MCA occlusion in Fisher-344 rats is an adequate model for space-occupying cerebral infarction. Rats may benefit from intervention aimed at reducing tissue shift and intracranial pressure, and at improving cerebral blood flow, if initiated before 24 hours after MCA occlusion. The value of treatment modalities depending on an intact blood brain barrier should be questioned. 42 Rat model of space-occupying infarction P atients with a hemispheric infarct and massive brain edema formation have a poor prognosis: in prospective intensive care (IC)-based series, the case fatality rate was about 80%, despite maximal medical therapy.1,2 Fatal space-occupying edema occurs in 1 to 5% of the patients with a supratentorial infarct.3 Several treatment strategies have been proposed to limit brain tissue shifts and to reduce intracranial pressure, such as sedation, hyperventilation, and osmotic therapy, but none has been proven effective.4-6 Because of these limitations, there have been proposals for decompressive surgery in patients with large hemispheric infarcts who deteriorate as a result of edema. This intervention leads to reversal of brain tissue shifts and normalization of intracranial pressure (ICP). Non-randomized studies have suggested that hemicraniectomy lowers mortality without increasing the rate of severely disabled survivors.7,8 Support for the efficacy of different treatment modalities has been sought in rat models of focal cerebral ischemia. In these models, decompressive surgery has been shown to reduce mortality and to improve histological and functional outcome.9-12 However, previous studies on the temporal evolution of cerebral infarction in rats have been performed with models of variable or moderate focal cerebral ischemia.13-17 Moreover, the optimal timing of therapy has remained uncertain. An adequate model of space-occupying infarction may provide information on the applicability and optimal timing of medical or surgical intervention. We present a model of space-occupying hemispheric infarction in rats and investigated the temporal evolution of lesion volume, edema formation, perfusion, and blood-brain barrier breakdown. We used permanent intraluminal occlusion of the middle cerebral artery (MCA) in Fisher rats, because in Fisher rats, MCA occlusion leads to larger and more reproducible infarcts than in Wistar or Sprague-Dawley rats.18 The MRI protocol, consisting of diffusion- (DWI), T2- (T2W), post-contrast T1- (T1W), and non-invasive perfusion weighted imaging (PWI) with flow-sensitive alternating inversion recovery (FAIR) was performed at six different time points in the first 48 hours after MCA occlusion. Methods Animal model The experiments were performed according to a protocol approved by the Utrecht University Animal Experiment Ethical Committee. Animals had free access to standard laboratory chow and water. Thirty-two male Fisher rats (F344, Iffa-Credo Broekman, Someren, the Netherlands), weighing 280-320g, were anesthetized by intraperitoneal injection of 0.16mg / kg fentanyl citrate, 5mg / kg fluanisone, and 2.5mg / kg midazolam, followed by subcutaneous injection of 0.05mg / kg atropine sulfate. They were intubated and mechanically 43 Chapter 3 ventilated (Amsterdam Infant Ventilator, MK3, Hoekloos, the Netherlands) with a gas mixture of 35% oxygen and 65% nitrous oxide. During all experiments, rectal temperature was continuously monitored and maintained between 36.5 and 37.5°C by means of a water heating pad. The left femoral artery was cannulated to monitor arterial blood pressure (Datascope 3000 Monitor, Datascope corp), and to obtain blood for arterial blood gas analyses before and after MCA occlusion (ABL 505 / OSM3, Radiometer, Copenhagen, Denmark). When necessary, respiratory adjustments were made to maintain normal blood gas levels. After surgery, the animals received 0.3mg / kg buprenorfine subcutaneously for relief of pain. MCA occlusion was achieved by a minor modification of the intraluminal filament technique, originally described by Koizumi et al.19 The right common (CCA), internal (ICA) and external (ECA) carotid arteries were exposed through a ventral cervical midline incision. The pterygopalatine artery was ligated with a 7.0 prolene suture. After coagulation of the proximal branches of the ECA, the ECA and CCA were ligated with a 7.0 silk suture. A microvascular clip was placed at the origin of the ICA. A 3.0 prolene suture, with a rounded tip and coated with poly-L-lysine (0.1% wt/vol in deionized water; Sigma) as described by Belayev et al.,20 was introduced into the CCA. A 7.0 silk suture was tightened around the CCA and the intraluminal thread, after which the microvascular clip was removed. The thread was gently advanced into the ICA until a slight resistance was felt and the MCA was occluded. Immediately thereafter the incision was closed. The animals were randomly assigned to the MRI measurement time points of 1, 3, 8, and 16 hours after MCA occlusion, and to later time points if they were still alive. For measurements at 1 and 3 hours after occlusion the rats remained under anesthesia with 1% halothane in a mixture of 35% oxygen and 65 % nitrous oxide. For measurements at later time points, animals were re-intubated and ventilated with 1% halothane in the same gas mixture. Rats were immobilized in a stereotactic holder and positioned in an animal cradle. During the MRI experiments, the exhaled CO2 level and blood oxygen saturation were continuously monitored (Datascope Multinex 4200 and Pulse oximeter 8600V, Nonin Medical). If necessary, respiratory adjustments were made. Each day, rats were weighed and examined neurologically according to the scale introduced by Bederson et al.21 and refined by Menzies et al.22 Seven days after MCA occlusion the surviving animals were killed by an intraperitoneal injection of 150 mg pentobarbital. The brains of all animals were carefully evaluated for the presence of subarachnoid hemorrhage. MRI experiments MRI experiments were performed on a 4.7T Varian (Palo Alto, USA) horizontal bore spectrometer. RF-excitation and signal detection were accomplished by means 44 Rat model of space-occupying infarction of a Helmholtz volume coil (diameter 9cm, length 10cm) and an inductively coupled surface coil (diameter 2cm), respectively. A spin-echo sequence was used for determination of the position of the animal in the magnet (echo time (TE) / repetition time (TR) = 40ms / 1s, matrix (M) = 128 x 64, field of view (FOV) = 5.0 x 3.0cm2, 21 1.0mm thick sagital slices, number of excitations (NEX) = 1). Eight contiguous 1.7mm thick transversal slices were planned with respect to the center of the eyes. The first slice was positioned 4mm anterior to the eyes, which is consistent with approximately 5mm posterior to the bregma. A single-scan diffusion-trace MRI sequence (4 b values: 100-1780s / mm2, TE / TR = 100ms / ≥ 2s, M = 128 x 64, FOV = 3.2 x 3.2cm2, NEX = 2) was used to generate quantified images of the tissue water trace apparent diffusion coefficient (ADC). For DWI a double spin-echo pulse sequence was used with four pairs of bipolar gradients with specific predetermined signs in each of the three orthogonal directions.23 The combination of gradient directions leads to a cancellation of all off-diagonal tensor elements, effectively measuring the trace of the diffusion tensor. This provides unambiguous and rotationally invariant ADC values in one experiment, circumventing the need for three separate experiments. To minimize the inherently high sensitivity of diffusion-weighted imaging to motion, data acquisition was triggered to the respiratory cycle. T2W-images were acquired with a multi-echo sequence (TE / TR = 17ms + 7 x 17ms / 3s, M = 256 x 128, FOV = 3.2 x 3.2cm2, NEX = 2). PWI was performed in a single slice through the infarct core (corresponding to slice number five from DWI and T2W images), with the use of the FAIR technique.24 A slice-selective and a non-selective inversion recovery image (Mss and Mns) were acquired with turbo-fast low angle shot (FLASH) acquisition and a sufficient inversion time (TI) to allow inflow of labeled spins into brain tissue. MR parameters were as follows: flip angle (α) = 20º, TE / TR / TI = 3ms / 6ms / 2000ms, predelay = 2.0s (total TR = 4.75s), M = 128 x 128, FOV = 3.2 x 3.2cm2, slice thickness = 1.7mm, NEX = 96. For normalization of the FAIR signal an equilibrium magnetization (M0) image was acquired with the same parameters, but without inversion. For quantification of the FAIR signal a slice-selective T1-map was acquired by Turbo-FLASH Look-Locker acquisition25 (α / TE / TR = 5º / 4.5ms / 11ms, 10 TIs (0.4 + 9 x 1.4s), M = 128 x 128, FOV = 3.2 x 3.2cm2, slice thickness = 1.7mm, NEX = 8). Finally, a pre-contrast T1W dataset (TE / TR = 12.5ms / 0.65s, M = 256 x 128, FOV = 3.2 x 3.2cm2, NEX = 2) and, 12 min after injection of a bolus of 0.1ml / 100mg Gadolinium-DTPA (Gd-DTPA) (469.01mg / ml), contrast-enhanced T1W images were acquired. To minimize interference at the slice boundaries, slices for multi-slice datasets were acquired in alternating order, thus maximizing the time between excitation of two neighboring slices. 45 Chapter 3 Data processing and analysis All images were zero-filled to 256 x 256 prior to analysis. ADC and T2 maps were generated by mono-exponential fitting with IDL (Research Systems, Boulder, USA). Parametric images were analyzed in anatomic regions of interest (ROIs) using in-house software. Calculations of lesion volume were based on ipsilateral ADC or T2 differences of more than 20% as compared to the mean value in the contralateral hemisphere. This threshold corresponds to a difference of more than 2 standard deviations (SD) and is close to the 23% drop in ADC found to correlate with ATP depletion 1 hour after MCA occlusion.26 Measurements of midline shift were performed with the software package ImageBrowser (SISCO / Varian) on slice number 5 of T2W images, according to midline shift = ipsilateral diameter – (total diameter / 2), using the third ventricle as a landmark. PWI data were processed with IDL. Relative FAIR images were obtained by subtracting the non-selective inversion recovery image from the slice selective inversion recovery image and dividing the resulting image by the M0 image ((Mss – Mns) / M0). T1 maps were obtained by mono-exponential fitting with a correction for magnetization saturation.27 From these relative FAIR images and T1 maps, CBF maps in milliliters per minute per 100g tissue were calculated by T1 correction according to Calamante et al.28 assuming perfect inversion, a homogeneous blood brain partition coefficient of 0.9ml / g and a blood T1 of 2.0s. Further analysis was performed on region of interest (ROI) basis with ImageBrowser. ROIs in the ADC maps were defined as those with a ≥ 20% reduction of ADC as compared to the mean value calculated from the contralateral hemisphere (ADC ROI), and in the CBF maps as those with a ≥ 20% reduction of CBF as compared to contralateral (CBF ROI). Areas of the specific ROIs as well as the mean CBF of the contralateral hemisphere were determined. The area with diffusion-perfusion mismatch was calculated by subtracting the area of the ADC ROI from the area of the CBF ROI. Pre- and postcontrast T1W datasets were analyzed with ImageBrowser as described by Blezer et al.29 A Gd-DTPA-enhancement ratio (GER) was calculated according to GER = [(T1W MRI post contrast – T1W MRI precontrast) / T1W MRI precontrast] x 100. Pixel intensities thus display the percentage signal increase due to Gd leakage, and can therefore be used as a semi-quantative measure for blood brain barrier leakage. Mean blood brain barrier leakage was determined in the ADC ROIs. Statistical analysis was carried out using SPSS 9.0 (SPSS Inc, Chicago, USA). Data were evaluated by one way analysis of variance to analyze the effect of time after occlusion, or by Student’s t-test where appropriate. Post hoc tests were performed using Bonferroni’s method. Outcome measures are expressed as means ± SD. Differences were considered significant at levels of P < 0.05. 46 Rat model of space-occupying infarction Table 1 Intra-operative physiological parameters of rats at different time points after MCA occlusion. 1h 3h 8h 16 h 24 h 48 h n=8 n=7 n=6 n=9 n=9 n=7 Weight (g) 287 ± 21 287 ± 19 290 ± 20 289 ± 14 278 ± 17 289 ± 20 Temperature (°C) 36.4 ± 0.7 36.3 ± 0.6 36.7 ± 0.4 36.3 ± 0.5 36.8 ± 0.4 36.8 ± 0.3 BP syst (mmHg) 116 ± 6 119 ± 2 145 ± 56 121 ± 8 120 ± 6 136 ± 45 BP diast (mmHg) 91 ± 7 89 ± 9 99 ± 15 90 ± 11 95 ± 11 100 ± 12 pCO2 (mmHg) 49 ± 4 47 ± 3 49 ± 3 49 ± 4 49 ± 4 49 ± 4 O2 saturation (%) 97 ± 2 96 ± 2 98 ± 1 97 ± 2 98 ± 2 98 ± 2 BP syst indicates systolic blood pressure; BP diast, diastolic blood pressure Results Twenty of the 32 animals were measured once, 10 animals twice and 2 animals were measured at 3 time points. All physiological variables remained within the normal range throughout the operative procedures. There were no differences between the groups for weight, body temperature, blood pressure, and arterial blood gases (Table 1). Eight of the 32 rats (25%) survived up to seven days. All other animals (24 (75%)) died within 48 hours after MCA occlusion (mean survival 24 ± 14 hours). On post-mortem examination, none of the animals showed evidence of subarachnoid hemorrhage. All surviving animals had a continuous decline in body weight up to seven days after MCA occlusion. At day 7, body weight was 68 ± 7% of the initial weight. Directly after MCA occlusion, all animals had a score of 4 on Bederson’s scale, indicating spontaneous contralateral circling.21 In the surviving animals this score started to improve after 4 (n = 5) or 5 (n = 3) days. DWI and T2W MRI Ischemic areas were visualized on ADC and T2 maps and covered fronto-parietal cortical and subcortical areas in all animals. Figure 1a shows a typical example of ADC maps acquired at different time points of a slice through the infarct core (slice number 5). Figure 1b shows the course of lesion development on T2 maps. Lesion volume as deduced from ADC maps gradually increased from 200 ± 65mm3 after 1 hour to a maximum of 400 ± 48mm3 at 24 hours after MCA occlusion (P < 0.001 for the effect of time on lesion volume, Figure 2a). Lesion volume on T2 maps increased from 41 ± 25mm3 after one hour to 420 ± 54mm3 after 48 hours of ischemia (P < 0.001, Figure 2b). 47 Chapter 3 Figure 1 ADC (a) and T2 (b) maps calculated from slice number 5 at different time-points after MCA occlusion. a b Midline shift Midline shift increased from 0.2 ± 0.1mm after one hour to 1.3 ± 0.3mm after 24 hours of ischemia (P < 0.001, Figure 3). Perfusion deficit and diffusion-perfusion mismatch In 12 of the 46 MRI experiments FAIR images could not be analyzed as a result of poor signal : noise ratios due to low global CBF. In the CBF maps of the other 34 experiments, regions with perfusion deficits covering the entire MCA territory were clearly visible (Figure 4a). The mean area of the perfusion deficit on slice number 5 was 64 ± 14mm2. The area of the perfusion deficit did not change significantly over 48 Rat model of space-occupying infarction Figure 2 Lesion volume as calculated from ADC (a) and T2 (b) maps at different time-points after MCA occlusion. Error bars indicate mean ± standard deviation. a b Figure 3 Midline shift at different time-points after MCA occlusion. Error bars indicate mean ± standard deviation. time (P = 0.1). The mean perfusion in the area of perfusion deficit was 28 ± 10ml / min / 100g, and did not change significantly over time (P = 0.7). Since the area with the perfusion deficit remained constant over time, and the area of the lesion on ADC maps increased (from 36 ± 11mm2 to 57 ± 16mm2), the area with diffusion perfusionmismatch decreased from 31 ± 12mm2 after one hour to 4 ± 5mm2 after 24 hours of ischemia (P = 0.007, Figure 4b). Mean CBF in the contralateral hemisphere was lowest at 24 hours after MCA occlusion: 46 ± 8ml / min / 100g compared with 74 ± 25ml / min / 100g at other time points (P = 0.024). 49 Chapter 3 Figure 4 Example of ADC and CBF maps (a) and error bar of the area with diffusion-perfusion mismatch at different time-points after MCA occlusion (b). Error bars indicate mean ± standard deviation. a b T1W MRI before and after Gd-DTPA GER increased from 7 ± 2% at 1 hour to 31 ± 12% at 3 hours of ischemia (P = 0.023), and remained essentially the same thereafter (P = 0.81). There was obvious enhancement in cortical and subcortical infarct regions in all animals. In the contralateral hemisphere we found no increase of Gd enhancement over time (P = 0.5). Discussion We studied changes in lesion volume, tissue shift, cerebral perfusion, and blood brain barrier disturbances after permanent MCA occlusion in Fisher-344 rats. In these rats, intraluminal occlusion of the MCA consistently led to complete, space-occupying MCA infarction. Lesion volumes from ADC and T2 maps gradually increased up to 24 and 48 hours respectively. Brain tissue shifts peaked at 24 hours after occlusion, whereas perfusion levels of the contralateral non-affected hemisphere tended to be lowest and regions with diffusion perfusion mismatch were smallest at 24 hours of ischemia. Gadolinium leakage across the blood-brain barrier increased in the first 3 50 Rat model of space-occupying infarction hours of ischemia, and remained essentially the same thereafter. In this study, permanent intraluminal MCA occlusion in Fisher-344 rats proved to be an adequate model of space-occupying cerebral infarction. The procedure consistently resulted in infarction of the complete MCA territory. The observed lesion volumes of 400 ± 48mm3 on DWI at 24 hours were substantially larger than those of 161 to 225mm3 reported in previous experimental studies on space-occupying cerebral infarction, in which other rat strains were used.9,10,12 The gradual increase in midline shift to a maximum of 1.3 mm also reflects the clinical syndrome of space-occupying infarction in man. In addition, the high mortality of 75% corresponds with mortality levels found in clinical studies.1,2 However, the mortality rate observed in our study may have been influenced by the MRI experiments under general anesthesia. Our finding of consistently large infarcts in Fisher-344 rats is in line with a previous study18 in which infarct volumes of different rat strains were compared after MCA occlusion according to Tamura et al.30 The smaller variability of infarct volume after MCA occlusion in this strain is probably a result of the smaller variability of the cerebral vasculature as compared to Wistar31 and Sprague Dawley32 rats and the lack of proximal MCA collaterals. The consistently large infarct volumes make Fisher-344 probably the preferred strain for studies on space-occupying hemispheric infarction. Histological studies have shown that the volume of the infarct does not grow in size beyond 3 to 4 hours after permanent MCA occlusion.33,34 In the present model lesion volume as calculated from the ADC maps reached its maximum at 24 hours after MCA occlusion. Whereas DWI may suffer from contamination of for instance T2 contrast, contrast on the calculated ADC maps is brought about purely by differences in the apparent diffusion coefficient of water. The ischemia-induced ADC reduction is thought to be related to cytotoxic edema formation and the associated decrease in extracellular space, as a result of a malfunctioning of energy-dependent ion pumps.35 Studies comparing MRI with histopathological data found that a marked ADC reduction after 2 hours of ischemia is related to irreversible tissue damage.35 Several studies of permanent MCA occlusion in rats have reported a gradual extension of the area of DWI hyperintensity.13-17 The fast growth of lesion volume during the first hours is in line with previous reports.13,36 Lesion size on T2W images increased up to 48 hours after occlusion and, at this time point, exceeded the size of the DWI abnormalities. Prolongation of brain tissue water T2, measured by T2W MRI, is thought to reflect vasogenic edema and is probably most pronounced in irreversibly damaged tissue.37-41 It has been demonstrated that the size of the infarct does not increase any more after 24 hours of ischemia, so infarct volume on T2W14 and DWI42 24 hours after occlusion can be considered as the final lesion size. The (non-significant) increase in T2 abnormality between 24 and 48 hours of ischemia therefore may reflect an increase in brain edema. This is in line with other findings suggesting overestimation of infarct volume as a result of the development of 51 Chapter 3 brain edema, especially in the first 3 days of ischemia.43,44 Edema formation and the consequent tissue shifts and ICP increase may compromise CBF in other vascular territories and thereby increase ischemic damage. We used FAIR for PWI. FAIR has been shown to quantify normal CBF values24 and decreases in CBF in a model of forebrain ischemia in gerbils45 as well as in patients with various cerebral diseases.46,47 In contrast to continuous arterial spin labeling, FAIR had not been applied in experimental focal cerebral ischemia before. Analysis of the CBF maps revealed a region with a clear perfusion deficit covering the entire MCA territory on all 34 measurement points. The area with the perfusion deficit remained constant over time, indicating permanent complete MCA occlusion. Since lesion volume increased on ADC maps, the area with diffusion-perfusion mismatch decreased up to 24 hours of ischemia. At 24 hours of ischemia, when midline shift reached a maximum, perfusion levels of the initially non-injured contralateral hemisphere were lowest. In patients with space-occupying cerebral infarction, tissue shifts may lead to compression of other arteries and thereby to additional ischemic damage.48 Despite the substantial midline shift and impaired perfusion of the contralateral hemisphere, we did not find evidence of ischemic damage beyond the ipsilateral MCA territory at 48 hours in this model. The increase of the area with diffusion-perfusion mismatch at 48 hours is caused by the decrease of lesion volume calculated from ADC maps. Since some animals died before the MRI measurement points of 24 and 48 hours, it is possible that the rats that were studied at these time points had less severe ischemic damage. This may partially explain the stabilization of lesion volume after 24 hours. However, these animals also had complete spaceoccupying MCA infarction and a severe neurological deficit, underscoring the validity of the model. The gradual extension of tissue damage as observed on the ADC and T2W maps and substantiated by the decrease in diffusion-perfusion mismatch is consistent with earlier findings of time-dependent perfusion thresholds for tissue damage,49,50 and is not caused by tissue shifts or a rise in ICP. Although CBF of the contra-lateral hemisphere is not the same in all groups of animals, we defined the CBF ROI as a percentage of the CBF in the contralateral hemisphere. We did not use absolute values, because of the fluctuations in resting CBF with this technique. The reduction of 20% corresponds with a difference of about two SD. With the 20% reduction we did not aim to define ischemic tissue, but tissue that is hemodynamicaly compromised, or so called ‘penumbra’ or ‘tissue at risk’ The Gd-DTPA-enhancement ratio, a semi-quantitative measure for blood-brain barrier disruption,29 increased up to 3 hours of ischemia and remained essentially the same thereafter. This finding is consistent with previous reports showing that the blood-brain barrier remains intact for only a few hours after permanent ischemia.51-53 After reperfusion, blood-brain barrier damage is probably even more pronounced.54 52 Rat model of space-occupying infarction This finding supports theoretical concerns on the use of osmotic agents in patients with massive space-occupying infarction. As a result of the destruction of the blood brain barrier, osmotic agents may accumulate in the affected tissue, thereby reversing the osmotic gradient between tissue and plasma and leading to an exacerbation of edema.55 Moreover, as osmotic agents are supposed to exert their main effect in the contralateral hemisphere where the blood brain barrier is still intact, the induced reduction in volume of this hemisphere could lead to an additional increase in brain tissue shifts.48 In conclusion, several observations in our study indicate that permanent intraluminal MCA occlusion in Fisher-344 rats is an adequate model of space-occupying cerebral infarction and may be superior to MCA occlusion in other rat strains. The consistently large infarct volume, substantial midline shift, and high mortality adequately reflect this clinical syndrome in man. In rats, midline shift was maximal at 24 hours, whereas perfusion of the contralateral hemisphere was lowest at this time point. In this model, potential therapies against ICP increase and tissue shifts should therefore be started before 24 hours of ischemia. The usefulness of treatment modalities depending on an intact blood-brain barrier, such as osmotherapy, should be questioned since the integrity of the blood-brain barrier is disturbed from the first hours of ischemia onwards. References 1. 2. 3. 4. 5. 6. 7. 8. 9. Hacke W, Schwab S, Horn M, Spranger M, De Georgia M, von Kummer R. ‘Malignant’ middle cerebral artery territory infarction: clinical course and prognostic signs. Arch Neurol 1996;53:309-315. Wijdicks EF, Diringer MN. Middle cerebral artery territory infarction and early brain swelling: progression and effect of age on outcome. Mayo Clin Proc 1998;73:829-836. Silver FL, Norris JW, Lewis AJ, Hachinski VC. Early mortality following stroke: a prospective review. Stroke 1984;15:492-496. Adams HP, Jr., Adams RJ, Brott T. Guidelines for the early management of patients with ischemic stroke: A scientific statement from the Stroke Council of the American Stroke Association. Stroke 2003;34:1056-1083. van der Worp HB, Kappelle LJ. Complications of acute ischaemic stroke. Cerebrovasc Dis 1998;8:124132. Hofmeijer J, van der Worp HB, Kappelle LJ. Treatment of space-occupying cerebral infarction. Crit Care Med 2003;31:617-625. Rieke K, Schwab S, Krieger D. Decompressive surgery in space-occupying hemispheric infarction: results of an open, prospective trial. Crit Care Med 1995;23:1576-1587. Schwab S, Steiner T, Aschoff A. Early hemicraniectomy in patients with complete middle cerebral artery infarction. Stroke 1998;29:1888-1893. Doerfler A, Forsting M, Reith W. Decompressive craniectomy in a rat model of “malignant” cerebral hemispheric stroke: experimental support for an aggressive therapeutic approach. J Neurosurg 1996;85:853-859. 53 Chapter 3 10. 11. 12. 13. 14. 15. 16. 17. 18. 19. 20. 21. 22. 23. 24. 25. 26. 54 Forsting M, Reith W, Schabitz WR. Decompressive craniectomy for cerebral infarction. An experimental study in rats. Stroke 1995;26:259-264. Engelhorn T, Doerfler A, Kastrup A. Decompressive craniectomy, reperfusion, or a combination for early treatment of acute “malignant” cerebral hemispheric stroke in rats? Potential mechanisms studied by MRI. Stroke 1999;30:1456-1463. Engelhorn T, von Kummer R, Reith W, Forsting M, Doerfler A. What is effective in malignant middle cerebral artery infarction: reperfusion, craniectomy, or both? An experimental study in rats. Stroke 2002;33:617-622. Hoehn-Berlage M, Norris DG, Kohno K, Mies G, Leibfritz D, Hossmann KA. Evolution of regional changes in apparent diffusion coefficient during focal ischemia of rat brain: the relationship of quantitative diffusion NMR imaging to reduction in cerebral blood flow and metabolic disturbances. J Cereb Blood Flow Metab 1995;15:1002-1011. Quast MJ, Huang NC, Hillman GR, Kent TA. The evolution of acute stroke recorded by multimodal magnetic resonance imaging. Magn Reson Imaging 1993;11:465-471. Knight RA, Ordidge RJ, Helpern JA, Chopp M, Rodolosi LC, Peck D. Temporal evolution of ischemic damage in rat brain measured by proton nuclear magnetic resonance imaging. Stroke 1991;22:802808. Dardzinski BJ, Sotak CH, Fisher M, Hasegawa Y, Li L, Minematsu K. Apparent diffusion coefficient mapping of experimental focal cerebral ischemia using diffusion-weighted echo-planar imaging. Magn Reson Med 1993;30:318-325. Verheul HB, Berkelbach van der Sprenkel JW, Tulleken CA, Tamminga KS, Nicolay K. Temporal evolution of focal cerebral ischemia in the rat assessed by T2-weighted and diffusion-weighted magnetic resonance imaging. Brain Topogr 1992;5:171-176. Duverger D, MacKenzie ET. The quantification of cerebral infarction following focal ischemia in the rat: influence of strain, arterial pressure, blood glucose concentration, and age. J Cereb Blood Flow Metab 1988;8:449-461. Koizumi J, Yoshida Y, Nakazawa T, Ooneda G. Experimental studies of ischemic brain edema, I: a new experimental model of cerebral embolism in rats in which recirculation can be introduced in the ischemic area. Jpn J Stroke 1986;8:1-8. Belayev L, Alonso OF, Busto R, Zhao W, Ginsberg MD. Middle cerebral artery occlusion in the rat by intraluminal suture. Neurological and pathological evaluation of an improved model. Stroke 1996;27:1616-1622. Bederson JB, Pitts LH, Tsuji M, Nishimura MC, Davis RL, Bartkowski H. Rat middle cerebral artery occlusion: evaluation of the model and development of a neurologic examination. Stroke 1986;17:472476. Menzies SA, Hoff JT, Betz AL. Middle cerebral artery occlusion in rats: a neurological and pathological evaluation of a reproducible model. Neurosurgery 1992;31:100-106. de Graaf RA, Braun KP, Nicolay K. Single-shot diffusion trace (1)H NMR spectroscopy. Magn Reson Med 2001;45:741-748. Kim SG. Quantification of relative cerebral blood flow change by flow- sensitive alternating inversion recovery (FAIR) technique: application to functional mapping. Magn Reson Med 1995;34:293-301. Haase A, Matthaei D, Bartkowski R, Duhmke E, Leibfritz D. Inversion recovery snapshot FLASH MR imaging. J Comput Assist Tomogr 1989;13:1036-1040. Olah L, Wecker S, Hoehn M. Relation of apparent diffusion coefficient changes and metabolic Rat model of space-occupying infarction 27. 28. 29. 30. 31. 32. 33. 34. 35. 36. 37. 38. 39. 40. 41. 42. 43. 44. 45. disturbances after 1 hour of focal cerebral ischemia and at different reperfusion phases in rats. J Cereb Blood Flow Metab 2001;21:430-439. Deichmann R, Haase A. Quantification of T1 values by SNAPSHOT-FLASH NMR imaging. J Magn Reson 1992;96:608-612. Calamante F, Williams SR, van Bruggen N, Kwong KK, Turner R. A model for quantification of perfusion in pulsed labelling techniques. NMR Biomed 1996;9:79-83. Blezer EL, Nicolay K, Goldschmeding R, Koomans HA, Joles JA. Reduction of cerebral injury in stroke-prone spontaneously hypertensive rats by amlodipine. Eur J Pharmacol 2002;444:75-81. Tamura A, Graham DI, McCulloch J, Teasdale GM. Focal cerebral ischaemia in the rat: 1. Description of technique and early neuropathological consequences following middle cerebral artery occlusion. J Cereb Blood Flow Metab 1981;1:53-60. Herz RC, Jonker M, Verheul HB, Hillen B, Versteeg DH, De Wildt DJ. Middle cerebral artery occlusion in Wistar and Fischer-344 rats: functional and morphological assessment of the model. J Cereb Blood Flow Metab 1996;16:296-302. Fox G, Gallacher D, Shevde S, Loftus J, Swayne G. Anatomic variation of the middle cerebral artery in the Sprague-Dawley rat. Stroke 1993;24:2087-2092. Memezawa H, Smith ML, Siesjo BK. Penumbral tissues salvaged by reperfusion following middle cerebral artery occlusion in rats. Stroke 1992;23:552-559. Kaplan B, Brint S, Tanabe J, Jacewicz M, Wang XJ, Pulsinelli W. Temporal thresholds for neocortical infarction in rats subjected to reversible focal cerebral ischemia. Stroke 1991;22:1032-1039. Moseley ME, Cohen Y, Mintorovitch J. Early detection of regional cerebral ischemia in cats: comparison of diffusion- and T2-weighted MRI and spectroscopy. Magn Reson Med 1990;14:330346. van Dorsten FA, Olah L, Schwindt W. Dynamic changes of ADC, perfusion, and NMR relaxation parameters in transient focal ischemia of rat brain. Magn Reson Med 2002;47:97-104. Brant-Zawadzki M, Pereira B, Weinstein P. MR imaging of acute experimental ischemia in cats. AJNR 1986;7:7-11. Kato H, Kogure K, Ohtomo H. Characterization of experimental ischemic brain edema utilizing proton nuclear magnetic resonance imaging. J Cereb Blood Flow Metab 1986;6:212-221. Hoehn-Berlage M, Tolxdorff T, Bockhorst K, Okada Y, Ernestus RI. In vivo NMR T2 relaxation of experimental brain tumors in the cat: a multiparameter tissue characterization. Magn Reson Imaging 1992;10:935-947. Olah L, Wecker S, Hoehn M. Secondary deterioration of apparent diffusion coefficient after 1-hour transient focal cerebral ischemia in rats. J Cereb Blood Flow Metab 2000;20:1474-1482. Horikawa Y, Naruse S, Tanaka C, Hirakawa K, Nishikawa H. Proton NMR relaxation times in ischemic brain edema. Stroke 1986;17:1149-1152. Loubinoux I, Volk A, Borredon J. Spreading of vasogenic edema and cytotoxic edema assessed by quantitative diffusion and T2 magnetic resonance imaging. Stroke 1997;28:419-426. Lin TN, He YY, Wu G, Khan M, Hsu CY. Effect of brain edema on infarct volume in a focal cerebral ischemia model in rats. Stroke 1993;24:117-121. Gerriets T, Stolz E, Walberer M. Noninvasive quantification of brain edema and the space-occupying effect in rat stroke models using magnetic resonance imaging. Stroke 2004;35:566-571. Pell GS, Lythgoe MF, Thomas DL. Reperfusion in a gerbil model of forebrain ischemia using serial magnetic resonance FAIR perfusion imaging. Stroke 1999;30:1263-1270. 55 Chapter 3 46. 47. 48. 49. 50. 51. 52. 53. 54. 55. 56 Arbab AS, Aoki S, Toyama K. Brain perfusion measured by flow-sensitive alternating inversion recovery (FAIR) and dynamic susceptibility contrast-enhanced magnetic resonance imaging: comparison with nuclear medicine technique. Eur Radiol 2002;11:635-641. Arbab AS, Aoki S, Toyama K. Quantitative Measurement of Regional Cerebral Blood Flow with FlowSensitive Alternating Inversion Recovery Imaging: Comparison with [Iodine 123]-Iodoamphetamin Single Photon Emission CT. AJNR 2002;23:381-388. Frank JI. Large hemispheric infarction, deterioration, and intracranial pressure. Neurology 1995;45:1286-1290. Dijkhuizen RM, Berkelbach van der Sprenkel JW, Tulleken KA, Nicolay K. Regional assessment of tissue oxygenation and the temporal evolution of hemodynamic parameters and water diffusion during acute focal ischemia in rat brain. Brain Res 1997;750:161-170. Hossmann KA. Viability thresholds and the penumbra of focal ischemia. Ann Neurol 1994;36:557565. Menzies SA, Betz AL, Hoff JT. Contributions of ions and albumin to the formation and resolution of ischemic brain edema. J Neurosurg 1993;78:257-266. Gotoh O, Asano T, Koide T, Takakura K. Ischemic brain edema following occlusion of the middle cerebral artery in the rat. I: The time courses of the brain water, sodium and potassium contents and blood-brain barrier permeability to 125I-albumin. Stroke 1985;16:101-109. Olsson Y, Crowell RM, Klatzo I. The blood-brain barrier to protein tracers in focal cerebral ischemia and infarction caused by occlusion of the middle cerebral artery. Acta Neuropathol (Berl) 1971;18:89102. Cole DJ, Matsumura JS, Drummond JC, Schultz RL, Wong MH. Time- and pressure-dependent changes in blood-brain barrier permeability after temporary middle cerebral artery occlusion in rats. Acta Neuropathol (Berl) 1991;82:266-273. Kaufmann AM, Cardoso ER. Aggravation of vasogenic cerebral edema by multiple-dose mannitol. J Neurosurg 1992;77:584-589. Perfusion in rats with MCA occlusion Chapter 4 Perfusion MRI by flow-sensitive alternating inversion recovery and dynamic susceptibility contrast in rats with permanent middle cerebral artery occlusion Jeannette Hofmeijer, Janneke Schepers, H. Bart van der Worp, L. Jaap Kappelle, and Klaas Nicolay Based on NMR in Biomedicine 2005;18:390-394 57 Chapter 4 Summary In this chapter cerebral blood flow (CBF) parameters obtained by dynamic susceptibility contrast magnetic resonance imaging (DSC-MRI) are compared with those obtained by flow-sensitive alternating inversion recovery (FAIR) in brain regions with different perfusion levels in rats with permanent middle cerebral artery (MCA) occlusion. MCA occlusion was performed in 19 rats. T2-weighted MRI, FAIR, and DSC-MRI were performed within 48 hours after occlusion. CBF parameters were analyzed in regions of interest with either prolonged or less prolonged mean transit time (MTT). Ratios of ipsi- versus contralateral CBF values were calculated and tested for correlation and differences between FAIR and DSC-MRI. FAIR-aCBF ratios correlated significantly with DSC-rCBF ratios (r2 = 0.719, P < 0.001). Mean FAIR-aCBF ratio was significantly lower than mean DSC-rCBF ratio in the area with prolonged MTT (0.39 ± 0.14 and 0.50 ± 0.14 respectively, P = 0.008). In the area with less prolonged MTT, mean FAIR-aCBF ratio and mean DSC-rCBF values did not differ significantly (0.78 ± 0.16 and 0.80 ± 0.19 respectively, P = 0.7). It is concluded that FAIR correlates with DSC-MRI if perfusion is preserved. FAIR provides lower CBF values than DSC-MRI if perfusion is reduced and MTT is prolonged. This probable underestimation of perfusion may be caused by transit delays. Care should be taken when quantifying CBF with FAIR and when comparing the results of FAIR-and DSC-MRI in areas with hypoperfusion. 58 Perfusion in rats with MCA occlusion M easurement of perfusion may guide therapeutic decisions in cerebral ischemia.1 Magnetic resonance imaging (MRI) allows perfusion-weighted imaging (PWI) by the use of exogenous contrast agents, and by endogenous contrast mechanisms. PWI by dynamic susceptibility contrast (DSC)-MRI depends on signal changes induced by the passage of a bolus of injected paramagnetic agent, such as a gadolinium chelate. Maps of hemodynamic parameters, including cerebral blood flow (CBF), cerebral blood volume (CBV), and mean transit time (MTT) can be calculated.2 Hemodynamic parameters can be quantified only if an arterial input function can be measured.3 Arterial spin labeling (ASL) is an endogenous contrast method, based on the signal difference detected between magnetically labeled and unlabeled spins in two imaging experiments. ASL is increasingly used for CBF measurements in experimental stroke research.4-6 ASL has been validated for PWI in healthy rat brain7 and after transient ischemia,8 but not in permanent cerebral ischemia. We compared CBF parameters obtained with Flow-sensitive Alternating Inversion Recovery (FAIR), a pulsed ASL technique, with those obtained with DSC-MRI in brain regions with different levels of hypoperfusion in rats with permanent middle cerebral artery (MCA) occlusion. Methods Animal model The protocol was approved by the Utrecht University Animal Experiment Ethical Committee. Animals had free access to food and water. Nineteen male Fisher rats (F344/Ico, Iffa-Credo Broekman, Someren, the Netherlands), weighing 280-320g, were anesthetized by intraperitoneal injection of 0.16 mg/kg fentanyl citrate, 5 mg/ kg fluanisone, and 2.5 mg/kg midazolam, followed by subcutaneous injection of 0.05 mg/kg atropine sulfate. They were intubated and mechanically ventilated (Amsterdam Infant Ventilator, MK3, Hoekloos, the Netherlands) with 35% oxygen and 65% nitrous oxide. During all experiments, rectal temperature was maintained between 36.5 and 37.5°C by means of a water heating pad, and the exhaled CO2 concentration was monitored. The left femoral artery was cannulated to monitor arterial blood pressure (Datascope 3000 Monitor, Datascope corp), and to obtain blood for arterial blood gas analyses before and after MCA-occlusion (ABL 505/ OSM3, Radiometer, Copenhagen, Denmark). When necessary, respiratory adjustments were made. After surgery, the animals received 0.3 mg/kg buprenorfine subcutaneously for relief of pain. MCA-occlusion was achieved by a minor modification of the intraluminal filament technique.9 In short, the right common (CCA), internal (ICA), and external (ECA) carotid arteries were exposed through a ventral cervical midline incision. The pterygopalatine artery, ECA and CCA were ligated with a 7.0 silk suture. A 3.0 prolene 59 Chapter 4 suture, with a rounded tip and coated with poly-L-lysine (0.1% wt/vol in deionized water; Sigma)10 was introduced into the CCA and advanced into the ICA until a slight resistance was felt. A 7.0 silk suture was tightened around the CCA and the intraluminal thread. Thereafter the incision was closed. Twenty-three MRI experiments were performed within 48 hours after MCA occlusion. The animals were re-induced by halothane inhalation followed by intubation. Anesthesia was maintained with 1.0% halothane in a 35% oxygen and 65% nitrous oxide mixture. During MRI experiments the animals were immobilized in an animal cradle using incisor and ear bars. A tail vein was cannulated for injection of gadolinium diethylene triamine pentaacetic acid (Gd-DTPA, Magnevist, Schering, Berlin, Germany). MR experiments Experiments were performed on a 4.7 T Varian INOVA system (Palo Alto, USA). RF excitation and signal detection were accomplished by a Helmholtz volume coil (diameter 9cm, length 10cm) and an inductively coupled surface coil (diameter 2cm), respectively. A spin-echo sequence was used for determination of the position of the animal. Single coronal slice FAIR, T1-weighted and DSC-MRI images were acquired approximately 5mm posterior to bregma. For FAIR PWI a slice-selective and a nonselective inversion recovery image (Mss and Mns) were acquired with turbo-fast low angle shot (FLASH) acquisition and a sufficient inversion time (TI) to allow inflow of labeled spins into brain tissue.8 MRI parameters were as follows: flip angle (α) = 20º, echo time (TE) / repetition time (TR) / TI = 3ms / 6ms / 2000ms, pre-delay = 2.0s (total TR = 4.75s), resolution = 250 x 250um2, slice thickness = 2mm, number of excitations (NEX) = 96. For normalization of the FAIR signal an equilibrium magnetization (M0) image was acquired with the same parameters, without inversion. For quantification of the FAIR signal a slice-selective and non selective T1-map were acquired by TurboFLASH Look-Locker acquisition11 (α / TE / TR = 5º / 3ms / 11ms, 10 TIs (0.4 + 9 x 1.4s), resolution = 250 x 250um2, slice thickness = 2mm, NEX = 8). The FAIR experiment took 15 minutes. For DSC-MRI (acquired immediately after FAIR PWI) FLASH acquisition (α / TE / TR = 12º / 7ms / 11ms, resolution = 500 x 500um2, slice thickness = 2 mm, and NEX = 1) was used following injection of 0.5 mmol / kg Gd-DTPA as a bolus of 0.1 mg / 100mg, injected after twelve images in less than 0.7s, with a temporal resolution of 0.7s per image. Generation of hemodynamic parameter maps All images were zero-filled to 256 x 256. T1 maps were generated by mono-exponential fitting on a pixel-by-pixel basis, and corrected for longitudinal saturation as described 60 Perfusion in rats with MCA occlusion by Deichmann et al.12 Relative FAIR images (FAIR-rCBF in %) were obtained by subtracting the non-selective inversion recovery image from the slice selective inversion recovery image and dividing the result by the M0 image ((Mss–Mns) / M0). From the relative FAIR images and T1 maps, CBF maps in milliliters per minute per 100g tissue were calculated by T1 correction according to Calamante et al.,13 assuming perfect inversion, a homogeneous blood-brain partition coefficient of 0.9ml / g, and an arterial blood T1 of 2.0s. The resulting FAIR-CBF values will be referred to as FAIR apparent CBF values (FAIR-aCBF in ml / min / 100g tissue), to emphasize that no correction was made for transit delays and that quantification may thus not be absolute. A gamma-variate fit was used to derive hemodynamic parameters from DSC-MRI images, yielding maps of relative CBF (DSC-rCBF), relative CBV, and relative MTT in arbitrary units. The annotation “relative” indicates that no absolute quantification was performed by deconvolution with an arterial input function. T1 and FAIR image calculations were performed in IDL (Interactive Data Language, Research Systems, Boulder, CO). The gamma-variate fits of DSC-MRI data were fitted as described in Kluytmans et al. using in-house software.14 Further analysis was performed on region of interest (ROI) basis with the software package ImageBrowser (Varian). ROIs were defined using an MTT map. Two ROIs were drawn in the ipsilateral hemisphere. The first ROI was drawn according to MTT (ROI) > 1.5 x mean MTT (contralateral), and will be referred to as the ‘ROI with prolonged MTT’. The second ROI included the rest of the hemisphere, and will be referred to as the ‘ROI with less prolonged MTT’. Parts of the hemisphere in which perfusion was too low to fit were excluded. Large cerebral venous structures were excluded using the CBV map, according to CBV > mean CBV (contralateral) + 2 standard deviations (SD). Ipsilateral ROIs were mirrored in the mid-sagittal line to obtain contralateral ROIs. Ratios of ipsi- versus contralateral CBF values were calculated for these ROIs. Statistical analysis Data are expressed as mean ± SD. For analysis, data from measurements at different time-points were pooled. The difference between the data obtained from the two perfusion MRI techniques was tested for significance using a Student’s t-test. Linear (Pearson’s) correlation coefficients between FAIR and DSC-CBF parameters were calculated. Correlation and differences were considered significant if P < 0.05. Results Intraoperative physiological variables remained within the normal range in all animals (data not shown). Ischemic areas were visualized on T2, MTT, DSC- and FAIR-perfusion maps, and covered the complete MCA territory in all animals. The development of tissue damage and edema formation in these rats has been described in a separate 61 Chapter 4 article.15 In the lesion core signal-to-noise ratio (SNR) was too low to determine MTT. This area was excluded from analysis in all rats. Regions with prolonged MTT values included the watershed areas between the MCA and anterior cerebral artery (ACA). Regions with less prolonged MTT were found in the ACA territory (Figure 1). FAIR-aCBF ipsi- versus contralateral CBF ratios were significantly correlated with DSC-rCBF ratios (r2 = 0.719, P < 0.001; Figure 2). The mean MTT ipsi- versus contralateral ratio was 2.0 ± 0.4 in ROIs with prolonged MTT and 1.0 ± 0.1 in ROIs with less prolonged MTT (P < 0.001). In the area with prolonged MTT, mean FAIRaCBF ratio was significantly lower than mean DSC-rCBF ratio (0.39 ± 0.14 and 0.50 ± 0.14 respectively, P = 0.008). In the area with less prolonged MTT, mean FAIR-aCBF ratio and mean DSC-rCBF values did not differ significantly (0.78 ± 0.16 and 0.80 ± 0.19 respectively, P = 0.7; Figure 3). Figure 1 Examples of maps of (a) local anatomy with regions of interest, (b) FAIR-aCBF, (c) DSC-rCBF, and (d) MTT. a b c d In Figure 1a, the infarct core is colored in black (excluded from analysis), the ROI with prolonged MTT (watershed area) in dark grey, and the ROI with less prolonged MTT (ACA area) in light grey. Figure 2 Scatter plot of ipsiversus contralateral FAIR-aCBF as a function of ipsi- versus contralateral DSC-rCBF per rat (each dot). 62 Perfusion in rats with MCA occlusion Figure 3 Ipsi- versus contralateral ratios for FAIR-aCBF and DSC-rCBF in areas with different MTT values. Error bars indicate means and standard deviations; *, a statistically significant difference (P < 0.05). Discussion In this study, both FAIR and DSC-MRI were sensitive to spatial differences in CBF in rats with permanent MCA occlusion. A significant linear correlation was found between the ratios of FAIR values and relative DSC-CBF indices. However, in regions with prolonged MTT, FAIR ipsi- versus contralateral CBF ratios were lower than DSC-MRI ratios. This indicates that FAIR detected larger CBF reductions than DSCrCBF in those regions. Underestimation of CBF with ASL in hypoperfused brain regions has been reported before.16,17 This is generally attributed to transit delays, which are longer in areas with reduced perfusion. These transit delays cause loss of label in ASL-based imaging, and may lead to a lower measured CBF.18 We found prolonged MTT values mainly in watershed areas. In permanent vessel occlusion, transit delays in watershed areas are probably longer, since blood supply is dependent on collateral pathways. Among the ASL methods, FAIR is reported to suffer only moderately from transit delays, because the distance between the site of labeling and the acquisition slice is relatively small. However, in regions with reduced perfusion, especially in regions supplied by collateral flow, transit delays may be significant even for FAIR imaging. They may lead to a reduction of measured CBF of up to 20% with transit delays of 500 ms.18 DSC-MRI may be considered as standard MRI modality in clinical practice, and in different animal models. Our study demonstrates that (semi-) quantitative 63 Chapter 4 ASL results cannot always be compared with qualitative or quantitative DSC-MRI parameters, especially in areas with low CBF.19 A reduction of the gap between the site of labeling and the acquisition slice may improve FAIR PWI, even in regions with CBF reductions.20 This would also improve SNR, which is known to be low for ASL-based PWI.20,21 Alternatively, transit delays may be fitted by analysis of FAIR signal acquired with multiple TI values. However, acquisitions with multiple TI values are time consuming. In addition, ASL implementations that compensate intrinsically for transit delays by means of saturation pulses may be used.22-24 These sequences set the transit delay, such that it is a known parameter in the quantification of perfusion. However, with very long transit delays (as can be found in severe stroke), these methods are less applicable. Still, the benefits of FAIR-based PWI may outweigh the above-mentioned disadvantages for most experimental purposes. Quantification of perfusion with DSCMRI is probably equally difficult, since determination of an arterial input function is problematic in small rodents, given the small diameter of the feeding arteries. FAIR images can be acquired with a better spatial resolution, since spatial resolution is restricted by the need for a good temporal resolution in bolus passage sampling, such as DSC-MRI. Moreover, ASL based imaging is non-invasive and has the potential of unlimited repeats without delays. The role of ASL in clinical stroke care is still limited, because of its low sensitivity in regions with reduced CBF, and the relatively long duration of scanning. However, in follow-up studies of stroke patients, ASL may be of use, especially if repeated measurements are necessary. FAIR has been used to measure reserve capacity of cerebral auto-regulation after acetazolamide challenge, which would have been difficult with injections of contrast agent.25 Continuous ASL offers the possibility to measure perfusion territories using a single labeling coil placed on one of the carotid arteries.26 Transit delays may even be used to detect hemodynamic compromise in an early stage.14 Moreover, ASL may be used to detect activation patterns in functional MRI.27 Our results emphasize that FAIR correlates with the commonly used DSC-MRI if perfusion is relatively preserved. Care should be taken when quantifying CBF with FAIR in regions with reduced perfusion, and further research is required to determine the nature of the FAIR signal under different levels of hypoperfusion. The use of ASL sequences that limit the effect of transit delays is recommended. References 1. 2. 64 Wu O, Koroshetz WJ, Ostergaard L. Predicting tissue outcome in acute human cerebral ischemia using combined diffusion- and perfusion-weighted MR imaging. Stroke 2001;32:933-942. Rosen BR, Belliveau JW, Vevea JM, Brady TJ. Perfusion imaging with NMR contrast agents. Magn Reson Med 1990;14:249-265. Perfusion in rats with MCA occlusion 3. 4. 5. 6. 7. 8. 9. 10. 11. 12. 13. 14. 15. 16. 17. 18. 19. 20. Rempp KA, Brix G, Wenz F, Becker CR, Guckel F, Lorenz WJ. Quantification of regional cerebral blood flow and volume with dynamic susceptibility contrast-enhanced MR imaging. Radiology 1994;193:637-641. Olah L, Wecker S, Hoehn M. Secondary deterioration of apparent diffusion coefficient after 1-hour transient focal cerebral ischemia in rats. J Cereb Blood Flow Metab 2000;20:1474-1482. Hilger T, Niessen F, Diedenhofen M, Hossmann KA, Hoehn M. Magnetic resonance angiography of thromboembolic stroke in rats: indicator of recanalization probability and tissue survival after recombinant tissue plasminogen activator treatment. J Cereb Blood Flow Metab 2002;22:652-662. Hofmeijer J, Schepers J, Veldhuis WB, Nicolay K, Kappelle LJ, Bar PR, van der Worp HB. Delayed decompressive surgery increases apparent diffusion coefficient and improves peri-infarct perfusion in rats with space-occupying cerebral infarction. Stroke 2004;35:1476-1481. Calamante F, Lythgoe MF, Pell GS. Early changes in water diffusion, perfusion, T1, and T2 during focal cerebral ischemia in the rat studied at 8.5 T. Magn Reson Med 1999;41:479-485. Schepers J, Veldhuis WB, Pauw RJ, de Groot JW, van Osch MJP, Nicolay K, van der Sanden BPJ. Comparison of FAIR perfusion kinetics with DSC-MRI and functional histoloy, in a model of transient ischemia. Mag Reson Med 2004;51:312-320. Koizumi J, Yoshida Y, Nakazawa T, Ooneda G. Experimental studies of ischemic brain edema, I: a new experimental model of cerebral embolism in rats in which recirculation can be introduced in the ischemic area. Jpn J Stroke 1986;8:1-8. Belayev L, Alonso OF, Busto R, Zhao W, Ginsberg MD. Middle cerebral artery occlusion in the rat by intraluminal suture. Neurological and pathological evaluation of an improved model. Stroke 1996;27:1616-1622. Haase A, Matthaei D, Bartkowski R, Duhmke E, Leibfritz D. Inversion recovery snapshot FLASH MR imaging. J Comput Assist Tomogr 1989;13:1036-1040. Deichmann R, Haase A. Quantification of T1 values by SNAPSHOT-FLASH NMR imaging. J Magn Reson 1992;96:608-612. Calamante F, Williams SR, van Bruggen N, Kwong KK, Turner R. A model for quantification of perfusion in pulsed labelling techniques. NMR Biomed 1996;9:79-83. Kluytmans M, van der Grond J, Viergever MA. Gray matter and white matter perfusion imaging in patients with severe carotid artery lesions. Radiology 1998;209:675-682. Hofmeijer J, Veldhuis WB, Schepers J, Nicolay K, Kappelle LJ, Bar PR, van der Worp HB. The time course of ischemic damage and cerebral perfusion in a rat model of space-occupying cerebral infarction. Brain Res 2004;1013:74-82. Hunsche S, Sauner D, Schreiber WG, Oelkers P, Stoeter P. FAIR and dynamic susceptibility contrastenhanced perfusion imaging in healthy subjects and stroke patients. J Magn Reson Imaging 2002;16:137-146. Yoneda K, Harada M, Morita N, Nishitani H, Uno M, Matsuda T. Comparison of FAIR technique with different inversion times and post contrast dynamic perfusion MRI in chronic occlusive cerebrovascular disease. Magn Reson Imaging 2003;21:701-705. Buxton RB, Frank LR, Wong EC, Siewert B, Warach S, Edelman RR. A general kinetic model for quantitative perfusion imaging with arterial spin labeling. Magn Reson Med 1998;40:383-396. Perthen JE, Calamante F, Gadian DG, Connelly A. Is quantification of bolus tracking MRI reliable without deconvolution? Magn Reson Med 2002;47:61-67. Sidaros K, Andersen IK, Gesmar H, Rostrup E, Larsson HB. Improved perfusion quantification in FAIR imaging by offset correction. Magn Reson Med 2001;46:193-197. 65 Chapter 4 21. 22. 23. 24. 25. 26. 27. 66 Schepers J, Garwood M, van der SB, Nicolay K. Improved subtraction by adiabatic FAIR perfusion imaging. Magn Reson Med 2002;47:330-336. Wong EC, Buxton RB, Frank LR. Quantitative imaging of perfusion using a single subtraction (QUIPSS and QUIPSS II). Magn Reson Med 1998;39:702-708. Wong EC, Buxton RB, Frank LR. Implementation of quantitative perfusion imaging techniques for functional brain mapping using pulsed arterial spin labeling. NMR Biomed 1997;10:237-249. Luh WM, Wong EC, Bandettini PA, Hyde JS. QUIPSS II with thin-slice TI1 periodic saturation: a method for improving accuracy of quantitative perfusion imaging using pulsed arterial spin labeling. Magn Reson Med 1999;41:1246-1254. Yen YF, Field AS, Martin EM. Test-retest reproducibility of quantitative CBF measurements using FAIR perfusion MRI and acetazolamide challenge. Magn Reson Med 2002;47:921-928. Zaharchuk G, Ledden PJ, Kwong KK, Reese TG, Rosen BR, Wald LL. Multislice perfusion and perfusion territory imaging in humans with separate label and image coils. Magn Reson Med 1999;41:1093-1098. Detre JA, Wang J. Technical aspects and utility of fMRI using BOLD and ASL. Clin Neurophysiol 2002;113:621-634. Decompressive surgery in rats with space-occupying infarction Chapter 5 Delayed decompressive surgery improves peri-infarct perfusion in rats with space-occupying cerebral infarction Jeannette Hofmeijer, Janneke Schepers, Wouter B. Veldhuis, Klaas Nicolay, L. Jaap Kappelle, Peter R. Bär, and H. Bart van der Worp Based on Stroke 2004;35:1476-81 67 Chapter 5 Summary There is no conclusive support that decompressive surgery in late stages of spaceoccupying cerebral infarction will improve outcome. In this chapter the effects of delayed decompressive surgery on the development of tissue damage, edema formation, and cerebral perfusion are studied with different magnetic resonance imaging (MRI) techniques in a rat model of space-occupying cerebral infarction. Permanent middle cerebral artery (MCA) occlusion was performed in 6 Fisher-344 rats. Decompressive surgery was performed 17 hours after the occlusion. Each animal was assessed before, 2 hours after, and 4 hours after surgery by means of diffusionweighted (DW), T2-weighted (T2W), and flow-sensitive alternating inversion recovery (FAIR) perfusion-weighted MRI. Ischemic damage was also evaluated in hematoxylineosin-stained brain sections. Lesion volume as derived from ADC maps decreased from 522 ± 98 mm3 before to 405 ± 100 mm3 (P = 0.016) 4 hours after decompressive surgery, whereas lesion volume from T2 maps increased from 420 ± 66 mm3 before to 510 ± 92 mm3 (P = 0.048). Midline shift decreased from 1.4 ± 0.1 mm to 0.5 ± 0.2 mm (P = 0.001). Blood flow in the non-infarcted area of the ipsilateral hemisphere improved from 25 ± 9 ml/min/ 100g tissue to 38 ± 9 ml/min/100g tissue (P = 0.035). Despite the pseudonormalization of ADC, irreversible damage was found in the entire MCA territory on histological evaluation. In rats with space-occupying cerebral infarction, delayed decompressive surgery leads to increased perfusion of non-infarcted areas, possibly preventing secondary damage. The observed decrease in lesion volume derived from ADC maps is probably a result of increased extracellular water formation. There are no signs that this reflects rescue of ischemic tissue. 68 Decompressive surgery in rats with space-occupying infarction F atal brain edema occurs in 1 to 5% of patients with a supratentorial infarct.1 Patients with a hemispheric infarct and space-occupying brain edema have a poor prognosis: in prospective series, mortality was about 80%.2 There are no medical treatment strategies of proven efficacy.3 Non-randomized studies have suggested that decompressive surgery lowers mortality without increasing the percentage of severely disabled survivors.4,5 Studies of decompressive surgery in rat models of brain infarction have shown that this procedure reduces infarct volume and improves cerebral blood flow when performed within 12 hours after occlusion of the middle cerebral artery (MCA).6-9 Infarct volume increased with time between MCA-occlusion and decompressive surgery.7,9 Clinical studies also favor early over delayed hemicraniectomy.5 Still, most clinicians will be inclined to perform this procedure only in later stages, when the patient has signs of impending herniation. There is no experimental support that therapy in this stage will improve outcome. In the present study, we investigated the effect of delayed hemicraniectomy on lesion volume, midline shift, and cerebral blood flow in rats with space-occupying cerebral infarction. MRI experiments, consisting of diffusion- (DW), perfusion- (PW), and T2weighted (T2W) imaging, were performed directly before and at two time points after decompression. Methods Animal model The experiments were performed according to a protocol approved by the Utrecht University Animal Experiment Ethical Committee. Animals had free access to food and water. Six male Fisher344 rats (Iffa-Credo Broekman, Someren, the Netherlands), weighing 280-320 g, were anesthetized by intraperitoneal injection of 0.16 mg/kg fentanyl citrate, 5 mg/kg fluanisone, and 2.5 mg/kg midazolam, followed by subcutaneous injection of 0.05 mg/kg atropine sulfate. They were intubated and mechanically ventilated (Amsterdam Infant Ventilator, MK3, Hoekloos, the Netherlands) with 35% oxygen and 65% nitrous oxide. During all experiments, rectal temperature was maintained between 36.5 and 37.5°C by means of a water heating pad. The left femoral artery was cannulated to monitor arterial blood pressure (Datascope 3000 Monitor, Datascope corp), and to obtain blood for arterial blood gas analysis before and after MCAocclusion (ABL 505/ OSM3, Radiometer, Copenhagen, Denmark). When necessary, respiratory adjustments were made. After surgery, the animals received 0.3 mg/kg buprenorfine subcutaneously for relief of pain. MCA-occlusion was achieved by a minor modification of the intraluminal filament technique.10 In short, the right common (CCA), internal (ICA), and external (ECA) 69 Chapter 5 carotid arteries were exposed through a ventral cervical midline incision. The pterygopalatine artery, ECA and CCA were ligated with a 7.0 silk suture. A 3.0 prolene suture, with a rounded tip and coated with poly-L-lysine (0.1% wt/vol in deionized water; Sigma)11 was introduced into the CCA and advanced into the ICA until a slight resistance was felt. A 7.0 silk suture was tightened around the CCA and the intraluminal thread. Thereafter the incision was closed. Sixteen hours after MCA occlusion the rats were neurologically examined according to the scale introduced by Bederson et al.,12 and refined by Menzies et al.13 Thereafter, they were intubated and ventilated with halothane 1% in a mixture of 35% oxygen and 65% nitrous oxide for MRI experiments and hemicraniectomy. They remained under anesthesia during the surgical procedure and all measurements. Decompressive surgery was performed 17 hours after MCA-occlusion as described by Forsting et al.7 Rats were immobilized in a stereotactic holder, the skull was exposed through a dorsal midline incision, and the temporal muscle was removed partially. A bone flap of 10 x 5 mm2 was created in the parietal and temporal bone using a highspeed mini-drill. The dura was opened by a large incision. Thereafter, the temporal muscle and the skin were adapted and sutured in place. MRI experiments MRI experiments were performed just before, and at 2 and 4 hours after decompressive surgery on a 4.7 T Varian (Palo Alto, USA) horizontal bore spectrometer. RF excitation and signal detection were accomplished by a Helmholtz volume coil (diameter 9 cm, length 10 cm) and an inductively coupled surface coil (diameter 2 cm), respectively. A spin-echo sequence was used for determination of the position of the animal (echo time (TE) / repetition time (TR) = 40 ms / 1 s, matrix (M) = 128 x 64, field of view (FOV) = 5.0 x 3.0 cm2, 21 1.0 mm thick sagital slices, number of excitations (NEX) = 1). Eight contiguous 1.7 mm thick transversal slices were planned, with the first slice 4mm anterior to the center of the eyes. A single-scan diffusion-trace MRI sequence (4 b values: 100-1780 s / mm2, TE / TR = 100 ms / 2 s, M = 128 x 64, FOV = 3.2 x 3.2 cm2, NEX = 2) was used to generate quantified images of the tissue water trace apparent diffusion coefficient (ADC). For DW imaging a double spin-echo pulse sequence was used with four pairs of bipolar gradients with specific predetermined signs in each of the three orthogonal directions.14 The combination of gradient directions leads to a cancellation of all off-diagonal tensor elements, measuring the trace of the diffusion tensor. This provides unambiguous and rotationally invariant ADC values in one experiment. To minimize the high sensitivity of DW imaging to motion, the acquisition was triggered to the respiratory cycle. T2W-images were acquired using a multi-echo sequence (TE / TR = 17 ms + 7 x 17 m s / 3 s, M = 256 x 128, FOV = 3.2 x 3.2 cm2, NEX = 2). To minimize interference at the slice boundaries, slices were acquired in alternating order. 70 Decompressive surgery in rats with space-occupying infarction PWI was performed in a single slice through the infarct core (corresponding to slice number five from diffusion and T2-weighted images), with use of Flow-sensitive Alternating Inversion Recovery (FAIR).15 A slice-selective and a non-selective inversion recovery image (Mss and Mns) were acquired with turbo-fast low angle shot (FLASH) acquisition and a sufficient inversion time (TI) to allow inflow of labeled spins into brain tissue.16 MR parameters were as follows: flip angle (α) = 20º, TE / TR / TI = 3 ms / 6 ms / 2000 ms, pre-delay = 2.0 s (total TR = 4.75 s), M = 128 x 128, FOV = 3.2 x 3.2 cm2, slice thickness = 1.7 mm, NEX = 96. For normalization of the FAIR signal an equilibrium magnetization (M0) image was acquired with the same parameters, without inversion. For quantification of the FAIR signal a slice-selective T1-map was acquired by Turbo-FLASH Look-Locker acquisition17 (α / TE / TR = 5º / 3 ms / 11 ms, 10 TIs (0.4 + 9 x 1.4 s), M = 128 x 128, FOV = 3.2 x 3.2 cm2, slice thickness = 1.7 mm, NEX = 8). Data processing and analysis All images were zero-filled to 256 x 256. ADC and T2 maps were generated by monoexponential fitting with IDL (Interactive Data Language, Research Systems, Boulder, USA). Parametric images were analyzed in anatomic regions of interest using in-house software. Calculations of lesion volume were based on ipsilateral ADC or T2 differences of more than 20% as compared to the mean value in the contralateral hemisphere. This threshold corresponds to a difference of more than 2 standard deviations (SD) but is less influenced by noise than the SD. Moreover, this threshold is close to the 23% drop in ADC found to correlate with ATP depletion 1 hour after MCA occlusion.18 Measurements of midline shift were performed with the software package ImageBrowser (SISCO/Varian) at the level of the infarct core, corresponding to slice number five of T2W images, according to the formula midline shift = (total diameter / 2) – contralateral diameter, using the third ventricle as a landmark. PWI data were processed with IDL. Relative FAIR images were obtained by subtracting the non-selective inversion recovery image from the slice selective inversion recovery image and dividing the result by the M0 image ((Mss – Mns) / M0). T1 maps were obtained by mono-exponential fitting with a correction for magnetization saturation.19 From these relative FAIR images and T1 maps, CBF maps in milliliters per minute per 100g tissue were calculated by T1 correction according to Calamante et al.,20 assuming perfect inversion, a homogeneous blood brain partition coefficient of 0.9 ml / g and a blood T1 of 2.0 s. Further analysis was performed on region of interest (ROI) basis with ImageBrowser. Infarct ROIs were drawn in the T2 maps and defined as those regions with a 20% increase of T2 as compared to the mean value of the contralateral hemisphere. Peri-infarct ROIs were defined as those covering the non-infarcted parts of the affected hemisphere, excluding the ventricles. Contralateral ROIs consisted of the whole contralateral hemisphere. These ROIs were transferred to 71 Chapter 5 the CBF maps and CBF was determined in the infarct core, in the peri-infarct region, and in the contralateral hemisphere. Statistical analyses of MR parameters before and 2 and 4 hours after surgical treatment were performed by means of repeated measures analysis of variance. Outcome measures are expressed as means ± SD. Differences were considered significant at levels of P < 0.05. Histology After the MRI experiments the rats were killed by an intraperitoneal injection of 150 mg pentobarbital. The brains were removed and stored in a 4% phosphate-buffered formaldehyde solution. After dehydration in a phosphate-buffered 25%-sucrose solution, coronal cryosections (25 μm) were cut and stained with hematoxylin and eosin for histopathological evaluation. Infarct was defined as the area of pallor caused by loss of affinity for hematoxylin affecting all cell types except infiltrated inflammatory cells. Cells were considered to be damaged irreversibly when the cytoplasm had become intensively eosinophilic while the nucleus had become pyknotic, or when the cellular nucleic acids had completely lost their affinity for hematoxylin (ghost neurons).21 Counts of neurons and glial cells were performed in specified areas of infarcted cortex and striatum, and in the corresponding areas of the contralateral hemisphere, in slices corresponding to slice number five of the MRI experiments. Results Intraoperative physiological variables remained within the normal range (data not shown). Before hemicraniectomy all rats had a neurological score of 4 according to Bederson et al., indicating spontaneous contralateral circling.12 DW and T2W MRI Before surgery, ischemic areas were visualized on ADC and T2 maps and covered the complete MCA territory in all animals (Figure 1). Mean ADC and T2 values of the contralateral hemisphere, threshold values, and values within the infarct and in periinfarct tissue are presented in Table 1. Values of the contralateral hemisphere (and thereby threshold values) did not change significantly over time. Mean lesion volume as calculated from the ADC maps decreased from 522 ± 98 mm3 before to 458 ± 92 mm3 at two hours, and 405 ± 100 mm3 at four hours after surgery (P = 0.016, Figure 2a). Of total lesion volume calculated from ADC maps, the percentage with increased ADC values increased from 4.6 ± 2.7% to 10.4 ± 6.7% (P = 0.05). In contrast to ADC lesion volumes, lesion volume as deduced from T2 maps increased from 420 ± 66 mm3 before to 457 ± 94 mm3 at two hours, and 510 ± 92 mm3 at four hours after surgery (P = 0.048, Figure 2b). 72 Decompressive surgery in rats with space-occupying infarction Figure 1 ADC (a), T2 (b), and CBF (c) maps of a slice through the infarct core before and four hours after hemicraniectomy. Midline shift Mean midline shift decreased from 1.4 ± 0.1 mm before to 0.7 ± 0.3 mm at two hours, and 0.5 ± 0.2 mm at four hours after surgery (P = 0.001, Figure 3). The total volume of the affected hemisphere as deduced from T2 maps increased from 767 ± 19 mm3 to 820 ± 58 mm3 at two hours, and 830 ± 52 mm3 at four hours after surgery (P = 0.033). PWI From 18 FAIR experiments the results of 5 could not be analyzed due to poor signalto-noise ratios as a result of low global CBF. The calculated perfusion maps of the remaining experiments showed regions with perfusion deficits covering the entire MCA territory. Figure 1c shows an example of a perfusion map before and four hours after surgery. Perfusion before and after surgery could be analyzed in 5 out of six rats. The mean CBF in the peri-infarct region increased from 25 ± 9 ml / min / 100 g tissue 73 Chapter 5 Table 1 ADC and T2 values of the contralateral hemisphere, threshold values, and values within the lesion and in peri-infarct tissue. Before surgery Two hours after surgery Four hours after surgery P ADC contralateral (x10-4mm2/s) 7.5 ± 0.5 7.3 ± 0.4 7.5 ± 0.5 0.4 ADC threshold for decreased values (x10-4mm2/s) 6.0 ± 0.4 5.8 ± 0.3 6.0 ± 0.4 0.4 ADC within part of lesion with decreased values (x10-4mm2/s) 4.4 ± 0.2 4.5 ± 0.3 4.4 ± 0.7 0.3 ADC threshold for increased values (x10-4mm2/s) 9.0 ± 0.5 8.8 ± 0.5 9.0 ± 0.6 0.4 ADC within part of lesion with increased values (x10-4mm2/s) 10.6 ± 0.5 11.1 ± 0.4 12.1 ± 1.6 0.8 ADC peri-infarct (x10-4mm2/s) 7.1 ± 0.4 7.4 ± 0.4 7.6 ± 0.4 0.7 T2 contralateral (ms) 53 ± 2 53 ± 4 53 ± 2 0.2 T2 threshold value (ms) 63 ± 3 64 ± 5 64 ± 3 0.6 T2 within lesion (ms) 80 ± 4 81 ± 3 83 ± 3 0.5 T2 peri-infarct (ms) 51 ± 1 52 ± 2 52 ± 2 0.6 Data are expressed as mean ± SD Figure 2 Lesion volume from ADC (a) and T2 (b) maps before and after hemicraniectomy. a Error bars indicate mean ± SD. 74 b Decompressive surgery in rats with space-occupying infarction Figure 3 Midline shift before and after decompressive surgery. Error bars indicate mean ± SD. Figure 4 Regional cerebral blood flow (rCBF) in noninfarcted tissue of the ipsilateral hemisphere before and after decompressive surgery. Error bars indicate mean ± SD. before to 37 ± 9 ml / min / 100 g tissue two hours, and 38 ± 5 ml / min / 100 g tissue four hours after surgery (P = 0.035, Figure 4). There were no differences in CBF of the infarcted region and the contralateral hemisphere before and after decompressive surgery (14 ml / min / 100 g tissue before and 12 ml / min / 100 g tissue four hours after surgery, P = 0.6, and 46 ml / min / 100 g tissue before and 44 ml / min / 100 g tissue four hours after surgery, P = 0.4 respectively). 75 Chapter 5 Histology Signs of irreversible tissue damage, including cytoplasmic eosinophilia affecting both neurons and glial cells, clumping of nuclear chromatin, nuclear pyknosis, and ghost neurons, were observed in the entire MCA territory, including the parts where ADC had normalized. Cell density in the infarct as compared to contralateral decreased with 40 ± 14% in the cortex (P = 0.001) and 43 ± 14% in the striatum (P = 0.001). Discussion In this model of space-occupying cerebral infarction, hemicraniectomy and durotomy performed when herniation was impending gave rise to a reversal of midline shift and an increase in blood flow in non-infarcted tissue of the ipsilateral hemisphere. Apparent lesion volume as measured on ADC maps decreased, suggesting a beneficial effect on ischemic damage. However, there was an increase in lesion volume calculated from T2 maps, and histological evaluation demonstrated irreversible damage in the complete MCA territory, including areas with (pseudo)normalized ADC values. ADC reductions in ischemic tissue are thought to be caused by cytotoxic edema and the associated decrease in extracellular water,22 and are a very sensitive indicator of early ischemic brain damage.23 Several investigators have observed reversal of DWI abnormalities after transient focal ischemia followed by delayed recurrence.23,24 It has been suggested that this phenomenon is caused by secondary ischemic damage, after an initial recovery of the energy status.24 This is the first report that describes an increase in ADC after hemicraniectomy in space-occupying cerebral infarction. As irreversible injury was observed on histological evaluation in the entire MCA territory, including the parts where ADC had (pseudo)normalized, the decrease in apparent lesion volume on ADC maps did not indicate tissue recovery. Given the increase in apparent lesion volume as deduced from the T2 maps, and the increase in volume of the entire hemisphere, we suggest that the increase of ADC values may be caused by an increased extracellular space by vasogenic edema. T2 prolongation of brain tissue water, measured by T2W MRI, is thought to reflect vasogenic edema and is probably most pronounced in irreversibly damaged tissue.25An increase in extracellular space in our model may be explained by a decreased interstitial pressure caused by the bony decompression. This is in line with earlier findings. Cooper et al. showed that bony decompression resulted in exacerbation of edema in a cold lesion model in dogs.26 In a rat model of MCA infarction, Engelhorn and colleagues found that lesion volume after a combination of reperfusion and decompressive surgery was significantly larger than lesion volume after reperfusion alone.8 Analysis of the CBF maps revealed a region with a clear perfusion deficit covering the entire MCA territory in each animal. After surgery, the area with the perfusion deficit slightly increased, probably as a result of the increase of lesion volume by edema. 76 Decompressive surgery in rats with space-occupying infarction CBF in the non-infarcted parts of the ipsilateral hemisphere improved significantly, supporting one of the rationales of decompressive surgery, which is to decrease ICP to improve cerebral perfusion.4,5 However, in contrast to a previous study,8 we observed no increase in CBF in the territory of the occluded artery. In this previous study, treatment was applied one hour after MCA occlusion, when there was no evidence of edema yet. It is unlikely that delayed hemicraniectomy as performed in our study (and in most patients), exerts a beneficial effect by saving penumbral tissue in the area of the occluded artery. Protection from secondary ischemic damage, if present, may be explained by the increase of CBF in the non-affected vascular territories, thereby preventing recruitment of these territories in the infarct. We did not compare ADC, T2, and CBF values after decompressive surgery with those in controls. However, in a previous study on the natural course of tissue damage in rats with space-occupying infarction we did not observe normalization of ADC values, nor reversal of midline shift or increase of CBF in the peri-infarct region in the time period between 16 and 24 hours after MCA-occlusion. In conclusion, delayed decompressive surgery might be beneficial by reversal of tissue shifts with improvement of CBF in vascular territories surrounding the infarct. We found no beneficial effect of surgery on infarcted tissue within the MCA territory. Therefore, the intervention may only prevent additional damaged in already severely affected patients. Randomized clinical studies focusing on functional outcome are required before implementing this strategy as a standard treatment modality.27 References 1. 2. 3. 4. 5. 6. 7. 8. Silver FL, Norris JW, Lewis AJ, Hachinski VC. Early mortality following stroke: a prospective review. Stroke 1984;15:492-496. Hacke W, Schwab S, Horn M, Spranger M, De Georgia M, von Kummer R. ‘Malignant’ middle cerebral artery territory infarction: clinical course and prognostic signs. Arch Neurol 1996;53:309-315. Hofmeijer J, van der Worp HB, Kappelle LJ. Treatment of space-occupying cerebral infarction. Crit Care Med 2003;31:617-625. Rieke K, Schwab S, Krieger D. Decompressive surgery in space-occupying hemispheric infarction: results of an open, prospective trial. Crit Care Med 1995;23:1576-1587. Schwab S, Steiner T, Aschoff A. Early hemicraniectomy in patients with complete middle cerebral artery infarction. Stroke 1998;29:1888-1893. Doerfler A, Forsting M, Reith W. Decompressive craniectomy in a rat model of “malignant” cerebral hemispheric stroke: experimental support for an aggressive therapeutic approach. J Neurosurg 1996;85:853-859. Forsting M, Reith W, Schabitz WR. Decompressive craniectomy for cerebral infarction. An experimental study in rats. Stroke 1995;26:259-264. Engelhorn T, Doerfler A, Kastrup A. Decompressive craniectomy, reperfusion, or a combination for early treatment of acute “malignant” cerebral hemispheric stroke in rats? Potential mechanisms studied by MRI. Stroke 1999;30:1456-1463. 77 Chapter 5 9. 10. 11. 12. 13. 14. 15. 16. 17. 18. 19. 20. 21. 22. 23. 24. 25. 26. 78 Engelhorn T, von Kummer R, Reith W, Forsting M, Doerfler A. What is effective in malignant middle cerebral artery infarction: reperfusion, craniectomy, or both? An experimental study in rats. Stroke 2002;33:617-622. Koizumi J, Yoshida Y, Nakazawa T, Ooneda G. Experimental studies of ischemic brain edema, I: a new experimental model of cerebral embolism in rats in which recirculation can be introduced in the ischemic area. Jpn J Stroke 1986;8:1-8. Belayev L, Alonso OF, Busto R, Zhao W, Ginsberg MD. Middle cerebral artery occlusion in the rat by intraluminal suture. Neurological and pathological evaluation of an improved model. Stroke 1996;27:1616-1622. Bederson JB, Pitts LH, Tsuji M, Nishimura MC, Davis RL, Bartkowski H. Rat middle cerebral artery occlusion: evaluation of the model and development of a neurologic examination. Stroke 1986;17:472476. Menzies SA, Hoff JT, Betz AL. Middle cerebral artery occlusion in rats: a neurological and pathological evaluation of a reproducible model. Neurosurgery 1992;31:100-106. de Graaf RA, Braun KP, Nicolay K. Single-shot diffusion trace (1)H NMR spectroscopy. Magn Reson Med 2001;45:741-748. Kim SG. Quantification of relative cerebral blood flow change by flow- sensitive alternating inversion recovery (FAIR) technique: application to functional mapping. Magn Reson Med 1995;34:293-301. Schepers J, Veldhuis WB, Pauw RJ, de Groot JW, van Osch MJP, Nicolay K, van der Sanden BPJ. Comparison of FAIR perfusion kinetics with DSC-MRI and functional histology, in a model of transient ischemia. Magn Reson Med 2004;51:312-320. Haase A, Matthaei D, Bartkowski R, Duhmke E, Leibfritz D. Inversion recovery snapshot FLASH MR imaging. J Comput Assist Tomogr 1989;13:1036-1040. Olah L, Wecker S, Hoehn M. Relation of apparent diffusion coefficient changes and metabolic disturbances after 1 hour of focal cerebral ischemia and at different reperfusion phases in rats. J Cereb Blood Flow Metab 2001;21:430-439. Deichmann R, Haase A. Quantification of T1 values by SNAPSHOT-FLASH NMR imaging. J Magn Reson 1992;96:608-612. Calamante F, Williams SR, van Bruggen N, Kwong KK, Turner R. A model for quantification of perfusion in pulsed labelling techniques. NMR Biomed 1996;9:79-83. Garcia JH, Yoshida Y, Chen H. Progression from ischemic injury to infarct following middle cerebral artery occlusion in the rat. Am J Pathol 1993;142:623-635. Moseley ME, Cohen Y, Mintorovitch J. Early detection of regional cerebral ischemia in cats: comparison of diffusion- and T2-weighted MRI and spectroscopy. Magn Reson Med 1990;14:330346. Dijkhuizen RM, Berkelbach van der Sprenkel JW, Tulleken KA, Nicolay K. Regional assessment of tissue oxygenation and the temporal evolution of hemodynamic parameters and water diffusion during acute focal ischemia in rat brain. Brain Res 1997;750:161-170. Neumann-Haefelin T, Kastrup A, de Crespigny A. Serial MRI after transient focal cerebral ischemia in rats: dynamics of tissue injury, blood-brain barrier damage, and edema formation. Stroke 2000;31:1965-1972. Olah L, Wecker S, Hoehn M. Secondary deterioration of apparent diffusion coefficient after 1-hour transient focal cerebral ischemia in rats. J Cereb Blood Flow Metab 2000;20:1474-1482. Cooper PR, Hagler H, Clark WK, Barnett P. Enhancement of experimental cerebral edema after Decompressive surgery in rats with space-occupying infarction 27. decompressive craniectomy: implications for the management of severe head injuries. Neurosurgery 1979;4:296-300. Hofmeijer J, Amelink GJ, Algra A, van Gijn, Macleod MR, Kappelle LJ, van der Worp HB; the HAMLET investigators. Hemicraniectomy After Middle cerebral artery infarction with Lifethreatening Edema Trial (HAMLET). Protocol for a randomised controlled trial of decompressive surgery in space-occupying hemispheric infarction. Trials 2006 Sep 11;7:29. 79 Part II Clinical studies Predictors of life-threatening edema in MCA infarction Chapter 6 Predictors of life-threatening brain edema in middle cerebral artery infarction. Jeannette Hofmeijer, Ale Algra, L. Jaap Kappelle, and H. Bart van der Worp Submitted 83 Chapter 6 Summary Clinical and preclinical data suggest that outcome after decompressive surgery in patients with middle cerebral artery (MCA) infarction and life-threatening edema formation is better if surgery is performed as early as possible. Early treatment requires identification of patients at risk. In this chapter a systematic review is presented to identify predictors of life-threatening edema formation in patients with MCA infarction. Medline and Embase have been searched from January 1966 and January 1974, respectively, to August 2006 for cohort and case-control studies on predictors of lifethreatening edema formation in patients with MCA infarction. Data were extracted by means of a predefined data extraction form and crude data were used to calculate risk ratios, odds ratios, or weighted mean differences. Infarct size was the major determinant for life-threatening edema formation. Cut-off values of 50% of the MCA territory for infarct size and 66% for perfusion deficit were found. Other associated signs were early mass effect, involvement of other vascular territories, higher body temperature, internal carotid artery occlusion, and need for mechanical ventilation. However, predictive values were moderate. In patients with MCA infarction, prediction of life-threatening edema formation remains tempting. The size of the ischemic area is the major determinant. However, single predictors lack sufficient predictive value to select candidates for surgical decompression before the onset of clinical signs of herniation. 84 Predictors of life-threatening edema in MCA infarction L arge middle cerebral artery (MCA) infarcts may be complicated by spaceoccupying and life-threatening edema formation, usually between the second and fifth day after stroke onset. Case fatality rates as high as 78% have been reported.1 Fatal edema occurs in 1 to 5% of all patients with a supratentorial infarct.2,3 Because of the limitations of medical treatment strategies to reduce edema formation,4 there have been proposals for decompressive surgery consisting of a large hemicraniectomy and a duraplasty.5,6 In rats with MCA infarction, surgical decompression reduced infarct volume when performed early after the onset of ischemia, but not when start of treatment was delayed.7 Although a recent systematic review of retrospective and uncontrolled clinical studies failed to show a significant effect of timing of surgery on outcome,8 a large prospective study not included in this review also suggested that functional outcome is better if treatment is started early, even before clinical deterioration.6 If this suggestion is confirmed in ongoing clinical trials, patients at risk for developing fatal edema should be identified as early as possible. Different early predictors of life-threatening edema are described, ranging from the presence of vomiting to infarct volume.9-30 In this chapter the results of a systematic review to identify predictors of life-threatening edema formation in patients with MCA infarction are presented. Methods Search strategy and selection criteria We searched Medline from January 1966 to August 2006 and Embase from January 1974 to August 2006 for cohort and case control studies on predictors of life-threatening cerebral edema formation in patients with MCA infarction. The search terms we used were (predict* OR prognos*) AND (stroke OR (middle cerebral artery)) AND (edema OR oedema OR swelling OR malignant OR herniation OR space-occupying). Publications were identified independently by the first and by the last author. Reference lists of all relevant publications were checked for additional articles. This method of cross-checking was continued until no further studies were found. Inclusion criteria Criteria for inclusion of studies were (1) cohort or case-control studies conducted among patients with acute MCA infarction, published as full articles and written in English, French, or German; (2) clear description of the cohort in cohort studies and the control group in case-control studies; and (3) outcome defined as neurological deterioration with a decrease in consciousness and signs of cerebral herniation as a result of cerebral edema formation. Studies were also included if patients died or if invasive therapeutic procedures, such as decompressive surgery, were performed, after 85 Chapter 6 the above outcome criteria had been met. For inclusion in the meta-analysis, the studies had to present crude data to allow recalculations in our analyses. However, we were not able to use individual patient data. Studies were excluded if (1) only a radiological outcome was described, without clinical information, or if (2) brain imaging was not performed and patients with other lesions than MCA infarcts could therefore not be excluded. Data were included only once if used in multiple publications. Data extraction Data were systematically extracted with use of a predefined data extraction form. Difficulties were resolved by consensus discussions between the first and the last author. Data analysis Cochrane Collaboration’s Review Manager software (version 4.2) was used for analysis. For analysis of crude data, data of at least two studies had to be available on the same potential risk factor. Crude dichotomous data were used to construct 2 x 2 tables and to calculate risk ratios (RR) or odds ratios (OR) where appropriate, as well as positive and negative predictive values. Data from cohort and case control studies were not combined. Crude continuous data were used to calculate weighted mean differences (WMD). If there was statistically significant heterogeneity among the results of included studies (P < 0.05), we used random- instead of fixed-effect models, because these include both within-study variance and between-study variation in the assessment of uncertainty of the meta-analysis.31 Data are presented as point estimates with 95% confidence intervals (CI). The boxes in the figures describe the study size, with larger boxes for larger studies. Results Of 955 citations identified after searching Medline and Embase with the above search terms, 84 were considered possibly relevant after screening of titles and abstracts, and reviewed in detail. Hand searching of the reference lists of these articles identified 8 additional publications that were reviewed in detail. Sixty-nine articles were excluded for one or more of the following reasons: no crude patient data (n=6), duplicate publication (n=3), intervention study without analysis of predictors of edema formation (n=2), inclusion of stroke subtypes different from MCA infarction (n=12), death as an outcome measure without adequate specification of its cause (n=20), other clinical outcome measure different from neurological deterioration as a result of life-threatening edema formation (n=23), insufficient definition of the outcome measure (n=6), and outcome defined as severe edema on CT or MRI without clinical information (n=11). Eventually, 23 articles were included in the present review. Details 86 Predictors of life-threatening edema in MCA infarction of these studies are given in Table 1. We were able to perform a meta-analysis of crude data on the following potentially predictive factors: age, sex, gaze palsy on admission, need for mechanical ventilation, infarct size, early signs of infarction on CT, side of infarction, involvement of other vascular territories besides the MCA territory, results of perfusion-weighted imaging, thrombolysis, recanalization, hemorrhagic transformation, blood glucose on admission, diabetes mellitus, temperature on admission, infarct etiology, and the presence of MCA or ICA occlusion. Because of the use of different cut-off points, it was not possible to combine data for some other predictive factors. For this reason, data on infarct severity, level of consciousness, blood pressure, fever, brain atrophy, neuro-monitoring, cerebral perfusion, and midline shift are presented in a narrative synthesis. Meta-analysis The results of the meta-analysis are summarized in Table 2. We found a total of 27 factors evaluated for their relation with life-threatening edema formation, of which 12 were statistically significant predictors of this complication. For infarct size, cut-off values of 50%, 66%, and 100% of the MCA territory were used,9,18,20,21,23,26,27,29,30 and for perfusion deficit a cut-off value of 66% of the MCA territory. A need for mechanical ventilation, infarction of more than 66% of the MCA territory, and a perfusion deficit larger than 66% of the MCA territory were found to be the strongest predictors in this meta-analysis. However, predictive values were only moderate, and highly dependent on the incidence of life-threatening edema, which ranged from 10 % to 78 % in the cohort studies included in this review (Table 2). MCA occlusion was associated with a significantly reduced risk. Meta-analyses of age, infarct size, involvement of other vascular territories, and thrombolysis are presented in Figures 1-4. Narrative synthesis The investigators of 11 studies reported on severity of the neurological deficits at admission and risk of life-threatening edema formation. Severity was expressed as a score on either the National Institutes of Health Stroke Scale (NIHSS)11,12,14,21,26,27,30 or the Scandinavian Stroke Scale (SSS)1,10,15,29 and results were reported either on the basis of mean or median scores, or dichotomized with different cut-off points. This made a formal meta-analysis impossible. In general, patients with life-threatening edema had higher severity scores, either with statistical significance,26,27,30 or not.1,11,12,14,15,21,29 Level of consciousness on admission was not significantly different in most studies.10,22,24,25,28 In only one study level of consciousness on admission was significantly lower in patients going on to life-threatening edema, but the definition of this factor was unclear.18 Neither mean arterial pressure on admission,11 nor systolic blood pressure 12 hours 87 Chapter 6 Mechanical ventilation Gaze palsy Severity Gender Age Time from onset of symptoms (hours) Population Incidence of life-threatening edema (%) Controlsb Cohorta Cases Year Table 1 Details of included studies. Berrouschot9 1998 11 108 10 ICU 6 - - - - - Berrouschot10 1998 37 53 70 ICU 12 + - + + - Bosche,11 2003 ICU 12 + + + - - - - - - 14 31 45 Dohmen,12 17 34 50 Heiss17 16 34 47 1998 6 20 30 New 6 - Foerch 2004 16 51 31 New 6 + + + - + Gerriets15 2001 12 42 29 New 16 +c +c + - - ICU 12 + c + + + + New 18 +c + - - - ICU 48 + + - - - Firlik 13 14 1 Hacke and 1996 43 55 78 Haring16 1999 31 18 2001 94 19 Krieger 1999 23 New 6 + - - - - Kucinski20 1998 17 74 23 New 6 +c - - - - Lee21 2004 10 31 32 New 6 + - + - - Limburg22 1990 6 26 23 New 24 - - - + - Manno23 2003 22 36 61 New 12 + + - - - Maramattom24 2004 14 24 58 ICU ? +c + - + - Mori25 2001 34 55 62 New 6 + + - - - Oppenheim26 2000 10 28 36 New 14 + + + - - 27 Ryoo 2004 11 27 41 New 6 - - + - - Stolz29 2002 9 21 43 New 120 + + + - - + + - Schwab28 Kasner 30 Thomalla 2003 11 31 201 47 112 37 30 New 6 + a c b Variables were included in this table only if studied by at least two authors. indicates cohort studies; , casecontrol studies; c, not included in the meta-analysis because of lack of sufficient crude data; d, not included in 88 Predictors of life-threatening edema in MCA infarction Perfusion-weighted imaging Thrombolysis Recanalization Hemorrhagic transformation Atrophy Blood glucose/ DM Temperature/ fever Blood pressure/ hypertension - +d - - - - - - - - - - - - - - - - - - - - + + - - - - - - - + - +d + - + - - + + - - + - - +c - - +d +c - + - - - - - - - - - + + - + - - - + - + + - - - + c c Midline shift Involvement of other territories - Neuro-monitoring Infarct side - Vessel occlusion Early signs + Etiology/ atrial fibrillation Infarct size Table 1 – Continued – - - - - - - - - - - - - + - + - - + - - + + - - - - - +d + + + + + - - - - - - - - - - + + - + + + - - - + - + + + + + + + - - + - - + - - - - + + + + + + - - + + - - - - + - - - - - - + - - + - + + + - - - - - - - - + - - - - - - +d - - - - - - - - - - - + - + - - + - - - - - - - + - - - - +d + - - - - - - - - - + - + - - +d + - + + + - - - - - - - + + - + + - - - - - - - - - + - - + +d - + + - - - - - - - - + - - + - - + - - - - - - - - - - - + + - + - +c + - - - - - - + + - - the meta-analysis because it was not possible to combine data; ICU, target population admitted to an intensive care unit; new, target population consists of newly admitted stroke patients; time from onset of symptoms, time window within which factors were assessed. 89 90 20,25,28 10,18,19 12,13,18,25 History of diabetes mellitus 14,18 Blood glucose levels on admission Hemorrhagic transformation Recanalization Thrombolysis 11,14,18,23,25,28,30 Low perfusion levels in other vascular territories a RR 1.2 (0.9-1.6) WMD 0.20 mmol/l (–0.7-1.0) RR 1.8 (0.9-3.7) RR 0.9 (0.2-2.5) RR 0.8 (0.6-1.1) RR 3.7 (2.0-6.6) a RR 7.7 (2.5-24) a Perfusion deficit larger than 66% of the MCA territory21,27 c 21,27 RR 2.6 (2.0-3.2) a Involvement of other vascular territories c 14,21,24-27,29 Early mass effect RR 1.1 (0.8-1.3) a RR 1.5 (1.2-2.0) RR 0.9 (0.7-1.2) RR 7.0 (2.5-19.4) Left-sided versus right-sided1,11,10,21,23,26,30 18,20 b 18,20 Obscuration of the lentiform nucleus on admission Infarction of the complete MCA territory RR 7.5 (3.9-14.3) Infarct size larger than 66% of the MCA territory 9,26,29,30 OR 9.2 (4.0-20.9) 9,21,27,29 Infarct size larger than 50% of the MCA territory a a RR 2.0 (1.5-2.6) 16,19 Infarct size larger than 50% of the MCA territory a 18,21,23 RR 10 (2.1-51) RR 1.2 (0.8-1.7) Conjugate gaze palsy on admission10,22,24 Mechanical ventilation during admission RR 0.9 (0.8-1.1) Men versus women1,11,14,18,23-26,29,30 a WMD –3.2 (–6.4-0.0)e Age10,11,14,18,19,21,23,25,26,29 d 1,14 Estimates (95% CI) Predictor Table 2 Results of the meta-analysis. fixed fixed random random random fixed fixed fixed fixed fixed fixed random fixed fixed fixed fixed fixed fixed random 44 40 49 45 50 36 36 44 47 40 40 21 22 46 56 55 50 42 49 75 30 43 85 73 86 47 51 39 71 86 65 72 60 48 60 54 43 47 78 86 69 53 64 58 91 90 68 96 55 48 Effect Overall incidence Positive predictive Negative modeling life-threatening value (%) predictive edema (%) value (%) Chapter 6 fixed fixed random fixed fixed fixed fixed fixed 45 37 46 38 31 31 61 42 63 51 21 25 33 55 82 57 40 63 70 Effect Overall incidence Positive predictive Negative modeling life-threatening value (%) predictive edema (%) value (%) indicates statistically significant difference; b, Sylvian fissure obscuration, effacement of sulci, and lateral ventricle compression on admission; c, anterior or posterior cerebral artery; d, the time between the onset of symptoms and the need for ventilation was between 3 hours and 5 days in one study;28 e,the mean age of patients with malignant edema was 3.2 years lower than the mean age of patients without. Perfusion-weighted imaging was performed by CT,13,21,3 MRI,30 positron emission tomography (PET),11,12,17 or single photon emission CT (SPECT).9,22 Recanalization was measured by digital subtraction angiography (DSA). Hemorrhagic transformation had to be well defined and primary intra-parenchymal hemorrhages had to be excluded. MCA occlusion was measured by DSA,1,20 CT angiography,29 or MR angiography,30 and internal cerebral artery (ICA) occlusion by DSA1,20 or MR angiography.26,30 a RR 1.0 (0.6-1.6) RR 2.8 (1.9-4.1) OR 4.0 (0.5-35.7) Hyperdense ICA sign on CT18,21 ICA occlusion 1,20,26,30 Hyperdense MCA sign on CT a RR 1.2 (0.9-1.5) a 16,19 RR 0.5 (0.3-0.7) RR 0.7 (0.4-1.2) RR 1.1 (0.5-2.3) 18,21,23,24 Hyperdense MCA sign on CT MCA occlusion 1,20,27,30 Cardioembolic infarction 14,30 Athero-thrombotic infarction WMD 0.3°C (0.0-0.6) a Core temperature on admission10,11 14,30 Estimates (95% CI) Predictor Table 2 – Continued – Predictors of life-threatening edema in MCA infarction 91 Chapter 6 Figure 1 Weighted mean differences in age between patients with and without life-threatening edema. Figure 2 Early infarct size and the risk of life-threatening edema formation. a b Figure 3 Involvement of other vascular territories besides the MCA territory and the risk of life-threatening edema formation. 92 Predictors of life-threatening edema in MCA infarction Figure 4 Thrombolysis and the risk of life-threatening edema formation. after the onset of symptoms,19 nor peak or through systolic pressure18 were identified as a risk factor for life-threatening edema. Cerebral atrophy, a history of hypertension, fever on admission, and atrial fibrillation did not lead to significantly altered risk in one cohort,14 and one case-control19 study. Lower levels of extracellular non-transmitter amino acids were found in non-infarcted ipsilateral tissue in the first 12 hours after the onset of symptoms in patients who developed life-threatening edema.12,17 In the first 12 hours after the onset of symptoms intra-cranial pressure (ICP) values could not discriminate between patients who did and did not go on to life-threatening edema.11,17 In one study higher ICP values were found in patients with life-threatening edema, but it is unclear at what time points these ICP measurements were done.1 Measurements of midline shift were presented in 5 cohort studies1,15,24,25,29 and 1 case-control study.16 Measurements were performed at different time points and several cut-off points were used. Generally, differences in midline shift between life-threatening and ‘non-life-threatening’ infarctions became statistically significant in the later stages of the disease. Maximum midline shift was found earlier in patients with life-threatening edema than in patients without (day 2-4 versus day 3-7),1 and the progression of midline shift was faster.29 Independent predictors Multivariate analysis of predictors of life-threatening edema formation was performed in 9 studies.9,10,18,19,21,23,26,27,29 Variables found as independent predictors are summarized in Table 3. 93 Chapter 6 Table 3 Variables found as independent predictors of life-threatening edema formation. Predictor Interval from onset Estimates (95% CI) of symptoms (hours) SPECT activity deficit of the complete MCA territory9 6 SPECT graded scale > 1409 6 RR 79 (11-569) 18 a Attenuated cortico-med contrast 16 18 RR 40 (10-161) History of hypertension OR 3.0 (1.2-7.6) History of congestive heart failure18 OR 2.1 (1.5-3.0) WBC 18 48 OR 1.07 / 1000 WBC/ ul (1.01-1.14) 48 OR 3.9 (2.4-6.4) >50% MCA on CT 48 OR 6.3 (3.5-11.6) Additional vascular territories on CT18 48 OR 3.3 (1.2-9.4) Nausea or vomiting 24 OR 5.1 (1.7-15.3) Systolic BP > 18019 12 OR 4.2 (1.4-12.9) Hyperdense MCA sign 12 OR 29 (1.6-524) >50% MCA CT23 12 OR 14 (1.04-189) Volume DWI > 145 cm 14 a Hypoperfusion on CT time to peak map27 6 OR 150 (a) WBC > 10.000/ul18 18 19 23 3 26 SPECT indicates single photon emission computed tomography; MCA, middle cerebral artery; WBC, white blood cells; BP, blood pressure; DWI, diffusion-weighted imaging; a, exact estimate could not be derived from the publication. Discussion The major determinants of developing fatal brain edema after MCA infarction are the size of the infarct and the size of the area with perfusion deficit. In this systematic review, infarct size larger than 50% of the MCA territory and a perfusion deficit larger than 66% were identified as risk factors. Also involvement of additional vascular territories besides that of the MCA was significantly associated with life-threatening edema formation. However, predictive values were low. To date, selection of patients for surgical decompression before the onset of clinical signs of herniation remains tempting. In two recent studies, lesion volume of more than 145 ml on diffusion-weighted imaging (DWI) within 14 hours26 and more than 82 ml on apparent diffusion coefficient maps in the first six hours30 predicted life-threatening edema formation with high predictive values. These findings could not be subjected to meta-analysis, because they were not tested in different studies. Still, measurement of infarct volume with 94 Predictors of life-threatening edema in MCA infarction DWI is simple, reliable, and promising and should be used in future studies on the prediction of life-threatening edema formation. Several findings were anticipated. First, in general, greater stroke severity was associated with an increased risk of the development of life-threatening edema. Second, we found a borderline significant association with age, in which lower age was associated with a greater risk. The lack significance is probably the result of the heterogeneity between the included studies. Third, body temperature on admission was marginally higher in patients with life-threatening edema, than in patients without. Stroke patients with fever have a worse prognosis than patients with a normal or low body temperature.32 Moreover, it has been shown that moderate hypothermia can help to control critically elevated ICP values in severe space-occupying edema after MCA infarction.33 Paradoxically, MCA occlusion was associated with a lower risk. This is probably a result of selection bias within the studies included in the meta-analysis, because the group of patients with an MCA occlusion consisted mainly of patients with distal MCA occlusions, whereas occlusions of the MCA trunk have been associated with poor outcome.34,35 The presence of a hyperdense MCA sign is usually indicative of MCA trunk occlusion36 and in the present meta-analysis a hyperdense MCA sign on CT led to an increased RR and OR, but the results were not statistically significant.16,18,19,21,23,24 Patients with ICA occlusions had a higher risk. This group had mainly intracranial carotid occlusions, which is considered a poor prognostic sign.1,20 The association between levels of non-transmitter and transmitter amino acids in peri-infarct tissue and life-threatening edema formation has been evaluated in two studies. Whereas levels of transmitter amino acids discriminated only in later stages of the disease,12,17 a statistically significant association was found between lower levels of extracellular non-transmitter amino acids within 12 hours after stroke onset and formation of life-threatening edema.11 This is possibly a result of dilution due to excessive vasogenic edema formation within the infarct, spreading into the extracellular space of peri-infarct tissue.11 Sensitivities and specificities of approximately 80% were found at specific cut-off values. These observations may emphasize the relevance of vasogenic edema in peri-infarct zones, but it is unlikely that invasive neuro-monitoring will be used on a large scale in future stroke units. For this reason, the clinical applicability of these results is probably low. The most important limitation of this systematic review and meta-analysis is the large variety of inclusion criteria and methods of analysis among the included studies, although we studied only articles on severe MCA infarction. The different incidences of life-threatening brain edema in the cohort studies reflect the large differences in inclusion criteria of these studies. As compared with newly admitted patients, predictive values in patients admitted to a neuro-critical care unit were higher, because a priori chances were higher (Table 1). Moreover, the more time passes after stroke onset, the 95 Chapter 6 more precise the prognostic reliability of the different parameters becomes. Especially signs that are directly related to edema formation, such as raised ICP, midline shift, and need for mechanical ventilation, are late rather than early outcome predictors. Early prediction of the development of life-threatening edema formation and selection of patients for decompressive surgery is difficult in patients with MCA infarction. The size of the ischemic area appears to be the major determinant. MRI with DWI is probably the most reliable tool. However, none of the clinical and radiological variables has sufficient predictive value to be useful for individual patients. Prediction would probably improve if combinations of symptoms and signs would be used. Future prognostic studies, in which all of the determinants described in the present review are evaluated, may help to establish reliable predictive models. References 1. 2. 3. 4. 5. 6. 7. 8. 9. 10. 11. 12. 13. 96 Hacke W, Schwab S, Horn M, Spranger M, De Georgia M, von Kummer R. ‘Malignant’ middle cerebral artery territory infarction: clinical course and prognostic signs. Arch Neurol 1996;53:309-315. Heinsius T, Bogousslavsky J, Van Melle G. Large infarcts in the middle cerebral artery territory. Etiology and outcome patterns. Neurology 1998;50:341-350. Silver FL, Norris JW, Lewis AJ, Hachinski VC. Early mortality following stroke: a prospective review. Stroke 1984;15:492-496. Hofmeijer J, van der Worp HB, Kappelle LJ. Treatment of space-occupying cerebral infarction. Crit Care Med 2003;31:617-625. Rieke K, Schwab S, Krieger D. Decompressive surgery in space-occupying hemispheric infarction: results of an open, prospective trial. Crit Care Med 1995;23:1576-1587. Schwab S, Steiner T, Aschoff A. Early hemicraniectomy in patients with complete middle cerebral artery infarction. Stroke 1998;29:1888-1893. Doerfler A, Forsting M, Reith W. Decompressive craniectomy in a rat model of “malignant” cerebral hemispheric stroke: experimental support for an aggressive therapeutic approach. J Neurosurg 1996;85:853-859. Gupta R, Connolly ES, Mayer S, Elkind MS. Hemicraniectomy for massive middle cerebral artery territory infarction: a systematic review. Stroke 2004;35:539-543. Berrouschot J, Barthel H, von-Kummer R, Knapp WH, Hesse S, Schneider D. 99m technetium-ethylcysteinate-dimer single-photon emission CT can predict fatal ischemic brain edema. Stroke 1998;29:2556-2562. Berrouschot J, Sterker M, Bettin S, Koster J, Schneider D. Mortality of space-occupying (‘malignant’) middle cerebral artery infarction under conservative intensive care. Intensive Care Med 1998;24:620623. Bosche B, Dohmen C, Graf R. Extracellular concentrations of non-transmitter amino acids in periinfarct tissue of patients predict malignant middle cerebral artery infarction. Stroke 2003;34:29082913. Dohmen C, Bosche B, Graf R. Prediction of malignant course in MCA infarction by PET and microdialysis. Stroke 2003;34:2152-2158. Firlik AD, Yonas H, Kaufmann AM. Relationship between cerebral blood flow and the development of Predictors of life-threatening edema in MCA infarction 14. 15. 16. 17. 18. 19. 20. 21. 22. 23. 24. 25. 26. 27. 28. 29. 30. 31. 32. 33. swelling and life-threatening herniation in acute ischemic stroke. J Neurosurg 1998;89:243-249. Foerch C, Otto B, Singer OC. Serum S100B predicts a malignant course of infarction in patients with acute middle cerebral artery occlusion. Stroke 2004;35:2160-2164. Gerriets T, Stolz E, Konig S. Sonographic monitoring of midline shift in space-occupying stroke: an early outcome predictor. Stroke 2001;32:442-447. Haring HP, Dilitz E, Pallua A. Attenuated corticomedullary contrast: An early cerebral computed tomography sign indicating malignant middle cerebral artery infarction. A case-control study. Stroke 1999;30:1076-1082. Heiss WD, Dohmen C, Sobesky J. Identification of malignant brain edema after hemispheric stroke by PET-imaging and microdialysis. Acta Neurochir Suppl 2003;86:237-240. Kasner SE, Demchuk AM, Berrouschot J. Predictors of fatal brain edema in massive hemispheric ischemic stroke. Stroke 2001;32:2117-2123. Krieger DW, Demchuk AM, Kasner SE, Jauss M, Hantson L. Early clinical and radiological predictors of fatal brain swelling in ischemic stroke. Stroke 1999;30:287-292. Kucinski T, Koch C, Grzyska U, Freitag HJ, Kromer H, Zeumer H. The predictive value of early CT and angiography for fatal hemispheric swelling in acute stroke. AJNR 1998;19:839-846. Lee SJ, Lee KH, Na DG. Multiphasic helical computed tomography predicts subsequent development of severe brain edema in acute ischemic stroke. Arch Neurol 2004;61:505-509. Limburg M, van Royen EA, Hijdra A, de Bruine JF, Verbeeten BW, Jr. Single-photon emission computed tomography and early death in acute ischemic stroke. Stroke 1990;21:1150-1155. Manno EM, Nichols DA, Fulgham JR, Wijdicks EF. Computed tomographic determinants of neurologic deterioration in patients with large middle cerebral artery infarctions. Mayo Clin Proc 2003;78:156160. Maramattom BV, Bahn MM, Wijdicks EF. Which patient fares worse after early deterioration due to swelling from hemispheric stroke? Neurology 2004;63:2142-2145. Mori K, Aoki A, Yamamoto T, Horinaka N, Maeda M. Aggressive decompressive surgery in patients with massive hemispheric embolic cerebral infarction associated with severe brain swelling. Acta Neurochir (Wien) 2002;143:483-491. Oppenheim C, Samson Y, Manai R. Prediction of malignant middle cerebral artery infarction by diffusion-weighted imaging. Stroke 2000;31:2175-2181. Ryoo JW, Na DG, Kim SS. Malignant middle cerebral artery infarction in hyperacute ischemic stroke: evaluation with multiphasic perfusion computed tomography maps. J Comput Assist Tomogr 2004;28:55-62. Schwab S, Spranger M, von-Kummer R, Hacke W. The ‘malignant MCA infarction’: Syndrome or artifact? Aktuelle Neurologie 1996;23:155-162. Stolz E, Gerriets T, Babacan SS, Jauss M, Kraus J, Kaps M. Intracranial venous hemodynamics in patients with midline dislocation due to postischemic brain edema. Stroke 2002;33:479-485. Thomalla GJ, Kucinski T, Schoder V. Prediction of malignant middle cerebral artery infarction by early perfusion- and diffusion-weighted magnetic resonance imaging. Stroke 2003;34:1892-1899. DerSimonian R, Laird N. Meta-analysis in clinical trials. Control Clin Trials 1986;7:177-188. Reith J, Jorgensen HS, Pedersen PM. Body temperature in acute stroke: relation to stroke severity, infarct size, mortality, and outcome. Lancet 1996;347:422-425. Schwab S, Schwarz S, Spranger M, Keller E, Bertram M, Hacke W. Moderate hypothermia in the treatment of patients with severe middle cerebral artery infarction. Stroke 1998;29:2461-2466. 97 Chapter 6 34. 35. 36. 98 Bozzao L, Bastianello S, Fantozzi LM, Angeloni U, Argentino C, Fieschi C. Correlation of angiographic and sequential CT findings in patients with evolving cerebral infarction. AJNR 1989;10:1215-1222. von-Kummer R, Meyding LU, Forsting M. Sensitivity and prognostic value of early CT in occlusion of the middle cerebral artery trunk. AJNR 1994;15:9-15. Bastianello S, Pierallini A, Colonnese C. Hyperdense middle cerebral artery CT sign. Comparison with angiography in the acute phase of ischemic supratentorial infarction. Neuroradiology 1991;33:207211. Algorithm for treatment allocation in small trials Chapter 7 A new algorithm for sequential allocation of two treatments in small clinical trials Jeannette Hofmeijer, Peter C. Anema and Ingeborg van der Tweel Journal of Clinical Epidemiology, under revision 99 Chapter 7 Summary In clinical trials, patients become available for treatment sequentially. Especially in trials with a small number of patients, loss of power may become an important issue, if treatments are not allocated equally or if prognostic factors differ between the treatment groups. In this chapter a new algorithm for sequential allocation of two treatments in small clinical trials is presented, to reduce both selection bias and imbalance. With the algorithm, an element of chance is added to the treatment decision according to minimization. The amount of chance depends on the actual amount of imbalance of treatment allocations of the patients already enrolled. The sensitivity to imbalance may be tuned. Trial simulations were performed with different numbers of patients and prognostic factors, in which loss of power and selection bias were quantified. With this new method, selection bias was smaller than with minimization, and loss of power was lower than with pure randomization or treatment allocation according to a biased coin principle. This new method combines the conflicting aims of reduction of bias by predictability and reduction of loss of power as a result of imbalance. The method may be of use in small trials. 100 Algorithm for treatment allocation in small trials I n clinical trials, patients become available for treatment sequentially. To assure baseline comparability with regard to prognosis, the treatments under study are usually allocated at random. However, in small trials, randomization may result in unbalance of patient numbers or prognostic factors between treatment groups. Thus, in patient allocation there are two important characteristics to optimize.1,2 One is the loss of power, which will increase if treatments are not allocated equally or if prognostic factors differ between the treatment groups.3 The other is the selection bias, caused by the knowledge of the treatment that is to be allocated next.4 Both should be as small as possible. In pure randomization there is no selection bias, since - in case of two treatments allocated equally - each patient has an equal probability of 1/2 of receiving the one or the other. However, this strategy may lead to imbalance of treatments among prognostic factors, especially in small trials. Various randomization procedures have been developed to overcome this problem. One method is random permuted blocks of treatment.5,6 In this design, blocks of patients are formed and a random permutation of treatments is determined for each block. This ensures that equal numbers of patients are achieved for each treatment. To achieve balance with regard to prognostic factors, random permuted blocks within strata have been recommended.6 However, this strategy restricts the number of prognostic factors that can be accounted for in trials with a small amount of patients. To prevent substantial imbalance between treatment groups in trials with a large number of prognostic factors, minimization is considered an alternative approach.6-8 Minimization is exclusively concerned with the balance of treatment allocations over various prognostic factors (or strata), since assignment of a treatment to a new patient is determined by the treatment assignments of previous patients in the relevant stratum. Therefore, at any moment the trial is as balanced as possible. However, minimization involves no random process, except when treatments are equally distributed among prognostic factors and minimization has been discouraged by experts because of predictability.9-11 The Committee for Proprietary Medicinal Products advises that dynamic allocation, such as minimization, should be strongly discouraged10 and the International Conference on Harmonization Guidelines advise that ‘deterministic dynamic allocation procedures should be avoided and an appropriate element of randomization should be incorporated for each treatment allocation’.11 In the biased coin design as described by Efron12 the allocation probability (P) is adapted on the basis of previous treatment allocations. The underrepresented treatment is assigned with P > 1/2 if there are just two treatments and no prognostic factors. This probability is fixed (for example equal to 2/3) and independent of the actual amount of imbalance between groups. We propose an allocation method, Parameterized Dynamic Minimization (PDM), in which the assigned probability is based on previous allocations. Unlike in Efron’s 101 Chapter 7 biased coin algorithm, the probability of allocating a certain treatment is not fixed, but depends on the actual level of imbalance of treatment allocations to the patients already enrolled. Example (first part) In a new trial for secondary prevention after transient ischemic attack or nondisabling stroke, testing treatment C versus treatment E, several prognostic factors for cardiovascular complications might be taken into account. For example, one expects a worse prognosis in patients with hypertension (HT) and in patients older than 65 years of age (OLD). The prognostic factors are categorized into two or more levels, to end up with several strata. In this example the four (2 x 2) strata are: (1) HT yes and OLD yes, (2) HT yes and OLD no, (3) HT no and OLD yes, and (4) HT no and OLD no. If random permuted blocks within strata are used, a separate randomization list is prepared for each of the strata. If a patient enters the clinical trial, one has to identify which stratum the patient is in, and obtain the next random treatment assignment from the corresponding randomization list. The purpose is to reach approximately equal numbers of each treatment modality for each type of patient. It might be necessary to stratify by more than two factors. For instance, in the abovementioned trial also the prognostic factors of high cholesterol (HC) (yes or no) and smoking (SM) (no or past or present) could be taken into account, resulting in 2 x 2 x 2 x 3 = 24 strata each requiring a separate randomization list. In small trials this large number of strata inevitably results in imbalance of prognostic factors between treatment groups. In small trials with a considerable number of prognostic factors to be taken into account, treatment groups with a similar distribution of the prognostic factors may be accomplished by the minimization method, which is ultimately concerned with balancing the marginal treatment totals for each level of each prognostic factor. Table 1 shows the number of patients for each of the treatments C and E according to each of the four prognostic factors after 80 inclusions. Suppose the next patient has hypertension, is older than 65 years of age, has a normal cholesterol level, and does not smoke. Then for each treatment one adds the number of patients in the corresponding rows of the table and assigns the treatment with the smallest sum of marginal totals: • • Treatment C: 30 + 18 + 9 + 19 = 76 Treatment E: 31 + 17 + 8 + 21 = 77 In this example the next patient is assigned treatment C. Especially in small trials the lack of any random process in treatment assignment could be a problem. The PDM method we propose adds an element of chance by assigning 102 Algorithm for treatment allocation in small trials Table 1 Treatment assignments and distribution over four prognostic factors for 80 patients in a fictitious trial on secondary prevention of brain ischemia. Prognostic factor Level Number on treatment C Number on treatment E Next patient HT Yes 30 31 ← No 10 9 Yes 18 17 No 22 23 Yes 31 32 No 9 8 ← ← OLD HC SM Sum of totals No 19 21 Past 8 7 Present 13 12 76 77 ← HT indicates hypertension; OLD, older than 65 years of age; HC, high cholesterol; SM, smoking. If the next patient has hypertension, is older than 65 years, has a normal cholesterol level, and does not smoke, then treatment C is the treatment with the smallest sum of marginal totals. According to the minimization method, this will be the next assigned treatment. the treatment of choice (based on minimization) with P < 1. The assignment probability depends on the amount of imbalance of treatment allocations of the patients already enrolled. For the above-mentioned patient entering the fictitious trial of secondary prevention, treatment C might be assigned with P = 0.6, and treatment E with P = 0.4. If the next patient to enter had no hypertension, was older than 65 years of age, had no high cholesterol, and was a smoker, the sum of allocated patients for each treatment would be: • Treatment C: 10 + 18 + 9 + 13 = 50 • Treatment E: 9 + 17 + 8 + 12 = 46 With the larger imbalance, treatment C might be assigned with P = 0.3 and treatment E with P = 0.7. For the first patient entering a trial and with equal treatment numbers, assignment is random. In the described algorithm, the sensitivity of the method for imbalance may be tuned. 103 Chapter 7 Methods Algorithm The amount of imbalance di between treatment C and E within stratum i (before allocation of the next patient) is defined as di = NCi - NEi (1) where NCi is the number of patients in stratum i who received treatment C, and NEi is the number of patients in stratum i who received treatment E. The weighted average imbalance davg is defined as davg = Σ ( wi di) (2) where wi is the pre-assigned relative importance of imbalance within stratum i and Σ wi = 1. The algorithm for the allocation probability P of a new patient to the most frequently allocated treatment is defined as P = 0.5 α |davg| (3) where α is a chosen constant with which the sensitivity of the method for imbalance may be tuned. Choosing α = 1 will result in pure randomization (P = 0.5), and α = 0 will result in purely deterministic allocation (P = 0) (Figure 1). Since P ≤ 0.5 (Figure 1), the allocation probability of the most frequently allocated treatment is calculated. This implies an allocation probability for the less frequently allocated treatment according to Pless allocated = 1 - P (4) Evaluation of the algorithm We evaluated the algorithm with respect to the two characteristics ‘loss of power’ and ‘selection bias’ as specified by Atkinson.2 Loss of power It is assumed that each of n patients receives one of two treatments C and E (denoted as –1 and +1, respectively). A balance vector b is defined as b = FT . a (5) where F is a matrix in which the constant term and prognostic factors are included, and a is the n x 1 vector of allocations of treatments + 1 and – 1. The superscript T stands for the transpose of a vector or matrix. ‘b’ is equal to zero if all prognostic factors are balanced across all treatments. 104 Algorithm for treatment allocation in small trials Figure 1 Probability as a function of weighted average imbalance for different values of α = P indicates allocation probability; davg indicates weighted average imbalance. The loss of power Ln after the inclusion of n patients is defined as a function of b, F, and a:2 Ln = bT . (FT . F)-1 . b (6) The loss of power in absence of prognostic factors is zero when each treatment has been allocated to an equal number of patients. If prognostic factors are present, the loss of power also depends on the balance over the factors. Selection bias According to Atkinson, the selection bias Bn is quantified as follows: Bn = (Gc – Gi) / n (7) where n is the number of patients included, Gc is the expected number of correct guesses of the next allocated treatment in the n patients and Gi is the expected number of incorrect guesses.2 With two treatments and completely deterministic allocation the treatment is always guessed correctly, and thus Bn = 1. With completely random allocation, the expected number of correct guesses equals the expected number of incorrect guesses, so that Bn = 0. In the proposed PDM, in which the probability of allocation of one of two treatments is different after every single inclusion, selection bias can be defined as follows: Bn = Σ (Pleast allocated – Pmost allocated) / n (8) where the summation is over the patients included (k=1,…,n), with Pmost allocated = 1 – Pleast allocated and thus Bn = Σ (2 Pleast allocated – 1) / n (9) where Pleast allocated is the allocation probability of the kth patient to the least allocated treatment, and Pmost allocated is the allocation probability of the kth patient to the most 105 Chapter 7 allocated treatment. Thus quantified, Bn has the correct value of 1 in completely deterministic and of 0 in completely random allocation. Data analysis Trial simulations were performed with different numbers of patients (20 up to 200) and prognostic factors (3 to 6, each consisting of 2 levels) and different values of α (0 to 1). Plots were made of the separate behavior of selection bias and loss of power, as well as curves of loss of power against selection bias. The plots show averages of 100 simulations. Values for pure randomization (α = 1), minimization (α = 0), and allocation according to Efron’s biased coin algorithm (P = 0.67) are indicated. Note that for the simulations all weights wi were equal. Results Results of the simulations are shown in figures 2 to 4. Figure 2 shows the loss of power. The largest losses occur in small trials with values of α close to one. Loss of power values for α = 1 (random allocation) were even higher, but for the sake of clarity scaling of the y-axis was not adjusted to include these values. With α closer to zero, the effect of trial size becomes smaller. With an increasing number of prognostic factors, values for loss of power become higher (Figure 2a with three prognostic factors versus Figure 2b with six such factors). Figure 3 shows the selection bias. The highest selection bias occurs with values of α approaching 0 (minimization). With an increasing number of prognostic factors, values for selection bias become slightly higher. The effect of trial size is marginal. Figure 4 shows plots of loss of power against selection bias for different values of α and trial sizes of up to n = 200. Allocation starts with random allocation (α=1), with high values of loss of power. Loss of power decreases and selection bias increases, with lower levels of α. Loss of power is higher in smaller trials. Loss of power and selection bias are smaller than in treatment allocation according to Efron’s biased coin. Since both loss of power and selection bias should be as low as possible, optimal values for α are represented in the lower left-hand corner of the plots and depend on the size of the trial and on the number of prognostic factors. Example (second part) Consider the above-mentioned trial on secondary prevention after 80 allocations to either treatment C or E according to Table 1. Suppose again the next patient has hypertension, is older than 65 years, has normal cholesterol and does not smoke. Treatment E is the most allocated treatment (example first part), so allocation of treatment E according to PDM will be as follows. The amount of imbalance is determined as 106 Algorithm for treatment allocation in small trials Figure 2 Loss of power as a function of trial size for trials with 3 (a), or 6 (b) strata. a b Figure 3 Selection bias as a function of trial size for trials with 3 (a), or 6 (b) strata. a b Figure 4 Loss of power as a function of selection bias for trials with 3 (a), or 6 (b) strata. a b 107 Chapter 7 dHT = 31-30, dOLD = 17-18, dHC = 8-9, and dSM = 21-19 (1) davg = Σ ( wi di) = wHT * 1 + wOLD * -1 + wHC * -1 + wSM * 2 (2) If imbalance within all strata is equally weighted, w = 0.25 for all strata, so davg = 0.25-0.25-0.25+0.5 = 0.25 If α = 0.5 and PC = 1- PE PE = 0.5 * 0.50.25 = 0.4 and PC = 0.6 (3) and (4) If a lower level of α is chosen, the sensitivity of the method for imbalance is higher. For example if α = 0.1 and PC = 1- PE PE = 0.5 * 0.10.25 = 0.3 and PC = 0.7 (3) and (4) In the other example treatment C is the most frequently allocated one. Allocation of treatment C will be as follows. First, again, the amount of imbalance is determined. dHT = 10-9, dOLD = 18-17, dHC = 9-8, and dSM = 13-12 (1) davg = Σ ( wi di) = wHT * 1 + wOLD * 1 + wHC * 1 + wSM * 1 = 1 (Again using equal weights for al strata.) (2) If α = 0.5 and PE = 1- PC PC = 0.5 * 0.51 = 0.25 and PE = 0.75 (3) and (4) If α = 0.1 and PE = 1- PC PC = 0.5 * 0.11 = 0.05 and PE = 0.95 (3) and (4) Discussion We developed a new method for assigning one of two treatments in clinical trials, which is aimed to improve the balance across prognostic factors (thus reducing loss of power) as well as to reduce bias by predictability (selection bias). Pure randomization may result in imbalance of treatment numbers across prognostic factors, whereas minimization is ultimately concerned with balance, but results in predictable treatment allocation.10,11 With our new method, an amount of random chance is added to the allocation decision based on minimization. The probability differs for each included patient, depending on the actual level of imbalance. It is not surprising that allocation rules that are more random (α approaching 1) result in lower selection bias and larger values of loss of power as a result of imbalance, whereas values of loss of power are relatively low if allocation becomes more deterministic (α approaching 0). 108 Algorithm for treatment allocation in small trials Minimization based allocation rules including a random element have been described before.13,14 The most important difference with our proposed method is that the random element in our algorithm is not fixed, but depends on the actual level of imbalance within a stratum at the moment of including the next patient. With a smaller imbalance the probability of assigning one of two treatments approaches 50%. This way bias by predictability is kept as small as possible. With a large amount of patients to be included, differences in loss of power become less important. Selection bias is less affected by sample size. However, values for selection bias are slightly higher with a greater number of prognostic factors, because with an increasing number of strata, treatment allocation becomes more deterministic with our method. In the absence of prognostic factors, 50 % of the treatment allocations are random. With the presence of prognostic factors, this percentage becomes smaller. Especially in trials with a small number of patients, loss of power may become an important issue. In choosing a rule for a smaller trial, the plots presented in Figure 4 may be used. Depending on the pre-specified number of patients to include and prognostic factors, α-values in the lower left-hand corner should be chosen, so that both loss of power and selection bias are relatively low. Consider the above-mentioned fictitious secondary prevention trial with three prognostic factors. If the trial aims to include 100 patients, an α value of 0.8 seems reasonable (Figure 4a). For trials with more prognostic factors, 0.4 may be chosen, depending on the number of patients (Figure 4b). In large trials, loss of power becomes less relevant, and the choice of α should probably be based on selection bias. A reasonable value for most situations seems to be 0.7 (Figure 4). In large trials pure randomization may be preferable. In our simulations we used prognostic factors with two levels, but factors with more than two levels can be implemented as well. In addition, the pre-assigned relative importance of imbalance over prognostic factors (wi) was the same for all factors. If balance across one or more specific prognostic factors is more important than balance across other, wi can be adjusted, as long as Σ wi = 1. Our proposed method may be of use in both blinded and unblinded trials, also with centralized treatment allocation. For example, the PDM algorithm has been implemented on a handheld computer and is currently being used in the multi-center Hemicraniectomy After Middle cerebral artery infarction with Life-threatening Edema Trial (HAMLET).15 HAMLET aims to include 112 patients and takes four prognostics factors into account. In conclusion, the PDM allocation method may be of use in relatively small clinical trials with stratification among prognostic factors. This method is preferable to minimization or pure randomization, since it combines the conflicting aims of reduction of selection bias caused by predictability and reduction of loss of power caused by imbalance of treatment numbers among the strata. Moreover, loss of power is smaller than in treatment allocation according to Efron’s biased coin. The sensitivity 109 Chapter 7 to imbalance may be tuned with α. The choice of α depends on the number of patients to be included and the number of pre-specified prognostic factors. References 1. 2. 3. 4. 5. 6. 7. 8. 9. 10. 11. 12. 13. 14. 15. 110 Atkinson AC. Optimum biased-coin designs for sequential treatment allocation with covariate information. Stat Med 1999 30;18:1741-1752. Atkinson AC. The comparison of designs for sequential clinical trials with covariate information. Journal of the Royal Statistical Society, Series A 2002;165:349-373. Atkinson AC. The distribution of loss in two-treatment biased-coin designs. Biostatistics 2003;4:179193. Blackwell D, Hodges JL. Design for the control of selection bias. Annals of mathematical statistics 1957;28:449-460. Matts JP, Lachin JM. Properties of permuted-block randomization in clinical trials. Control Clin Trials 1988;9:327-344. Pocock SJ. Methods of Randomization. Clinical Trials: A Practical Approach. Chichester: John Wiley & Sons, 1983:66-89. Pocock SJ, Simon R. Sequential treatment assignment with balancing for prognostic factors. Biometrics 1975;31:103-115. Taves DR. Minimization: a new method of assigning patients to treatment and. Clin Pharmacol Ther 1974;15:443-453. International Conference on Harmonisation of Technical Requirements for Registration of Pharmaceuticals for Human Use (ICH) adopts Consolidated Guideline on Good Clinical Practice in the Conduct of Clinical Trials on Medicinal Products for Human Use. Int Dig Health Legis 1997;48:231234. Committee for Proprietary Medicinal Products (CPMP): points to consider on adjustment for baseline covariates. Stat Med 2004;15:701-709. ICH Harmonised Tripartite Guideline. Statistical principles for clinical trials. International Conference on Harmonisation E9 Expert Working Group. Stat Med 1999;15:1905-1942. Efron B. Forcing a sequential experiment to be balanced. Biometrika 1971; 58 :403-417. Scott NW, McPherson GC, Ramsay CR, Campbell MK. The method of minimization for allocation to clinical trials. a review. Control Clin Trials 2002;23:662-674. Brown S, Thorpe H, Hawkins K, Brown J. Minimization-reducing predictability for multi-centre trials whilst retaining balance within centre. Stat Med 2005;30:3715-3727. Hofmeijer J, Amelink GJ, Algra A, van Gijn J, Macleod MR, Kappelle LJ, van der Worp HB. Hemicraniectomy After Middle cerebral artery infarction with Life-threatening Edema Trial (HAMLET). Protocol for a randomised controlled trial of decompressive surgery in space-occupying hemispheric infarction. Trials 2006;7:29. HAMLET protocol Chapter 8 Hemicraniectomy After Middle cerebral artery infarction with Life-threatening Edema Trial (HAMLET). Protocol of a randomized controlled trial of decompressive surgery in space-occupying hemispheric infarction Jeannette Hofmeijer, G. Johan Amelink, Ale Algra, Jan van Gijn, Malcolm R. Macleod, L. Jaap Kappelle, and H. Bart van der Worp for the HAMLET investigators Based on Trials 2006;11:7-29 111 Chapter 8 Summary Patients with a hemispheric infarct and massive space-occupying brain edema have a poor prognosis. Despite maximal medical treatment, the case fatality rate may be as high as 80%, and most survivors are left severely disabled. Non-randomized studies have suggested that decompressive surgery reduces mortality substantially and improves functional outcome of survivors. The Hemicraniectomy After Middle cerebral artery infarction with Life-threatening Edema Trial (HAMLET) is designed to compare the efficacy of decompressive surgery to improve functional outcome with that of conservative treatment in patients with supratentorial infarction, who deteriorate as a result of brain edema. HAMLET is a multi-center, randomized clinical trial, which will include 112 patients aged between 18 and 60 years with a large hemispheric infarct with space-occupying edema that leads to a decrease in consciousness. Patients will be randomized to receive either decompressive surgery in combination with medical treatment or best medical treatment alone. Randomization will be stratified for the intended mode of conservative treatment (intensive care or stroke unit care). The primary outcome measure will be functional outcome, as determined by the score on the modified Rankin Scale, at one year. HAMLET started recruiting September 1st 2002 (Current Controlled Trials ISRCTN94237756). As of January 1st 2006, 58 patients had been included. 112 HAMLET protocol L arge cerebral infarcts are commonly associated with variable degrees of brain edema. In severe cases, this may lead to transtentorial or uncal herniation. Serious edema formation usually manifests itself between the second and fifth day after stroke onset.1-4 Brain tissue shifts rather than raised intracranial pressure (ICP) are probably the most likely cause of the initial decrease in consciousness.4,5 Fatal space-occupying brain edema occurs in 1 to 5% of patients with a supratentorial infarct.6,7 However, in younger patients with ischemic stroke, herniation accounts for about half of the deaths in the first month.8 Patients with a hemispheric infarct and massive space-occupying brain edema have a poor prognosis: in recent intensive care (IC)-based prospective series, the case fatality rate was about 80%, despite maximal conservative therapy.9,10 Several conservative treatment strategies have been proposed to limit brain tissue shifts and reduce ICP, such as sedation with barbiturates or propofol, hyperventilation, and osmotic therapy with glycerol, mannitol, or hypertonic saline hydroxyethyl starch (HES).11-13 However, no trials have addressed the efficacy of these therapies to improve clinical outcome14 and several reports suggest that these are ineffective9,10,15 or even detrimental.16,17 The value of ICP monitoring has also not been established. ICP monitoring may reduce iatrogenic errors,18 but is probably not helpful in guiding long-term treatment.5 Despite this lack of evidence hyperventilation, osmotherapy, and the use of ICP monitors are recommended and used by experts for patients with ischemic stroke whose condition is deteriorating secondary to edema formation.11,12,19 Because of the limitations of medical therapies, there have been proposals for decompressive surgery in patients with neurological deterioration due to large hemispheric infarction and edema. The rationale of this therapy is to prevent brain tissue shifts and to normalize intracranial pressure, and thereby to preserve cerebral blood flow and to prevent secondary damage.19 The technique of decompressive surgery is relatively simple and consists of a large hemicraniectomy and a duraplasty. It can be performed in every neurosurgical center. Animal studies have shown that decompressive surgery reduces mortality and improves histological and functional outcome.20,21 Case reports and small retrospective studies have suggested that this intervention lowers mortality without increasing the rate of severely disabled survivors.19,22,23 These findings were supported by two prospective series. Mortality was reduced from about 80% in controls to 34% and 16% in surgically treated patients, respectively, and poor functional outcome from 95% to 50%.24,25 Reports from other studies suggest that decompressive surgery is less effective in elderly patients,26,27 and that substantial recovery extends into the second half year and thereafter.28 Although the above results suggest a very substantial benefit of decompressive surgery, the studies had too many flaws to be readily translated into clinical practice. Most importantly, the groups were not constituted by random selection. The control groups consisted of patients with a significantly higher age, more co-morbidity, 113 Chapter 8 and more frequent lesions in the dominant hemisphere than those in the surgical groups.24,25 In addition, information on functional outcome of the surviving patients was insufficient.24,25 The aim of the current study is to test if decompressive surgery improves functional outcome in patients with neurological deterioration as a result of large hemispheric infarction and edema, as compared with ‘standard’ best medical care, on either a stroke unit (SU) or an ICU, in a randomized controlled clinical trial. Methods and design HAMLET is a multi-center, open, randomized treatment trial with masked outcome assessment. Participating centers have adequate experience with the management of acute ischemic stroke and intensive care treatment of patients with an elevated ICP, and neurosurgical facilities are available on a 24-hours/day basis. The trial protocol has been approved by the medical ethical committees of all participating centers. Enrollment criteria Patients can be enrolled in the study if they have 1. a diagnosis of acute ischemic stroke in the territory of the middle cerebral artery, with an onset within 96 hours prior to the planned start of the trial treatment. Within this timeframe, it is advised to start treatment as soon as possible; 2. a score on the National Institutes of Health Stroke Scale (NIHSS)29 ≥ 16 for right-sided lesions or ≥ 21 for left-sided lesions; 3. a gradual decrease in consciousness to a score of 13 or lower on the Glasgow Coma Scale (GCS) for right-sided lesions, or an eye and motor score of 9 or lower for leftsided lesions; 4. hypodensity on CT involving two thirds or more of the territory of the middle cerebral artery (MCA), and space-occupying edema formation. (displacement of midline structures on CT is not a requirement for inclusion); and 5. age 18 up to and including 60 years. In addition, there must be a possibility to start trial treatment within 3 hours after randomization, and written informed consent must be obtained from a representative of the patient. Patients will be excluded from the study if they have 1. ischemic stroke of the entire cerebral hemisphere (anterior, middle, and posterior cerebral artery territories); 2. a decrease in consciousness at least partially explained by a cause other than edema formation, such as metabolic disturbances or medication; 3. two fixed dilated pupils; 4. been treated with a thrombolytic agent in the 12 hours preceding randomization; 5. a known systemic bleeding disorder; 6. a pre-stroke score on the modified Rankin Scale (mRS)30 of more than 1 or on the Barthel Index (BI)31 of less than 95; 7. a life expectancy < 3 years; or 8. a serious illness that may confound treatment assessment. Informed consent Written informed consent for this study will be obtained from the patient’s authorized 114 HAMLET protocol representative prior to the performance of any protocol-specific procedure. However, several of these assessments or tests may be performed as part of the patient’s routine clinical evaluation (i.e. not specifically performed for this trial). The study will be conducted in accordance with the provisions of the Declaration of Helsinki, as amended in South Africa (1996). Treatment allocation After informed consent is obtained, patients will be randomized to either surgical or conservative treatment. Randomization will be stratified for the intended mode of conservative treatment (intensive care or stroke unit care; see below). The choice of conservative treatment is left at the discretion of the local investigator, and will usually be the standard mode of treatment in the participating center. Allocation to treatment will be made via a telephone call to a 24 hour randomization service. Ideally, each treatment arm will consist of 56 patients. The total number of patients included in the trial will be 112. I. decompressive surgery Decompressive surgery will consist of a large hemicraniectomy and a duraplasty. In summary, a large (reversed) question mark-shaped skin incision based at the ear will be made. A bone flap with a diameter of at least 12 cm (including parts of the frontal, parietal, temporal, and occipital squama) will be removed. Additional temporal bone will be removed so that the floor of the middle cerebral fossa can be explored. The dura will be opened and an augmented dural patch will be inserted. The dura will be fixed at the margins of the craniotomy to prevent epidural bleeding. The temporal muscle and the skin flap will then be re-approximated and secured. Infarcted brain tissue will not be resected. A sensor for registration of intracranial pressure may be inserted. In surviving patients, cranioplasty will be performed after at least six weeks, using the stored bone flap or acrylate. After surgery, patients will be transferred to an IC unit. Anti-edema therapy as described under ‘IIa’ may be used but will usually not be necessary. IIa. intensive care Because no mode of IC treatment has been proven superior to the others, treatment options may vary depending on local traditions. However, to improve consistency of treatment between centers, the following treatment modalities are recommended: a. Osmotherapy. Osmotherapy should be started as soon as possible after randomization. The use of mannitol or glycerol is recommended in a dose sufficient to reach a serum osmolality of 315 to 320 mOsm. b. Intubation and mechanical ventilation. Patients will be intubated if the score on the GCS is lower than or equal to 10, or if there are signs of respiratory insufficiency, 115 Chapter 8 c. d. e. f. g. h. i. such as a pO2 ≤ 60 mm Hg, a pCO2 ≥ 60 mm Hg, or if the airway is compromised. However, earlier intubation is left at discretion of the treating physician. Mechanical ventilation with use of intermittent mandatory or assist/control ventilation will often be sufficient, especially in the early phase. Hyperventilation. The use of hyperventilation is discouraged. If hyperventilation is started, it is advised to monitor venous oxygen saturation with jugular bulb oxymetry and to maintain oxygen saturation higher than 50%. If venous oxygen saturation is not monitored, the pCO2 may be reduced to 28 - 32 mm Hg. ICP monitoring. Invasive monitoring of the intracranial pressure is left at the discretion of the treating physician. If used, the ICP monitor should preferably be inserted ipsilateral to the side of the infarct. Sedation. If mechanical ventilation requires sedation, and in the case of further neurological deterioration, or an uncontrolled increase in ICP, patients may be sedated with propofol. The use of barbiturates is discouraged, because this may reduce cerebral perfusion pressure and does often not lead to a sustained control of ICP. If necessary, muscle relaxants may be used. Blood pressure control. In general, spontaneous elevation of blood pressure to a level of 220/120 mm Hg should be accepted. Sustained higher blood pressures can be treated with labetolol or nitroprusside. In case of sustained hypotension or a critical reduction of cerebral perfusion pressure catecholamines may be used. Elevation of the head (30°) to optimize venous drainage. Aim for normothermia. Antipyretics and a cooling blanket may be used. Aim for normoglycemia. Consider the use of insulin. IC treatment should be continued at least until day 5 after stroke onset, or until there is clinical or radiological improvement deemed to be sufficient to transfer the patient back to the stroke unit or medium care unit. IIb. stroke unit care The patient will be admitted to a stroke unit or medium care unit and is treated according to local practice, supplemented with the treatment options that can be performed in a medium care unit as described above under ‘IIa’. Treatment strategy ‘I’ is the experimental option, strategy ‘IIa’ is recommended by international experts, and strategy ‘IIb’ is the most common clinical practice in the Netherlands, and probably also in a significant number of other European countries. The choice of ancillary care measures, e.g., prophylactic administration of (lowmolecular-weight) heparin and the treatment of complicating illnesses will be at the discretion of the treating physician. However, administration of low-dose aspirin is recommended, whereas treatment with intravenous heparin and hemodilution with plasma expanders are discouraged. 116 HAMLET protocol Outcome assessment The primary outcome measure will be functional outcome, as determined by the score on the mRS, at one year. Outcome will be dichotomized as ‘good’ (mRS 0 to 3) or poor (mRS 4 to dead). The score on the mRS will be determined in a standard way independently by three blinded investigators, on the basis of a narrative written by an unblinded independent study nurse, if necessary followed by a consensus meeting. Secondary analyses will be performed in which ‘good outcome’ will be defined as mRS 0 to 2, and ‘poor outcome’ as mRS 3 to dead, and with case fatality as the measure of outcome. Other outcome measures will include the scores on the NIHSS, the BI, the Montgomery and Asberg Depression Rating Scale (MADRS),32 quality of life as measured with the SF36, and quality of life measured with a visual analogue scale (VAS)33 at one year and at three years. In addition, the mRS, NIHSS, and BI will be determined at 3 and 6 months. Data collection At baseline, the medical history will be assessed and a general and neurological examination will be carried out. The scores on the GCS and NIHSS will be used to assess stroke severity on admission and directly prior to randomization. Retrospective scoring of the NIHSS on admission is reliable when a published algorithm is used.34 Date and time of stroke onset and of neurological deterioration (i.e. GCS ≤ 13) will be noted. Laboratory investigations will include a blood cell count, electrolytes, serum glucose, creatinine, and liver enzymes. Brain CT or MRI scans will be performed on admission and prior to randomization. Lateral displacement of the pineal gland and the septum pellucidum will be measured by the trial co-ordinator. The GCS, pupillary reflexes, body temperature, and blood pressure will be assessed every 4 hours during the first 4 days after randomization. If ICP is recorded, ICP and cerebral perfusion pressure (CPP) will be noted at 2-hour intervals. Dose and time of administration of osmotic agents and sedatives, concurrent medication, and neurological and systemic adverse events will be recorded. The GCS, NIHSS, mRS, and BI will be determined at day 14. CT or MRI of the brain will be performed at day 7 ± 2 days for measurement of infarct volume35,36 and lateral displacement of the pineal gland and septum pellucidum. At three and six months, the mRS, BI, and NIHSS will be assessed. Serious adverse events and the number of days the patient was admitted on an IC unit, in the hospital, and / or rehabilitation center or chronic nursing home will be recorded. At one and three years, the mRS, BI, MADRS, VAS, and SF36 will be assessed by the research fellow or data manager. He / she will perform a standardized evaluation of each patient, including functional status, and will include the obtained information in an essay. Based on this essay, three members of the executive committee, who are blinded to treatment allocation, will determine the scores on the mRS and BI. 117 Chapter 8 Statistical aspects In the primary analysis, outcome of all patients in the surgical group will be compared with that of all patients in the conservative treatment group. However, subgroup analyses will be performed based on the mode of conservative treatment (intensive care vs. stroke unit care), side of the lesion, and time of randomization (within 48 hours versus between 48 and 96 hours). The sample size is based on the desire that in each of the two subgroups the superiority of surgical decompression over conservative treatment can be proved. All primary analyses will be performed on an intention-totreat basis, but additional on-treatment analyses will also be performed. An interim analysis will be performed after the one-year follow-up of the first 30 patients. Power calculations are hampered by the scarcity of data on clinical outcome in this patient group. In the non-randomized trial by Rieke et al., which is most comparable to the proposed study regarding patient group and treatment, 95% (20/21) of the patients in the IC treatment group had a poor outcome, compared with 50% (16/32) in the surgically treated group.24 There are no data on outcome after ‘standard’ treatment, but this is not likely to be better than IC treatment. Based on an α of 0.05 and a β of 0.20, extrapolation of these data suggests that only 12 patients would have to be included in each group to demonstrate the superiority of surgical decompression over conservative treatment in a randomized trial. Assuming that 60% of the patients in the surgery group will have a poor outcome and 85% of the patients in each of the two conservative treatment subgroups, a group size of 28 patients would suffice to prove the superiority of decompressive surgery. Applying the same assumption to the complete treatment, a group size of 56 patients is needed. Safety In the conservative treatment group, patients will receive the standard care of the center to which they have been admitted. For this reason, the components of the conservative treatment strategies will not be considered experimental, and need no detailed explanation in patient information letters. Decompressive surgery consisting of a hemicraniectomy and a duraplasty may be complicated by the occurrence of local postoperative bleeding. In the two open, prospective studies of decompressive surgery for space-occupying hemispheric infarction, including a total of 63 patients, 3 patients (5%; 95% CI 0 to 13) experienced an epidural hematoma, one (2%; 95% CI 0 to 9) a subdural hematoma, and three (5%; 95% CI 0 to 13) a space-occupying subarachnoid hygroma over the trepanation site. None of these complications led to additional neurological deficit.9,25 An adverse event is any unfavorable and unintended sign, symptom, or disease occurring during the follow-up period of the study. Adverse events occurring after randomization will be recorded on the adverse event page of the CRF. A serious 118 HAMLET protocol adverse event is defined as any adverse event that results in death, a life-threatening condition, inpatient hospitalization or prolongation of existing hospitalization, or persistent or significant disability / incapacity. An important medical event that may not result in one of the above conditions may be considered a serious adverse event when, based upon medical judgment, it may jeopardize the patient and may require medical or surgical intervention to prevent one of the outcomes above. A reasonablyrelated adverse event is defined as one that is possibly, probably, or definitely related to the trial treatment. Adverse events that are serious and reasonably related to the trial treatment, and all deaths, require completion of the safety report, which should be faxed to the trial co-ordination center within 5 working days of observation or notification of the event. The Data Monitoring Committee performs analyses of the unblinded interim data and formulates recommendations for the Steering Committee on the continuation of the trial. The Data Monitoring Committee may also offer unsolicited recommendations. Publication of the trial results The trial results will be published by the members of the Executive Committee, on behalf of all HAMLET investigators. Before submission of any manuscript all local principal investigators will have the opportunity to comment on the manuscript. Discussion We present the protocol of a randomized clinical trial designed to test the efficacy of decompressive surgery to improve functional outcome in patients with spaceoccupying hemispheric infarction. Case reports, retrospective studies and two trials have suggested that this intervention lowers mortality without increasing the rate of severely disabled survivors,19,22-25 but this has never been tested in a randomized, controlled clinical trial. The timing of surgery and the choice of enrolment criteria form the crux of the present trial. It has been suggested that decompressive surgery is most efficacious if performed within the first 24 hours after stroke onset and before the occurrence of clinical signs of herniation,25 but this may carry the risk of inclusion of patients who do not need such an invasive procedure for survival and recovery. In addition, in a systematic review of studies on decompressive surgery in space-occupying hemispheric infarction, the timing of surgery did not affect outcome.27 In different studies, several clinical and radiological parameters have been identified as independent early predictors of fatal space-occupying edema formation: nausea and vomiting within 24 hours after stroke onset,37 a systolic blood pressure of 180 mm Hg or more at 12 hours after onset,37 a history of hypertension or heart failure,38 elevated white blood cell count,38 an activity deficit covering the complete MCA 119 Chapter 8 territory on SPECT imaging,39 a hypodensity of more than 50% on the initial CT scan,37,38 attenuated corticomedullary contrast on CT,40 involvement of additional vascular territories,38 and volume of diffusion-weighted imaging (DWI) abnormalities of more than 145 ml.41 In addition, in the Lubeluzole-International-9 trial it was found that the minimum baseline score in patients who ultimately died from brain swelling was 15 for right-sided lesions and 20 for left-sided lesions.37 Unfortunately, all studies were relatively small and the value of the suggested predictors has not been confirmed in other prospective series. With the proposed timing of therapy and choice of enrolment criteria we expect to largely prevent inclusion of patients who cannot benefit from hemicraniectomy, either because they do not need decompressive surgery since their disease will follow a more benign course, or because of extensive and irreversible damage. In the Heidelberg studies, complete global aphasia was an exclusion criterion for decompressive surgery, and the majority of patients receiving surgical therapy had a lesion in the right hemisphere.24,25 In our study, patients with a large infarct in the dominant hemisphere and severe aphasia may be included, as it is unproven that they will fare worse than patients with an infarct in the non-dominant hemisphere. In fact, it has been shown that with the exception of the ability to communicate, the long-term quality of life of patients with left-sided lesions is slightly better than that of patients with a lesion in the right hemisphere.42 In this trial, a fully blinded evaluation of the functional outcome of patients is virtually impossible, as patients in the decompressive surgery group will be easily recognized by surgical marks. Even in case of follow up by telephone, patients are likely to note the sequels of surgery. However, sufficient blinding will be assured by the indirect assessment of scores on the functional outcome scales, as described above. The trial started September 1st 2002. As of January 1st 2006, 58 patients were included in seven participating centers in the Netherlands. Two other Dutch centers and seven centers in the UK are expected to join the trial shortly. Other centers that have adequate experience with the management of acute ischemic stroke and intensive care treatment of patients with an elevated ICP, and that have neurosurgical facilities available on a 24-hours/day basis are welcome to participate. References 1. 2. 3. 120 Shaw C, Alvord Jr E, Berry E. Swelling of the brain following ischemic infarction with arterial occlusion. Arch Neurol 1959;1:161-177. Ropper AH, Shafran B. Brain edema after stroke. Clinical syndrome and intracranial pressure. Arch Neurol 1984;41:26-29. Wardlaw J, Dennis M, Lindley R, Warlow C, Sandercock P, Sellar R. Does early reperfusion of a cerebral infarct influence cerebral infarct swelling in the acute stage or the final clinical outcome? Cerebrovasc Dis 1993;3:86-93. HAMLET protocol 4. 5. 6. 7. 8. 9. 10. 11. 12. 13. 14. 15. 16. 17. 18. 19. 20. 21. 22. 23. 24. Frank JI. Large hemispheric infarction, deterioration, and intracranial pressure. Neurology 1995;45:1286-1290. Schwab S, Aschoff A, Spranger M, Albert F, Hacke W. The value of intracranial pressure monitoring in acute hemispheric stroke. Neurology 1996;47:393-398. Silver FL, Norris JW, Lewis AJ, Hachinski VC. Early mortality following stroke: a prospective review. Stroke 1984;15:492-496. Heinsius T, Bogousslavsky J, Van Melle G. Large infarcts in the middle cerebral artery territory. Etiology and outcome patterns. Neurology 1998;50:341-350. Biller J, Adams-HP J, Bruno A, Love BB, Marsh EE. Mortality in acute cerebral infarction in young adults--a ten-year experience. Angiology 1991;42:224-230. Hacke W, Schwab S, Horn M, Spranger M, De Georgia M, von Kummer R. ‘Malignant’ middle cerebral artery territory infarction: clinical course and prognostic signs. Arch Neurol 1996;53:309-315. Wijdicks EF, Diringer MN. Middle cerebral artery territory infarction and early brain swelling: progression and effect of age on outcome. Mayo Clin Proc 1998;73:829-836. Adams HP, Jr., Adams RJ, Brott T. Guidelines for the early management of patients with ischemic stroke: A scientific statement from the Stroke Council of the American Stroke Association. Stroke 2003;34:1056-1083. Wijdicks EF. Management of massive hemispheric cerebral infarct: is there a ray of hope? Mayo Clin Proc 2000;75:945-952. Schwarz S, Schwab S, Bertram M, Aschoff A, Hacke W. Effects of hypertonic saline hydroxyethyl starch solution and mannitol in patients with increased intracranial pressure after stroke. Stroke 1998;29:15501555. Hofmeijer J, van der Worp HB, Kappelle LJ. Treatment of space-occupying cerebral infarction. Crit Care Med 2003;31:617-625. Schwab S, Spranger M, Schwarz S, Hacke W. Barbiturate coma in severe hemispheric stroke: useful or obsolete? Neurology 1997;48:1608-1613. Muizelaar JP, Marmarou A, Ward JD. Adverse effects of prolonged hyperventilation in patients with severe head injury: a randomized clinical trial. J Neurosurg 1991;75:731-739. Kaufmann AM, Cardoso ER. Aggravation of vasogenic cerebral edema by multiple-dose mannitol. J Neurosurg 1992;77:584-589. Ropper AH. Neurological intensive care. Ann Neurol 1992;32:564-569. Schwab S, Rieke K, Aschoff A, Albert F, von Kummer R, Hacke W. Hemicraniotomy in space-occupying hemispheric infarction: useful early intervention or desparate activism? Cerebrovasc Dis 1996;6:325329. Forsting M, Reith W, Schabitz WR. Decompressive craniectomy for cerebral infarction. An experimental study in rats. Stroke 1995;26:259-264. Doerfler A, Forsting M, Reith W. Decompressive craniectomy in a rat model of “malignant” cerebral hemispheric stroke: experimental support for an aggressive therapeutic approach. J Neurosurg 1996;85:853-859. Carter BS, Ogilvy CS, Candia GJ, Rosas HD, Buonanno F. One-year outcome after decompressive surgery for massive nondominant hemispheric infarction. Neurosurgery 1997;40:1168-1175. Koh MS, Goh KY, Tung MY, Chan C. Is decompressive craniectomy for acute cerebral infarction of any benefit? Surg Neurol 2000;53:225-230. Rieke K, Schwab S, Krieger D. Decompressive surgery in space-occupying hemispheric infarction: 121 Chapter 8 25. 26. 27. 28. 29. 30. 31. 32. 33. 34. 35. 36. 37. 38. 39. 40. 41. 42. 122 results of an open, prospective trial. Crit Care Med 1995;23:1576-1587. Schwab S, Steiner T, Aschoff A. Early hemicraniectomy in patients with complete middle cerebral artery infarction. Stroke 1998;29:1888-1893. Holtkamp M, Buchheim K, Unterberg A. Hemicraniectomy in elderly patients with space occupying media infarction: improved survival but poor functional outcome. J Neurol Neurosurg Psychiatry 2001;70:226-228. Gupta R, Connolly ES, Mayer S, Elkind MS. Hemicraniectomy for massive middle cerebral artery territory infarction: a systematic review. Stroke 2004;35:539-543. Manai R, Srour A, Crozier S, Vandamme X, Samson Y, Cornu P, Rancurel G. Long-term functional outcome of hemicraniectomy in middle cerebral artery malignant infarcts [abstract]. J Neurol 2001;248(suppl 2):121-122. Brott T, Adams HP, Jr., Olinger CP. Measurements of acute cerebral infarction: a clinical examination scale. Stroke 1989;20:864-870. Bamford JM, Sandercock PA, Warlow CP, Slattery J. Interobserver agreement for the assessment of handicap in stroke patients. Stroke 1989;20:828. Mahoney FI, Barthel DW. Functional evaluation: the Barthel Index. Md State Med J 1965;14:61-65. Montgomery S, Asberg M. A new depression scale designed to be sensitive to change. Br J Psychiatry 1979;134:382-389. Indredavik B, Bakke F, Slordahl SA, Rokseth R, Haheim LL. Stroke unit treatment improves long-term quality of life: a randomized controlled trial. Stroke 1998;29:895-899. Williams LS, Yilmaz EY, Lopez-Yunez AM. Retrospective assessment of initial stroke severity with the NIH Stroke Scale. Stroke 2000;31:858-862. Brott T, Marler JR, Olinger CP. Measurements of acute cerebral infarction: lesion size by computed tomography. Stroke 1989;20:871-875. van der Worp HB, Claus SP, Bar PR. Reproducibility of measurements of cerebral infarct volume on CT scans. Stroke 2001;32:424-430. Krieger DW, Demchuk AM, Kasner SE, Jauss M, Hantson L. Early clinical and radiological predictors of fatal brain swelling in ischemic stroke. Stroke 1999;30:287-292. Kasner SE, Demchuk AM, Berrouschot J. Predictors of fatal brain edema in massive hemispheric ischemic stroke. Stroke 2001;32:2117-2123. Berrouschot J, Barthel H, von-Kummer R, Knapp WH, Hesse S, Schneider D. 99m technetium-ethylcysteinate-dimer single-photon emission CT can predict fatal ischemic brain edema. Stroke 1998;29:2556-2562. Haring HP, Dilitz E, Pallua A. Attenuated corticomedullary contrast: An early cerebral computed tomography sign indicating malignant middle cerebral artery infarction. A case-control study. Stroke 1999;30:1076-1082. Oppenheim C, Samson Y, Manai R. Prediction of malignant middle cerebral artery infarction by diffusion-weighted imaging. Stroke 2000;31:2175-2181. de Haan RJ, Limburg M, Van der Meulen JH, Jacobs HM, Aaronson NK. Quality of life after stroke. Impact of stroke type and lesion location. Stroke 1995;26:402-408. Informed consent in HAMLET Chapter 9 Appreciation of the informed consent procedure in a randomized trial of decompressive surgery for space-occupying hemispheric infarction Jeannette Hofmeijer, G. Johan Amelink, Helena M. den Hertog, Ale Algra, L. Jaap Kappelle, and H. Bart van der Worp, on behalf of the HAMLET and the PAIS investigators Journal of Neurology, Neurosurgery, and Psychiatry, under revision 123 Chapter 9 Summary The efficacy of surgical decompression to improve functional outcome in patients with space-occupying hemispheric infarction is currently studied in the randomized Hemicraniectomy After Middle cerebral artery infarction with Life-threatening Edema Trial (HAMLET). As non-randomized studies have suggested that this treatment may reduce mortality, randomization may be considered unethical. In this chapter it is studied how representatives of the patients had experienced the informed consent procedure and what information they could recall in comparison with representatives of patients participating in the randomized trial of Paracetamol (Acetaminophen) In Stroke (PAIS). One year after study inclusion, 60 consecutive representatives who had given informed consent for participation of their relative in HAMLET or PAIS were contacted. Appreciation of the informed consent procedure and recall of trial details were investigated with standardized questionnaires and compared between the two groups. For HAMLET and PAIS 28 and 30 representatives were interviewed, respectively. 86% of HAMLET representatives and 40% of PAIS representatives remembered participation of their relative in a clinical trial (p< 0.001). HAMLET representatives remembered more trial details. With respect to appreciation of the informed consent procedure there were no differences between the groups: in each trial four representatives had considered the question of randomization unacceptable. Recall of information about a clinical trial by representatives of patients with a neurological emergency is better if the potential implications of the treatment under study are greater. Participation of patients in a randomized controlled trial of surgical decompression for space-occupying infarction is generally considered acceptable by their legal representatives. 124 Informed consent in HAMLET I nformed consent from patients or their representatives is essential in any clinical trial. In studies of acute stroke,1 myocardial infarction,2 or other diseases,3-8 the amount of information of the informed consent procedure that study participants could understand and recall has varied. If treatment and thereby study inclusion require urgency because of an acute life-threatening situation, it may be difficult to obtain truly informed consent. The Hemicraniectomy After Middle cerebral artery infarction with Life-threatening Edema Trial (HAMLET) is an ongoing randomized controlled trial to study the effect of decompressive surgery on functional outcome in patients with middle cerebral artery (MCA) infarction and life-threatening edema formation.9 Non-randomized studies have suggested that this intervention may reduce mortality from 78% to 50% or 16%.10,11 The inclusion criteria for participation in HAMLET require a reduced level of consciousness. Consequently, informed consent is asked from patients’ legal representatives. Since patients can deteriorate within hours, there is little time to contemplate participation. The life-threatening condition and time pressure may compromise the ability for relatives to adequately perceive all relevant details of the trial. Moreover, because of the reputed reduction of mortality caused by the treatment under study, randomized treatment allocation may be considered unethical and unacceptable by the patients’ legal representatives. Legal representatives of patients included in HAMLET were interviewed to ascertain what information about the trial they could recall and how they had experienced the informed consent procedure. To study the impact of the life-threatening condition and the invasive experimental treatment, also legal representatives of patients included in a trial in which less vital issues are at stake, the randomized Paracetamol (Acetaminophen) In Stroke (PAIS) trial,12 were interviewed and results were compared. Methods Subjects All legal representatives of patients who had given informed consent for participation in HAMLET at the University Medical Center in Utrecht or the Academic Medical Center in Amsterdam, the Netherlands, between 1 September 2002 and 1 August 2005, and those who had given informed consent for patients included in PAIS between 1 January 2005 and 1 January 2006 at the same hospital in Utrecht and the Erasmus Medical Center in Rotterdam, the Netherlands were contacted. In PAIS, informed consent should be obtained from the patient but may also be given by a legal representative in case of a reduced consciousness or aphasia. We interviewed the first 30 representatives who could be contacted from both trials. The interviews took place approximately 1 year after inclusion. Both trial protocols had been approved by the ethics committees of the participating hospitals. 125 Chapter 9 Trial physicians had sought written informed consent as soon as possible after a reduction in consciousness caused by edema formation (HAMLET) or after admission because of acute stoke (PAIS). The time window for inclusion in HAMLET is 96 hours after stroke onset, whereas this is 12 hours in PAIS. However, in both trials treatment is required to start as soon as possible after neurological deterioration or stroke onset, respectively. The information given about each of the trials included an explanation about the disease and the intervention under study, an explanation of the possible benefits and side effects of the treatment, the controlled design, the voluntary nature of participation, the duration of the trial, and the right to withdraw at any moment. Data collection Legal representatives who had originally given consent were interviewed by telephone by one investigator (JH). The first question was whether the representative could recall having given informed consent for participation of his / her relative in a clinical trial. If not, the interviewer disclosed that the relative had participated in a trial by referring to the meeting with a study physician shortly after clinical deterioration (HAMLET) or admission to the hospital (PAIS). Second, with a standard questionnaire (tables 1 and 2), recall of trial details and appreciation of the informed consent procedure were investigated. Finally, representatives were asked whether they would again give permission for participation in a clinical trial under similar circumstances. Data analysis Answers from representatives of patients participating in HAMLET and PAIS are presented as frequencies (n) and proportions (%). A Chi square test was used for testing differences between the groups. Differences in age were tested by means of Student’s t-test and differences in initial stroke severity as expressed by the score on the National Institutes of Health Stroke Scale (NIHSS)13 with the Mann-Whitney U test. P < 0.05 was considered statistically significant. Results Of 30 representatives of HAMLET patients contacted, 28 were interviewed; one representative refused to participate and one spoke Dutch insufficiently to communicate by telephone. Of 30 representatives of PAIS patients we contacted, all were interviewed. The median time between the informed consent procedure and the interview did not differ between the groups (HAMLET, 13 months (range 10-16); PAIS, 13 months (range 9-17)). As expected, because of the maximum age limit of 60 years in HAMLET, mean age of the patients at study inclusion was significantly lower in HAMLET (48 years ± 8) than in PAIS (69 years ± 13; mean difference 22, 95% CI 126 Informed consent in HAMLET Table 1 Recall of trial details. HAMLET (n (%)) PAIS (n (%)) n = 28 n = 30 P Do you remember your family member having participated in a study? Yes 23 (82) 21 (70) No 5 (18) 9 (30) 0.22 Do you remember the treatment under study? * Correct 24 (86) 12 (40) Incorrect 0 (0) 2 (7) Don’t know 4 (14) 16 (53) 0.001 Do you remember the presumed effects of the treatment under study? * Correct 17 (61) 1 (3) Incorrect 5 (18) 10 (33) Don’t know 6 (21) 19 (64) Correct 20 (71) 0 (0) Incorrect 1 (4) 2 (6) Don’t know 7 (25) 28 (94) Yes 26 (93) 26 (88) No 0 (0) 1 (2) Don’t know 2 (7) 3 (10) Yes 24 (86) 18 (60) No 3 (10) 6 (20) Don’t know 1 (4) 6 (20) Yes 21 (75) 12 (40) No 2 (7) 3 (10) Don’t know 5 (18) 15 (50) <0.001 How was treatment allocation achieved? * <0.001 Was participation voluntary? 0.57 Did you receive written information? 0.068 Did you read the written information? * 0.022 Did you understand the written information? * Yes 18 (64) 11 (37) No 2 (7) 0 (0) Don’t know 8 (29) 19 (63) 0.017 * indicates a statistically significant difference between HAMLET and PAIS. 127 Chapter 9 Table 2 Trial appreciation. HAMLET (n (%)) PAIS (n (%)) n = 28 n = 30 P Was the information provided clear to you? * Yes 27 (96) 14 (47) No 0 (0) 1 (3) Don’t know 1 (4) 15 (50) <0.001 Did you perceive the outcome of treatment allocation as a matter of life or death? * Yes 23 (82) 10 (33) No 4 (14) 16 (54) Don’t know 1 (4) 4 (13) 0.001 Do you have negative feelings about the informed consent procedure? * Yes 9 (32) 1 (3) No 18 (64) 28 (94) Don’t know 1 (4) 1 (3) 0.014 Do you feel that the question for participation and randomization was acceptable at that moment? Yes 19 (68) 16 (54) No 4 (14) 4 (13) Don’t know 5 (18) 10 (33) 0.40 Were you capable of deciding about participation? * Yes 17 (61) 13 (43) No 8 (29) 4 (14) Don’t know 3 (10) 13 (43) 0.018 Did you feel participation was voluntary? Yes 18 (65) 23 (77) No 8 (28) 3 (10) Don’t know 2 (7) 4 (13) 0.18 Do you regret having given permission? Yes 3 (11) 0 (0) No 23 (82) 25 (83) Don’t know 2 (7) 5 (17) 0.12 Are you glad with the treatment your relative received? Yes 14 (50) 19 (63) No 5 (18) 7 (23) Don’t know 9 (32) 4 (13) 128 0.17 Informed consent in HAMLET Table 2 – Continued – HAMLET (n (%)) PAIS (n (%)) n = 28 n = 30 P Would you have preferred the doctor having taken the responsibility for treatment choices? Yes 11 (39) 6 (20) No 15 (54) 17 (57) Don’t know 2 (7) 7 (23) 0.12 Would you have preferred the doctor having taken the responsibility for study participation? Yes 3 (11) 0 (0) No 21 (75) 20 (67) Don’t know 4 (14) 10 (33) 0.063 Would you approve participation again under similar circumstances? Yes 17 (61) 20 (67) No 2 (7) 1 (3) Don’t know 9 (32) 9 (30) 0.78 How do you feel about being interviewed again? Positive 23 (82) 23 (77) Negative 3 (11) 2 (7) Neutral 2 (7) 5 (17) 0.49 * indicates a statistically significant difference between HAMLET and PAIS. 16-27). In both trials, informed consent was most frequently given by the patients’ partner. In PAIS, informed consent was given more frequently by patients’ (grand) children (n = 10) than in HAMLET (n = 3, P = 0.046). Mean stroke severity as assessed with the score on the NIHSS was significantly higher in patients included in HAMLET (median, 23; range, 20-31) than in PAIS (median, 14; range, 3-26; P = 0.02) and in HAMLET the survival rate at follow up (n = 18, 60%) was significantly lower than in PAIS (n = 26, 87%, P = 0.03). The results of the interviews on recall of the details of the trial are summarized in Table 1 and the results of the interviews on appreciation of the informed consent procedure are summarized in Table 2. Relatives of HAMLET participants remembered significantly more trial details. With respect to appreciation of the informed consent procedure, we found fewer differences between the groups. Of the representatives of HAMLET participants, 8 (28%) felt participation to be obligatory, as compared with 3 (10%) of those in PAIS (P = 0.1). Upon inquiry, it appeared that these representatives felt this obligation only with respect to their relatives, because of the reputed reduction in mortality. Of the representatives of HAMLET participants, 9 (32%) had negative feelings about the informed consent procedure, as compared with 1 (3%) of those in PAIS (P = 0.01). Upon inquiry, these negative feelings concerned the acute and emotional situation and not the question of study participation in all but one 129 Chapter 9 representatives. With regard to these negative feelings in representatives of HAMLET patients, there were no differences between representatives of patients who received surgical or best medical treatment (n = 4 (27%) and n = 5 (36%) respectively (P = 0.4)) or between representatives of patients who died or survived (n = 3 (27% of all deaths), and n = 6 (33% of all survivors) respectively (P = 0.6). Discussion Despite the reputed large impact on survival of decompressive surgery, representatives of patients included in HAMLET were equally satisfied with the informed consent procedure as those in a study assessing the effect of acetaminophen on stroke outcome. In both trials, most representatives considered trial participation and randomization acceptable, despite the serious illness of their relatives. The majority would give informed consent again in similar circumstances. HAMLET representatives remembered significantly more trial details. Several factors have been put forward to explain poor recall of study details of patients and representatives participating in clinical trials.1,14 Being admitted with an acute and life-threatening disease has been proposed as the major factor compromising the ability to retain information.1 It has been suggested that knowledge of details of a clinical trial may be better in relatives than in the patients themselves, not only when the relative had given consent, but also when the patient had done so.1 The present study implies that recall of information may be better in life-threatening situations than in other neurological emergencies. The impact of the treatment under study undoubtedly plays an important role. Previous studies have shown low recall rates of complex information, such as randomization15 and other methodological details.16 None of the representatives of PAIS participants could recall the process of random treatment allocation, whereas this was recalled by two thirds of the representatives in HAMLET. Since treatment allocation in HAMLET is not blinded, the concepts of drawing lots and probability are much more explicit and probably easier to retain. HAMLET differs from other acute neurological emergencies by the high case fatality rate of the disease and the presumed large effect on the death rate of the treatment under study. Three quarters of the representatives indeed considered the outcome of randomization a matter of life or death. Still, about two thirds considered participation in a clinical trial and randomization acceptable and only about one third expressed negative feelings with respect to the informed consent procedure or study participation. It has been suggested that positive answers with regard to appreciation of trials may be biased by the hospital connection of the interviewer.1 Still, the results of the present interviews show that even in a study like HAMLET, in which the clinical situation is perceived as life-threatening, asking permission for study inclusion is generally considered acceptable. 130 Informed consent in HAMLET The present study has limitations. First, bias may have been introduced by not interviewing representatives of patients who had refused to participate in either of the trials. However, of the 56 representatives asked for participation of a relative in HAMLET up to 1 August 2006, only three refused, in all cases to avoid survival with severe disability. Second, patients included in HAMLET were younger than those included in PAIS. In PAIS informed consent was more frequently given by the patients’ children. Nevertheless, it is likely that most representatives having given informed consent for HAMLET were younger than those having given consent for PAIS. Age may have influenced understanding and recall of the information. Third, we do not have data on understanding of the trial information at the time of inclusion, so it is impossible to ascertain whether the information was initially understood and then forgotten. We conclude that perception of information about a clinical trial by representatives of patients with a neurological emergency is probably better if the condition is more severe and if the reputed effect of the treatment under study is larger. Participation of patients in a randomized controlled trial of surgical decompression for spaceoccupying infarction is generally considered acceptable by their legal representatives. References 1. 2. 3. 4. 5. 6. 7. 8. 9. Schats R, Brilstra EH, Rinkel GJ, Algra A, van Gijn J. Informed consent in trials for neurological emergencies: the example of subarachnoid haemorrhage. J Neurol Neurosurg Psychiatry 2003;74:988991. Williams BF, French JK, White HD. Informed consent during the clinical emergency of acute myocardial infarction (HERO-2 consent substudy): a prospective observational study. Lancet 2003;361:918-922. Griffin JM, Struve JK, Collins D, Liu A, Nelson DB, Bloomfield HE. Long term clinical trials: How much information do participants retain from the informed consent process? Contemp Clin Trials 2006. Sugarman J, McCrory DC, Hubal RC. Getting meaningful informed consent from older adults: a structured literature review of empirical research. J Am Geriatr Soc 1998;46:517-524. Joffe S, Cook EF, Cleary PD, Clark JW, Weeks JC. Quality of informed consent in cancer clinical trials: a cross-sectional survey. Lancet 2001;358:1772-1777. Cassileth BR, Zupkis RV, Sutton-Smith K, March V. Informed consent - why are its goals imperfectly realized? N Engl J Med 1980;302:896-900. Harrison K, Vlahov D, Jones K, Charron K, Clements ML. Medical eligibility, comprehension of the consent process, and retention of injection drug users recruited for an HIV vaccine trial. J Acquir Immune Defic Syndr Hum Retrovirol 1995;10:386-390. Howard JM, DeMets D. How informed is informed consent? The BHAT experience. Control Clin Trials 1981;2:287-303. Hofmeijer J, Amelink GJ, Algra A, van Gijn J, Kappelle LJ, van der Worp HB. Hemicraniectomy After Middle cerebral artery infarction with Life-threatening Edema Trial (HAMLET). Protocol for a 131 Chapter 9 10. 11. 12. 13. 14. 15. 16. 132 randomised controlled trial of decompressive surgery in space-occupying hemispheric infarction. Trials 2006;7:29. Rieke K, Schwab S, Krieger D. Decompressive surgery in space-occupying hemispheric infarction: results of an open, prospective trial. Crit Care Med 1995;23:1576-1587. Schwab S, Steiner T, Aschoff A. Early hemicraniectomy in patients with complete middle cerebral artery infarction. Stroke 1998;29:1888-1893. van Breda EJ, van der Worp HB, van Gemert HM. PAIS: paracetamol (acetaminophen) in stroke; protocol for a randomized, double blind clinical trial [ISCRTN 74418480]. BMC Cardiovasc Disord 2005;5:24. Brott T, Adams HP, Jr., Olinger CP. Measurements of acute cerebral infarction: a clinical examination scale. Stroke 1989;20:864-870. Oddens BJ, Algra A, van Gijn J. Informing patients about clinical trials. Clin Investig 1993;71:572573. Ross S, Grant A, Counsell C, Gillespie W, Russell I, Prescott R. Barriers to participate in randomised controlled trials: a systematic review. J Clin Epidemiol 1999:52:1143-1156. Miller C, Searight H, Grable D, Schwartz R, Barbarash R. Comprehension and recall of the informational content of the informed consent document: an evaluation of 168 patients in a controlled clinical trial. J Clin Res Drug Dev 1994;8:237-248. Cognitive outcome after decompressive surgery Chapter 10 Long-term cognitive outcome after decompressive surgery for space-occupying hemispheric infarction Jeannette Hofmeijer, H. Bart van der Worp, G. Johan Amelink, L. Jaap Kappelle, Gudrun M.S. Nys, and Martine J.E. van Zandvoort In preperation 133 Chapter 10 Summary In patients with space-occupying hemispheric infarction, decompressive surgery may reduce mortality without increasing the number of severely disabled survivors. However, cognitive functioning in surviving patients has been largely neglected. Ten consecutive patients with a space-occupying hemispheric infarction, who had been included in the surgical arm of the randomized Hemicraniectomy After spaceoccupying Middle cerebral artery (MCA) infarction with Life-threatening Edema Trial (HAMLET) underwent neuropsychological testing approximately one year after decompressive surgery. The tests comprised the following cognitive domains: general cognitive functioning, language, memory, visual perception, neglect, and executive functioning. Testing of general cognitive functioning was severely hampered in all five patients with infarcts in the dominant hemisphere, and in three of the five patients with infarcts in the non-dominant hemisphere cognitive functioning was severely impaired. Testing of language or verbal memory was not feasible or disclosed severe impairments in patients with infarction in the dominant hemisphere. Non-verbal memory was impaired in all patients with infarction in the non-dominant hemisphere and in three of the patients with infarction in the dominant hemisphere. Visual perception was impaired in four patients and below average in two. All patients with infarction in the non-dominant hemisphere suffered profound hemispatial neglect. Executive functioning was below average or impaired in all patients. In general, patients with infarction in the non-dominant hemisphere performed better than patients with infarction in the dominant hemisphere. In addition to the expected severe focal deficits, such as aphasia and neglect, global cognitive decline was found in the majority of patients. Cognitive functioning in patients with infarction in the dominant hemisphere was worse than in patients with infarction in the non-dominant hemisphere. Before implementing decompressive surgery as standard treatment in patients with space-occupying hemispheric infarction, results of cognitive assessments of patients included in ongoing trials should be awaited. 134 Cognitive outcome after decompressive surgery L arge cerebral infarcts may cause space-occupying edema that puts the patient at risk of transtentorial or uncal herniation.1 Fatal space-occupying brain edema occurs in 1-5% of patients with a supratentorial infarct.2,3 The case fatality rate of space-occupying infarcts has been reported to be as high as 78%, despite maximal medical therapy on an intensive care unit.1,4 The aim of decompressive surgery in patients with space-occupying hemispheric infarction is to revert brain tissue shifts and to normalize intracranial pressure, thereby presumably preserving cerebral blood flow and preventing secondary damage.5 Case reports and non-randomized patient series have suggested a substantial reduction in mortality after decompressive surgery without an increase in the number of severely disabled survivors.6-10 For assessment of functional outcome, most investigators used the modified Rankin Scale (mRS) and defined favorable outcome as a score of 4 or lower, representing moderately severe disability at best.11 Reports on cognitive outcome after decompressive surgery are scarce. The only study so far reports on 14 patients subjected to decompressive surgery for space-occupying infarction, however, this study selectively included patients with right-sided infarction and a relatively mild handicap.12 Over the last few years, cognitive functioning has become an important outcome measure in stroke research, as this has shown to be crucial to dependence in daily life,13,14 long-term survival,15 and more subjective measures such as quality of life.16 To study long-term cognitive impairments in unselected patients after decompressive surgery for space-occupying hemispheric infarction, we performed comprehensive neuropsychological testing one year after surgery. Methods Patients Between September 2003 and November 2006, all consecutive patients with a spaceoccupying hemispheric infarct who had been included in the surgical arm of the Hemicraniectomy After space-occupying Middle cerebral artery (MCA) infarction with Life-threatening Edema Trial (HAMLET) underwent neuropsychological testing approximately one year after surgery. Patients were included in HAMLET if the following inclusion criteria had been met:17 (1) age 18 up to and including 60 years, (2) clinical deficits consistent with infarction in the territory of the middle cerebral artery with a score on the National Institutes of Health Stroke Scale (NIHSS) > 15,18 (3) decrease in consciousness to a score of 13 or lower on the Glasgow Coma Scale for patients without aphasia, or an Eye and Motor score of 9 or lower for patients with aphasia, (4) signs on CT or MRI of a unilateral infarct in at least 50% of the territory of the MCA, with or without additional infarction of the territories of the anterior (ACA) or posterior cerebral artery (PCA) on the same side, (5) inclusion within 96 hours 135 Chapter 10 after the onset of symptoms, and (6) written informed consent by the patient or a legal representative. Exclusion criteria were: (1) pre-stroke mRS11 ≥ 2, (2) presence of two fixed dilated pupils, (3) contralateral ischemia or other serious brain lesions, (4) spaceoccupying hemorrhagic transformation of the infarct, defined as ≥ PH2 according to ECASS criteria,19 (5) life expectancy <3 years, (6) other serious illness that may affect outcome, (7) known coagulopathy or systemic bleeding disorder, (8) contra-indication for anesthesia, and (9) pregnancy. Data collection Demographic data were collected at the time of admission. Stroke severity was assessed on admission and before surgical treatment. Infarct side and location were assessed on CT scans made at randomization and follow-up. Neuropsychological examination was performed at the University Medical Center Utrecht approximately one year after surgery. The duration of the test procedure was dependent on the patient’s performance and varied between one and two hours. The neuropsychological examination comprised (1) a standardized battery for general cognitive functioning [Cambridge Cognitive Examination (CAMCOG)]20 and nine tasks tapping the following cognitive domains: (2) language [Boston Naming Task, Token Task (21-item short form), Category Fluency (animals)], (3) memory [Rey Auditory Verbal Learning Task (verbal), Location Learning Task (non-verbal)], (4) visual perception [Judgment of Line Orientation], (5) neglect [Star Cancellation], and (6) executive functioning [Ruff Figural Fluency Test and Letter Fluency (N,A)]. This test battery was developed for screening all major cognitive domains, but without being too extensive, given the expected severe cognitive deficits. Memory and Executive functioning were assessed in both a verbal and a non-verbal way in order to overcome possible feasibility problems with either language (dominant hemisphere dysfunctioning) or non-verbal visuospatial information (non-dominant hemisphere dysfunctioning). Outcome measures and analysis Data are presented in a qualitative way, using the population norms of the neuropsychological tests, corrected for age and level of education.20 Either test-specific cut-off values were used to describe the level of functioning (CAMCOG, Token Task), or the raw scores were converted into percentiles of the general population, to adjudge a level of functioning ranging from impaired (< -2 standard deviations (SD)), below average (≥ -2SD and < -1SD), average (≥ -1SD and ≤ 1SD), high average (> 1SD and ≤ 2SD), or superior (> 2SD). To compare performance of patients with infarcts in the dominant and the non-dominant hemisphere, the number of test scores above the level of ‘impaired’ was calculated. The Mann Whitney U test was used to test differences between the groups. P < 0.05 was considered statistically significant. 136 Cognitive outcome after decompressive surgery Table 1 Baseline data. Patient number Sex Age Infarct side (years) Infarct location NIHSS at randomization mRS 1 year 1 F 49 Left MCA 27 4 2 M 50 Left MCA+ACA 22 4 3 F 59 Left MCA+PCA 26 4 4 M 38 Left MCA+PCA 23 5 5 M 57 Right MCA+ACA 24 3 6 M 57 Right MCA 22 4 7 M 54 Right MCA 22 4 8 F 44 Right MCA+ACA 29 4 9 M 59 Right MCA 26 4 10 F 44 Right MCA 23 4 F indicates female; M, male; MCA, middle cerebral artery; ACA, anterior cerebral artery; PCA, posterior cerebral artery; NIHSS, National Institutes of Health Stroke Scale; mRS, modified Rankin Scale. Results As of December 1st, 2006 twelve patients had been eligible for neuropsychological examination one year after hemicraniectomy. Two patients refused to participate, because of fear to be confronted with cognitive decline. The mean interval ± SD between stroke onset and testing was 14.4 ± 1.6 months. Baseline data are summarized in Table 1. Patient number 9 had an infarct in the right hemisphere with aphasia. This patient was left-handed. One patient had an mRS score of 5, indicating severe disability, eight patients had an mRS score of 4, indicating moderately severe disability and one patient was functionally independent but needed help with some daily activities (mRS score of 3). Hetero-anamnestic information from patients’ relatives disclosed that, in general, patients were cognitively impaired. Relatives often mentioned inability to plan and structure activities interfering with daily life functioning, and problems with impulse regulation. Moreover, the capacity to adapt to new situations was often impaired, leading to frequent frustrations. Test performances are summarized in Table 2. Testing of general cognitive functioning was severely hampered in the four patients with left hemisphere infarction and in one patient with right hemisphere infarction as a result of language impairments. These five patients met the criteria for global aphasia (severe impairment in both comprehension and production of language) prohibiting verbal communication. General cognitive functioning was impaired in three of the five patients with infarcts in the dominant hemisphere. Verbal memory was not testable in patients with infarcts in the dominant 137 Chapter 10 Rey Auditory Verbal Learning Rey Auditory Verbal Learning del Location Learning Task displ Location Learning Task learn Location Learning Task del Judgment of Line Orientation Star Cancellation Figural Fluency Number of tasks above ‘impaired’ na avg avg na imp avg imp 3 na na na imp imp imp avg avg bavg 3 na na na avg bavg avg bavg avg na 5 na na na na na na na na na 0 avg avg nvg imp imp avg avg negl imp 8 imp na bavg navg imp imp imp imp negl imp 2 avg avg bavg navg imp imp havg imp negl imp 7 imp negl imp 6 avg avg imp 4 negl imp 7 1 na na na 2 na imp imp 3 na na imp 4 na na na 5 avg sup sup 6 imp imp 7 avg avg 8 imp avg avg avg bavg nvg imp imp avg 9 na imp imp na na na imp bavg bavg 10 imp avg avg imp avg navg bavg imp havg bavg Patient number Token Task na Boston Naming Task na CAMCOG Category Fluency Table 2 Neuropsychological test performance. na indicates not assessable; del, delayed recall; displ, displacement score; learn, learning index; imp, impaired; bavg, below average; avg, average; havg, high average; sup, superior; negl, neglect. Figure Cognitive functioning defined as the number of test-scores above the level of impaired for patients with infarcts in the dominant and the non-dominant hemisphere. Box plots indicate Medians, Quartiles, and Ranges. 138 Cognitive outcome after decompressive surgery hemisphere, and below average in four of the five patients with infarcts in the nondominant hemisphere. Non-verbal memory was impaired in all patients with infarcts in the non-dominant hemisphere and in three of the patients with infarcts in the dominant hemisphere. Visual perception was impaired in four patients and below average in two. All patients with infarcts in the non-dominant hemisphere suffered severe hemispatial neglect. Executive functioning was below average or impaired in all patients. In general, patients with infarcts in the non-dominant hemisphere tended to perform better than patients with infarcts in the dominant hemisphere (Figure), but this difference did not reach statistical significance (P = 0.08). Discussion This is the first report of neuropsychological examination in unselected patients after decompressive surgery for space-occupying MCA infarction. Cognitive functioning was poor in the majority. In addition to the expected severe focal deficits, such as aphasia and neglect, executive functioning and memory were affected in all patients resulting in below average or impaired performance. Several studies have shown a decline in cognitive functioning after all types of stroke.21,22 Higher cognitive functions are not solely located in a specific area of the brain, but are the result of complex and dynamic neural networks which are scattered throughout the brain.23 For this reason, neuropsychological functioning is highly dependent on the overall integrity of the brain, and may be impaired even after lacunar or brainstem infarction,24,25 let alone after large cortical infarcts. Irrespective of the side of infarction, most patients had a score on the mRS of 4. Nevertheless, patients with infarction in the dominant hemisphere performed worse than patients with infarcts in the non-dominant hemisphere, having impairments on both verbal and non-verbal tasks. Similar findings have been reported earlier in a more general stroke population.22 Language probably has a far-reaching impact on cognitive functioning and should be considered as the means by which most cognitive functions are articulated.21 Executive functioning, as tested with the Ruff Figural Fluency Test, was below average or impaired in all patients. This cannot be completely attributed to the presence of aphasia, amnesia, or a reduction in motor speed. The impaired performance reflects an inflexibility in information processing and an impairment in generation of ideas. In daily life this means that patients may become locked into one strategy and do not possess the ability to change to another. Relatives of patients with lesions in the non-dominant as well as relatives of patients with lesions in the dominant hemisphere have confirmed this observation. According to their relatives, the patients’ inability to initiate, plan and structure activities interferes with daily life functioning. Moreover, the capacity to adapt to new situations appears to be impaired to such an amount that 139 Chapter 10 this leads to frequent frustrations. Next to executive functioning, both verbal and non-verbal memory were impaired in most patients. Memory and the ability to learn probably deteriorate even more by disturbances of attention and concentration. In a previous study of cognitive functioning in patients with large MCA infarction subjected to decompressive surgery, profound disturbances of attention were found. In this study only patients with rightsided infarction and with a relatively preserved level of functioning were included.12 We performed neuropsychological examination more than one year after the onset of symptoms. Although further cognitive recovery after the first year following stroke has been described,26 a lack of improvement or even cognitive decline in the second year after stroke onset has also been reported.27 Patients with aphasia not only tend to perform worse, but also show less improvement.27,28 Thus, in our patients further improvement is probably very limited. The current study has limitations. First, the small sample size precludes definitive conclusions on the effects of cognitive and functional outcome. Despite the overall low performance, two patients (patient 5 and 7) performed average or above average on most tasks. Neuropsychological evaluation of all patients participating in randomized trials of decompressive surgery in space-occupying infarction is clearly warranted. Secondly, because of the small sample size, data were mainly presented in a descriptive way. Nevertheless, the serious impact of life-threatening space-occupying infarction on cognition is evident. Thirdly, in patients with infarcts in the dominant hemisphere language was too poor to be quantified by means of any of the tasks included in the test battery. In general, neuropsychological examination is feasible in the majority of patients with aphasia caused by stroke.14 Although floor effects prevented quantification in our series, these may still be seen as an indication of the magnitude of the damage in these patients. Finally, the present study did not include a control group of patients with space-occupying MCA infarcts that did not undergo surgery. For this reason, we do not know the impact of decompressive surgery on cognition in this patient population. In conclusion, although decompressive surgery increased the probability of a favorable outcome in patients with space-occupying MCA infarction in a pooled analysis of ongoing trials (chapter 11), the present results suggest that survivors are likely to be left with significant cognitive impairments. Despite similar functional dependence as assessed with the mRS, cognitive functioning in patients with infarction in the dominant hemisphere was worse than in patients with infarction in the non-dominant hemisphere. Before implementing decompressive surgery as a standard treatment modality in patients with MCA infarction, who deteriorate as a result of space-occupying edema, results of cognitive assessments of patients included in ongoing trials should be awaited. Moreover, the possible cognitive consequences, especially for patients with a stroke in the dominant hemisphere, should be considered before a decision to perform surgery is taken. 140 Cognitive outcome after decompressive surgery References 1. 2. 3. 4. 5. 6. 7. 8. 9. 10. 11. 12. 13. 14. 15. 16. 17. 18. Hacke W, Schwab S, Horn M, Spranger M, De Georgia M, von Kummer R. ‘Malignant’ middle cerebral artery territory infarction: clinical course and prognostic signs. Arch Neurol 1996;53:309-315. Silver FL, Norris JW, Lewis AJ, Hachinski VC. Early mortality following stroke: a prospective review. Stroke 1984;15:492-496. Heinsius T, Bogousslavsky J, Van Melle G. Large infarcts in the middle cerebral artery territory. Etiology and outcome patterns. Neurology 1998;50:341-350. Wijdicks EF, Diringer MN. Middle cerebral artery territory infarction and early brain swelling: progression and effect of age on outcome. Mayo Clin Proc 1998;73:829-836. Schwab S, Rieke K, Aschoff A, Albert F, von Kummer R, Hacke W. Hemicraniotomy in space-occupying hemispheric infarction: useful early intervention or desparate activism? Cerebrovasc Dis 1996;6:325329. Carter BS, Ogilvy CS, Candia GJ, Rosas HD, Buonanno F. One-year outcome after decompressive surgery for massive nondominant hemispheric infarction. Neurosurgery 1997;40:1168-1175. Delashaw JB, Broaddus WC, Kassell NF. Treatment of right hemispheric cerebral infarction by hemicraniectomy. Stroke 1990;21:874-881. van Leusen HJ, Tans JT, Wurzer JA. [Hemicraniectomy for treatment of malignant medial cerebral artery infarction in 3 patients]. Ned Tijdschr Geneeskd 2001;145:639-643. Schwab S, Steiner T, Aschoff A. Early hemicraniectomy in patients with complete middle cerebral artery infarction. Stroke 1998;29:1888-1893. Rieke K, Schwab S, Krieger D. Decompressive surgery in space-occupying hemispheric infarction: results of an open, prospective trial. Crit Care Med 1995;23:1576-1587. van Swieten JC, Koudstaal PJ, Visser MC, Schouten HJ, van Gijn J. Interobserver agreement for the assessment of handicap in stroke patients. Stroke 1988;19:604-607. Leonhardt G, Wilhelm H, Doerfler A. Clinical outcome and neuropsychological deficits after right decompressive hemicraniectomy in MCA infarction. J Neurol 2002;249:1433-1440. Tatemichi TK, Desmond DW, Stern Y, Paik M, Sano M, Bagiella E. Cognitive impairment after stroke: frequency, patterns, and relationship to functional abilities. J Neurol Neurosurg Psychiatry 1994;57:202207. Nys GM, Van Zandvoort MJ, De Kort PL. The prognostic value of domain-specific cognitive abilities in acute first-ever stroke. Neurology 2005;64:821-827. Tatemichi TK, Paik M, Bagiella E, Desmond DW, Pirro M, Hanzawa LK. Dementia after stroke is a predictor of long-term survival. Stroke 1994;25:1915-1919. Nys GM, Van Zandvoort MJ, van der Worp HB, de Haan EH, de Kort PL, Jansen BP, Kappelle LJ. Early cognitive impairment predicts long-term depressive symptoms and quality of life after stroke. J Neurol Sci 2006;247:149-156. Hofmeijer J, Amelink GJ, Algra A, van Gijn J, Macleod MR, Kappelle LJ, van der Worp HB. Hemicraniectomy After Middle cerebral artery infarction with Life-threatening Edema Trial (HAMLET). Protocol for a randomised controlled trial of decompressive surgery in space-occupying hemispheric infarction. Trials 2006 11;7:29. Goldstein LB, Bertels C, Davis JN. Interrater reliability of the NIH stroke scale. Arch Neurol 1989;46:660-662. 141 Chapter 10 19. 20. 21. 22. 23. 24. 25. 26. 27. 28. 142 Larrue V, von Kummer RR, Muller A, Bluhmki E. Risk factors for severe hemorrhagic transformation in ischemic stroke patients treated with recombinant tissue plasminogen activator: a secondary analysis of the European-Australasian Acute Stroke Study (ECASS II). Stroke 2001;32:438-441. Lezak MD, Howieson DB, Loring DW. Neuropsychological assessment, 4 ed. New York: Oxford University Press, 2004. Luria A. Higher cortical functions in man. New York: Basic Books, 1966. Hochstenbach J, Mulder T, van LJ, Donders R, Schoonderwaldt H. Cognitive decline following stroke: a comprehensive study of cognitive decline following stroke. J Clin Exp Neuropsychol 1998;20:503517. Hom J, Reitan RM. Generalized cognitive function after stroke. J Clin Exp Neuropsychol 1990;12:644654. van Zandvoort M, de Haan EH, van Gijn J, Kappelle LJ. Cognitive functioning in patients with a small infarct in the brainstem. J Int Neuropsychol Soc 2003;9:490-494. Van Zandvoort MJ, De Haan EH, Kappelle LJ. Chronic cognitive disturbances after a single supratentorial lacunar infarct. Neuropsychiatry Neuropsychol Behav Neurol 2001;14:98-102. del Ser T, Barba R, Morin MM. Evolution of cognitive impairment after stroke and risk factors for delayed progression. Stroke 2005;36:2670-2675. Hochstenbach JB, den Otter R, Mulder TW. Cognitive recovery after stroke: a 2-year follow-up. Arch Phys Med Rehabil 2003;84:1499-1504. Nys GM, Van Zandvoort MJ, De Kort PL, Jansen BP, van der Worp HB, Kappelle LJ, de Haan EH. Domain-specific cognitive recovery after first-ever stroke: a follow-up study of 111 cases. J Int Neuropsychol Soc 2005 Nov;11:795-806. Decompressive surgery in MCA infarction: pooled analysis of 3 trials Chapter 11 Early decompressive surgery in spaceoccupying hemispheric infarction: a pooled analysis of three randomized controlled trials Katayoun Vahedi,* Jeannette Hofmeijer,* Eric Juettler,* Eric Vicaut, Bernard George, Ale Algra, G. Johan Amelink, Peter Schmiedeck, Stefan Schwab, Peter M. Rothwell, Marie-Germaine Bousser, H. Bart van der Worp, and Werner Hacke for the DECIMAL, DESTINY, and HAMLET investigators *These authors contributed equally Based on Lancet Neurology 2007; 6:215-222 143 Chapter 11 Summary Space-occupying middle cerebral artery (MCA) infarction has a mortality rate of about 80%. Non-randomized studies have suggested that decompressive surgery reduces mortality without increasing the number of severely disabled survivors. To obtain sufficient data to reliably estimate the effects of decompressive surgery as soon as possible, data from three European randomized controlled trials (DECIMAL, DESTINY, and HAMLET) were pooled. The trials were ongoing when the pooled analysis was planned. Individual data of patients between 18 and 60 years of age with space-occupying MCA infarction included in one of the three trials and treated within 48 hours after stroke onset were pooled. The protocol was designed prospectively when the trials were still recruiting and outcomes were defined blind to the results of the individual trials. The primary outcome measure was the score on the modified Rankin Scale (mRS) at one year dichotomized between favorable (0-4) and unfavorable (5 and death). Secondary outcome measures included a dichotomization of the mRS between 0-3 and 4 to death and case fatality rate at one year. Data analysis was performed by an independent data monitoring committee. Absolute risk reductions (ARR), odds ratios, and 95% confidence intervals (95% CI) were calculated for the specified outcomes in each trial and then pooled by the Mantel-Haenszel method. P < 0.05 was considered statistically significant. Ninety-three patients were included. After decompressive surgery, more patients had an mRS≤4 (75% vs 24%; pooled ARR [95%CI]: 51%[34-69]), an mRS≤3 (43% vs 21%; ARR: 23%[5-41]), and survived (78% vs 29%; ARR: 50%[33-67]) indicating numbers needed to treat of 2 for survival with an mRS≤4, 4 for survival with an mRS≤3, and 2 for survival irrespective of outcome. The effect of surgery was highly consistent across the three trials. In patients with space-occupying MCA infarction, decompressive surgery reduces case fatality by 50% with an mRS≤3 at one year in 55% of the survivors. The decision to perform decompressive surgery should be made on an individual basis in every patient. 144 Decompressive surgery in MCA infarction: pooled analysis of 3 trials L ife-threatening space-occupying brain edema occurs in 1 to 10% of patients with a supratentorial infarct, and usually manifests itself between the second and the fifth day after stroke onset.1-3 However, up to one third of the patients may have neurological deterioration within 24 hrs of symptom onset.4 The prognosis of these space-occupying or ‘malignant’ MCA infarcts is poor, with case fatality rates in intensive care-based series of nearly 80%.5,6 No medical therapy has been proven effective.7 Different predictors of fatal brain edema formation have been identified, such as major early CT hypodensity involving >50% of the MCA territory and involvement of additional vascular territories besides that of the MCA.8 However, to date no single prognostic factor with sufficient prognostic value has been identified. Prospective, non-randomized studies have suggested that decompressive surgery, consisting of a hemicraniectomy and duraplasty, reduces mortality in patients with space-occupying MCA infarction without increasing the number of severely disabled survivors.9-12 However, evidence from randomized trials is lacking. While most clinicians agree that the procedure is probably life-saving, no convincing data are available regarding functional outcome of survivors. The effect of decompressive surgery on functional outcome in patients with space-occupying MCA infarction has been studied in three European randomized controlled trials: the French DECIMAL [DEcompressive Craniectomy In MALignant middle cerebral artery infarcts], the German DESTINY [DEcompressive Surgery for the Treatment of malignant INfarction of the middle cerebral arterY], and the Dutch HAMLET [Hemicraniectomy After Middle cerebral artery infarction with Lifethreatening Edema Trial].13 Two of these trials interrupted recruitment early in 2006: DECIMAL because of slow recruitment and a statistically significant difference in mortality between the treatment groups favoring surgery, and DESTINY because a pre-defined sequential analysis showed a statistically significant benefit of surgery on mortality. HAMLET is ongoing. The three trials have a similar design and share the same primary outcome measure, i.e. favorable versus unfavorable functional outcome as determined by the score on the modified Rankin Scale (mRS).14 A collaborative protocol for a pooled analysis of individual patient data from the three trials was planned before the interruption of the first two trials. The principal aim of this pooled analysis was to obtain sufficient data to reliably estimate the effects of decompressive surgery as soon as possible so as to avoid unnecessary (and unethical) continuation of randomization in the individual trials. Methods Design We combined individual patient data from DECIMAL (NCT00190203), DESTINY (ISRCTN01258591), and HAMLET (ISRCTN94237756), which are multi-center, 145 Chapter 11 randomized, controlled clinical trials assessing the effect of decompressive surgery in patients with space-occupying MCA infarction. When the pooled analysis was planned, the trials were still ongoing and there was no knowledge on outcome data except for mortality rates in DECIMAL and DESTINY. At the time of the analysis, DECIMAL and DESTINY had been interrupted, while HAMLET was still ongoing. Randomization, treatment, and outcome assessment were performed according to the individual study protocols that were approved by the relevant institutional review boards. Informed consent was obtained from the patients or the patients’ legal representatives. Trial characteristics DECIMAL was designed to include a maximum of 60 patients, 30 in each group. Recruitment stopped after inclusion of 38 patients in March 2006 because of slow enrollment, a significant difference in mortality favoring decompressive surgery, and the opportunity of a pooled analysis with DESTINY and HAMLET. DESTINY aimed to include a maximum of 68 patients. Recruitment was interrupted in February 2006 after a planned interim analysis including 32 patients showed a statistically significant benefit of surgery on 30-day mortality, and was stopped definitively after a revised sample size projection indicated that 188 patients would be required to show a statistically significant difference with regard to the primary endpoint (mRS 0 to 3 versus 4 to death at six months). HAMLET aims to include 112 patients.13 Eligibility criteria for the pooled analysis The inclusion and exclusion criteria of the three trials were largely similar. The main differences included: a longer interval from stroke onset to treatment start allowed in HAMLET (96 h) than in DECIMAL (30 h) and DESTINY (36 h). For the pooled analysis, a maximum time window from stroke onset to randomization of 45h (i.e. 48h to treatment) was adopted. Neuro-imaging criteria differed between the three trials and could therefore not be included in the pooled analysis. These criteria were a DWI infarct volume more than 145 cm3 in DECIMAL, brain CT ischemic changes involving more than 2/3 of the MCA territory and including the basal ganglia in DESTINY and brain CT ischemic changes involving at least 2/3 of the MCA territory with spaceoccupying edema in HAMLET. Patients included before 1 November 2005 in any of the three trials and fulfilling the prospectively defined eligibility criteria listed in Table 1 were included in this pooled analysis. Randomization and treatment Patients were randomized to either decompressive surgery or conservative treatment in all trials. In DECIMAL, patients were centrally randomized in blocks of four using a pre-established randomization list. In DESTINY, randomization was done according to a central computer generated randomization list for each participating center. 146 Decompressive surgery in MCA infarction: pooled analysis of 3 trials Table1 Eligibility criteria for the pooled analysis. Inclusion criteria Age 18 - 60 years. Clinical deficits suggestive of infarction in the territory of the middle cerebral artery (MCA) with a score on the National Institutes of Health Stroke Scale (NIHSS) > 15. Decrease in the level of consciousness to a score of 1 or greater on item 1a of the NIHSS. Signs on CT of an infarct of at least 50% of the MCA territory, with or without additional infarction in the territory of the anterior (ACA) or posterior cerebral artery (PCA) on the same side, or DWI infarct volume > 145 cm3. Inclusion within 45 hours after onset of symptoms. Written informed consent by the patient or a legal representative. Exclusion criteria Pre-stroke score on the mRS ≥ 2. Two fixed dilated pupils. Contralateral ischemia or other brain lesion that may affect outcome. Space-occupying hemorrhagic transformation of the infarct. Life expectancy < 3 years. Other serious illness that may affect outcome. Known coagulopathy or systemic bleeding disorder. Contra-indication for anesthesia. Pregnancy. In HAMLET, randomization was performed centrally, with a computerized algorithm in which an element of chance was added to the treatment decision of minimization (chapter 7). Decompressive surgery consisted of creating a large bone flap and duraplasty. In summary, a large (reversed) question mark-shaped skin incision based at the ear was made. A bone flap with a diameter of at least 12 cm (always including the frontal, temporal, and parietal bones) was removed. Additional temporal bone was removed so that the floor of the middle cerebral fossa could be reached. The dura was opened and a dural patch, consisting of pericranium or a commercially available dura substitute, was inserted and secured to enlarge the intradural space. To prevent epidural bleeding, dural tacking sutures were used when considered necessary. The temporal muscle and the skin flap were then re-approximated and sutured. Infarcted brain tissue was not resected. In surviving patients, cranioplasty was performed after at least six weeks, using the stored bone flap or acrylate. After surgery, patients were transferred to an intensive care unit, but anti-edema therapy was usually not necessary.15 In the 147 Chapter 11 conservative group, patients received best medical treatment, based on published guidelines for the management of acute ischemic stroke and space-occupying brain edema .16-18 Data collection A broad range of baseline characteristics and outcome measures was collected in the individual trials. For the pooled analysis the following pre-specified baseline characteristics were used: age, sex, time between stroke onset and randomization, medical history, physical examination (blood pressure, body temperature), presence of aphasia, and score on the National Institutes of Health Stroke Scale (NIHSS) at randomization.19 Outcome measures The trials used largely similar outcome measures. In the pooled analysis, the primary outcome measure was the score on the mRS at one year dichotomized between ‘favorable’ (mRS 0 to 4) and ’unfavorable’ (mRS 5 and death). Secondary analyses included a dichotomization of the mRS in which ‘favorable’ was defined as a score of 0 to 3 and ‘unfavorable’ as a score of 4 to death, and case fatality at one year. The mRS measures functional outcome after stroke.14 The scores range from 0 to 6, 0 indicating no symptoms at all, 1 indicating no significant disability despite symptoms, being able to carry out all usual duties and activities, 2 indicating slight disability, being unable to carry out all previous activities, but able to look after own affairs without assistance, 3 indicating moderate disability, requiring some help, but being able to walk without assistance, 4 indicating moderately severe disability, being unable to walk without assistance and unable to attend to own bodily needs without assistance, 5 indicating severe disability, being bedridden, incontinent and requiring constant nursing care and attention, and 6 indicating death. In DECIMAL outcomes were assessed by a neurologist blinded to treatment allocation, in DESTINY outcome was assessed unblinded, and in HAMLET the score on the mRS was determined independently by three blinded investigators, based on a narrative written by an unblinded independent study nurse, if necessary followed by a consensus meeting. Statistical analyses Data analysis was performed according to a pre-specified protocol, by an independent Data Monitoring Committee. The distributions of the mRS, with death scored as 6, were compared between the treatment groups with the Mann-Whitney U test. To assess the effect of surgical treatment, absolute risk reductions (ARR), odds ratios (OR), and 95% confidence intervals (95% CI) were calculated for the specified outcomes in each trial and then pooled by the Mantel-Haenszel method. Heterogeneity of ARR and OR between trials was determined by the Breslow-Day test. The influence of baseline 148 Decompressive surgery in MCA infarction: pooled analysis of 3 trials differences between the treatment groups was assessed by the comparison of crude and adjusted ORs. Data were also analyzed in a cumulative logit model.20 These ORs represent the odds of obtaining higher rather than lower mRS scores after surgical treatment compared with conservative treatment. Subgroup analyses were performed according to age (dichotomized at 50 years), timing of randomization (dichotomized at 24 hours), and presence of aphasia. Analyses were performed on an intention-totreat basis. The SPSS software package was used for all analyses and P < 0.05 was considered statistically significant. Results All patients randomized in DECIMAL (38 patients) and DESTINY (32 patients) and 28 patients randomized in HAMLET were eligible for the pooled analysis. From HAMLET, 29 of a total of 57 patients were excluded because they were randomized after 45 hours from stroke onset or were included after November 1st 2005. For all other patients, there were no missing data on primary or secondary outcome measures. Thus, 93 patients were included, of whom 51 were randomized to decompressive surgery and 42 to conservative treatment. There was one cross-over in DESTINY from conservative treatment to decompressive surgery. There were no cross-overs in the other trials. Of two patients from DESTINY the primary outcome measure was assessed at ten months. Baseline characteristics Baseline characteristics are shown in Table 2. Treatment groups within the individual trials were broadly similar. There were two minor differences: in DESTINY, the conservatively treated group had a higher NIHSS score than the surgically treated group, and in DECIMAL, mean systolic blood pressure was higher in the conservatively treated group than in the surgically treated group. There were baseline differences between the three trials: Time to randomization in DESTINY and HAMLET was significantly longer than in DECIMAL. Time to randomization in HAMLET was also longer than in DESTINY. NIHSS scores in DESTINY and DECIMAL were lower than in HAMLET. Body temperature was lower in DESTINY and DECIMAL than in HAMLET. History of TIA or stroke was more frequent in HAMLET than in DESTINY, and a history of ischemic heart disease was more common in DESTINY than in DECIMAL. Outcome Figure 1 shows the distributions of the scores on the mRS after 12 months according to randomized treatment. The difference in distributions of the scores on the mRS between the two treatment groups was highly significant (p < 0.001; Figure 1). 149 150 DECIMAL 9 (45%) 2/19 (11%) 0/19 (0%) 0/19 (0%) 6/19 (32%) 0/19 (0%) 9/18 (50%) 36.9 (0.5) 135.5 (19.3) 75.8 (15.6) 12 (60%) 21.5 (18-25) History of TIA/stroke Ischemic heart disease Atrial fibrillation Hypertension Diabetes Current smoker Temperature (mean [SD]) Systolic BP (mean [SD]) Diastolic BP (mean [SD]) Aphasia NIHSS (median [IQR]) Hours to randomization (median [IQR]) 16 (11-21) 43.4 (9.7) Male sex 16 (11-20) 21.5 (18-26) 11 (61%) 83.4 (15.1) 154.0 (25.1) 37.0 (0.6) 4/16 (25%) 4 (22%) 5 (28%) 1 (6%) 1 (6%) 0 (0%) 9 (50%) 43.4 (7.3) Surgery (n=20) Conservative (n=18) Age (mean [SD]) Table 2 Baseline characteristics. 16 (11-21) 21.5 (18-25) 23 (61%) 79.4 (15.6) 144.2 (23.8) 37.0 (0.6) 13/34 (38%) 5/37 (14%) 11/37 (30%) 1/37 (3%) 1/37 (3%) 2/37 (5%) 18 (47%) 43.4 (8.5) Total (n=38) 24 (18-29) 21 (19.5-23) 10 (59%) 74.7 (15.6) 139.4 (16.6) 37.0 (0.5) 7 (41%) 2 (12%) 9 (53%) 3 (18%) 3 (18%) 0 (0%) 8 (47%) 43.2 (9.7) 23 (17-33) 24 (22-26) 11 (73%) 72.3 (11.2) 133.3 (14.2) 37.2 (0.6) 5 (33%) 3 (20%) 7 (47%) 3 (20%) 4 (27%) 0 (0%) 7 (47%) 46.1 (8.4) 24 (18-29) 22 (20.3-24) 21 (66%) 73.6 (13.5) 136.6 (15.6) 37.1 (0.5) 12 (38%) 5 (16%) 16 (50%) 6 (19%) 7 (22%) 0 (0%) 15 (47%) 44.6 (9.1) Surgery (n=17) Conservative (n=15) Total (n=32) DESTINY Chapter 11 HAMLET 9 (64%) 3 (21%) 2 (14%) 4 (29%) 4 (29%) 2 (14%) 5/12 (42%) 37.5 (0.6) 147.5 (27.4) 74.6 (16.5) 6 (43%) 23 (21.8-27) Male sex History of TIA/stroke Ischemic heart disease Atrial fibrillation Hypertension Diabetes Current smoker Temperature (mean [SD]) Systolic BP (mean [SD]) Diastolic BP (mean [SD]) Aphasia NIHSS (median [IQR]) 30 (20-41) 27 (22.5-32) 3 (33%) 76.3 (21.9) 148.1 (23.0) 37.5 (0.5) 4 (44%) 0 (0%) 1 (11%) 0 (0%) 0 (0%) 2/8 (25%) 3 (33%) 43.0 (12.6) 30 (24-40) 24 (22-28) 9 (39%) 75.3 (18.3) 147.7 (25.2) 37.5 (0.5) 9/21 (43%) 2 (9%) 5 (22%) 4 (17%) 2 (9%) 5 (22%) 12 (52%) 48.2 (9.9) Total (n=23) a 0.90b 0.77 b 1.00 0.13a 0.014 0.55 a 0.17 0.18 1.00 0.49 0.49 0.49 b 0.65b 0.0033 0.47 0.63a 0.28 a 0.34 a 0.73 0.65 1.00 1.00 0.68 1.00 1.00 0.39a 0.98a 1.00 DESTINY 0.91b 0.13b 1.00 0.83a 0.96a 0.96a 1.00 0.50 0.61 0.13 0.50 1.00 0.21 0.086a HAMLET Comparison of treatment groups DECIMAL Data are number (%) umless otherwise indicated. Group comparisons are Fisher’s exact test unless otherwise indicated. SD indicates standard deviation; IQR, interquartile range; a, t-test or ANOVA as appropriate; b, Mann-Withney U or Kruskal-Wallis test as appropriate. Hours to randomization (median [IQR]) 31 (23-39) 51.6 (6.1) Surgery (n=14) Conservative (n=9) Age (mean [SD]) Table 2 – Continued – Decompressive surgery in MCA infarction: pooled analysis of 3 trials 151 Chapter 11 Figure 1 Distributions of the scores on the mRS and death after 12 months for patients treated with or without surgical decompression. Significantly fewer patients had an unfavorable outcome defined as an mRS score of 5 or death at 12 months after surgery than after conservative treatment (13/51 (25%) patients in the surgical group vs 32/42 (76%) in the conservative group; pooled ARR [95%CI]: 51.2% [33.9-68.5]; Figure 2a). Significantly fewer patients had a mRS > 3 at 12 months after surgical treatment than after conservative treatment (29/51 (57%) in the surgical group vs 33/42 (79%) in the conservative group; pooled ARR [95%CI]: 22.7% [4.6-40.9]; Figure 2b). The survival rate at 12 months was higher after surgical treatment than after conservative treatment (40/51 (78%) in the surgical group vs 12/42 (29%) in the conservative group; pooled ARR [95%CI]: 50.3% [33.3-67.4]; Figure 2c). The results of our analyses remained essentially the same after adjustment for baseline incomparabilities. With regard to all three outcome measures, there was no statistically significant heterogeneity between the three trials. If baseline differences between the treatment groups were taken into account, the reduction in ORs remained essentially the same for all three analyses. The resulting numbers needed to treat [95%CI] for the three outcomes are 2 [1.5-3] for the prevention of mRS 5 or death, 4 [2-22] for the prevention of mRS 4 to death and 2 [1.5-3] for survival irrespective of outcome. Surgery was beneficial (p < 0.01) in all pre-defined subgroups [age (above and below 50 years), presence of aphasia, and time to randomization (above and below 24 hours)], as defined by mRS ≤ 4 at 12 months, with no statistically significant subgrouptreatment effect interactions (Figure 3). 152 Decompressive surgery in MCA infarction: pooled analysis of 3 trials Figure 2 Absolute Risk Reductions and Odds Ratios for unfavorable outcome at 12 months defined as mRS > 4 (a), mRS > 3 (b), and death (c). Outcome / Patients Conservative Surgery ARR (%) 95% CI OR 95% CI 0.10 0.15 0.05 0.02-0.43 0.03-0.73 0.00-0.54 0.10 0.04-0.27 0.29 0.41 0.31 0.07-1.18 0.09-1.81 0.03-3.38 0.33 0.13-0.86 0.10 0.19 0.03 0.02-0.43 0.04-0.94 0.00-0.39 0.10 0.04-0.27 (a) mRS > 4 at 12 months DECIMAL DESTINY HAMLET 14 / 18 10 / 15 8/9 5 / 20 4 / 17 4 / 14 52.8 43.1 60.3 25.8-79.8 11.9-74.4 29.0-91.6 TOTAL 32 / 42 13 / 51 51.2 33.9-68.5 -10 0 Significance: p < 0.0001 Heterogeneity: p = 0.74 10 20 30 40 50 60 70 80 (b) mRS > 3 at 12 months DECIMAL DESTINY HAMLET 14 / 18 11 / 15 8/9 10 / 20 9 / 17 10 / 14 27.8 20.4 17.5 -1.4-56.9 -12.2-53.0 -13.9-48.8 TOTAL 33 / 42 29 / 51 22.7 4.6-40.9 -10 0 Significance: p = 0.014 Heterogeneity: p = 0.89 10 20 30 40 50 60 70 80 (c) Death at 12 months DECIMAL DESTINY HAMLET 14 / 18 8 / 15 8/9 5 / 20 3 / 17 3 / 14 52.8 35.7 67.5 25.8-79.8 4.6-66.8 37.7-97.2 TOTAL 30 / 42 11 / 51 50.3 33.3-67.4 Significance: p < 0.0001 Heterogeneity: p = 0.34 -10 0 10 20 30 40 50 60 70 80 % Absolute Risk Reduction (95% CI) Discussion This pooled analysis of randomized trials confirms suggestions from uncontrolled studies that decompressive surgery performed within 48 hours of stroke onset reduces mortality and increases the number of patients with a favorable functional outcome after space-occupying hemispheric infarction.9,10,12 The numbers needed to treat for survival, survival with an mRS of 4 or lower, and survival with an mRS of 3 or lower are 2, 2, and 4 respectively. The present study is the first in the field of stroke in which a pooled analysis of individual patient data from three independent randomized trials was planned while these trials were still ongoing. This has the obvious advantage of being able to keep the number of patients included to a minimum and to report the results several years earlier than would have been possible based on the individual trials alone. The (expected) results of this pooled analysis have led to premature termination of DESTINY. DECIMAL had 153 Chapter 11 Figure 3 Subgroup analyses of outcome according to age, timing of randomization, and presence of aphasia. Outcome / Patients ARR (%) 95% CI OR 95% CI 3 / 16 1 / 13 4/6 54.6 52.3 16.7 25.1-84.0 18.7-86.0 -31.4-64.8 0.08 0.06 0.40 0.02-0.46 0.01-0.61 0.03-6.18 8 / 35 46.9 26.7-67.0 0.10 0.03-0.35 0.14 0.75 0.01 0.00-4.47 0.03-17.51 0.00-0.51 0.13 0.02-0.76 0.08 0.06 2.00 0.02-0.42 0.00-0.82 0.05-78.25 0.12 0.04-0.43 0.33 0.33 0.02 0.01-16.80 0.04-2.87 0.00-0.42 0.13 0.03-0.54 0.06 0.13 0.03 0.00-0.82 0.01-2.18 0.00-0.74 0.06 0.01-0.31 0.11 0.14 0.25 0.02-0.78 0.02-1.03 0.01-4.73 0.14 0.04-0.50 Conservative Surgery Age < 50 years DECIMAL DESTINY HAMLET 11 / 15 6 / 10 5/6 TOTAL 22 / 31 -20 -10 0 Significance: p < 0.0001 Heterogeneity: p = 0.39 Age 50 years DECIMAL DESTINY HAMLET TOTAL 3/3 4/5 3/3 2/4 3/4 0/8 37.5 5.0 81.9 -17.0-92.0 -50.0-60.0 46.2-117.6 10 / 11 5 / 16 44.5 17.0-72.1 -20 -10 0 Significance: p = 0.0015 Heterogeneity: p = 0.044 Time to randomization < 24 hours 13 / 16 DECIMAL 7/8 DESTINY HAMLET 1/2 TOTAL 21 / 26 5 / 19 2/7 2/3 54.9 58.9 -16.7 27.4-82.5 18.4-99.5 -104.1-70.8 9 / 29 49.7 27.6-71.9 -20 -10 0 Significance: p < 0.0001 Heterogeneity: p = 0.28 Time to randomization 24 hours 1/2 0/1 DECIMAL 3/7 2 / 10 DESTINY 7/7 2 / 11 HAMLET 25.0 22.9 72.9 -57.5-107.5 -21.4-67.1 44.5-101.4 TOTAL 46.9 22.3-71.4 11 / 16 4 / 22 -20 -10 0 Significance: p = 0.0002 Heterogeneity: p = 0.099 No aphasia DECIMAL DESTINY HAMLET TOTAL 5/7 3/4 6/6 1/8 2/7 2/8 58.9 46.4 65.1 18.4-99.5 -7.6-100.5 30.1-100.0 14 / 17 5 / 23 58.2 34.1-82.3 -20 -10 0 Significance: p < 0.0001 Heterogeneity: p = 0.85 Aphasia DECIMAL DESTINY HAMLET 9 / 11 7 / 11 2/3 4 / 12 2 / 10 2/6 48.5 43.6 33.3 13.4-83.6 5.9-81.4 -32.0-98.7 TOTAL 18 / 25 8 / 28 44.2 20.2-68.1 Significance: p = 0.0003 Heterogeneity: p = 0.92 154 -20 -10 0 10 20 10 20 10 20 10 20 10 20 10 20 30 30 30 30 30 30 40 50 40 50 40 50 40 50 40 50 40 50 60 60 60 60 60 60 70 80 70 80 70 80 70 80 70 80 70 80 % Absolute Risk Reduction (95% CI) 90 90 90 90 90 90 Decompressive surgery in MCA infarction: pooled analysis of 3 trials been terminated after the difference in mortality had become significant and the data monitoring committee had recommended stopping enrollment Although a score on the mRS of ≤ 3 is generally accepted as a favorable outcome in stroke research, an mRS ≤ 4 after 12 months was chosen as the primary outcome in this pooled analysis. Given that survival with no or only slight disability after large MCA infarction is rare, the primary aim of the study was to assess whether decompressive surgery reduced mortality without an increase in the number of severely disabled survivors (mRS score of 5). However, decompressive surgery did in fact result in a statistically significant increase in survival with an mRS ≤ 3 after 12 months. The present study shows that after decompressive surgery, the probability of survival increases from 28% to nearly 80% and the probability of survival with an mRS of ≤ 3 almost doubles. However, the probability of surviving in a condition requiring assistance from others (mRS of 4) increases more than ten times, although very severe disability (mRS of 5) is not increased. The choice of performing decompressive surgery in an individual patient with space-occupying hemispheric infarction will therefore depend on the willingness to accept survival with moderate disability. Information on quality of life of survivors is essential for guiding such decisions. Previous studies on quality of life after decompressive surgery for space-occupying infarction have reported divergent results.21-23 Even patients with aphasia may improve significantly.24 Information on quality of life will be provided in the separate publications of the trials. In the three trials under study, patients were excluded if they were older than 55 or 60 years of age. The results can probably not be generalized to patients who are older. In a systematic review of uncontrolled studies on decompressive surgery, 80% of the patients older than 50 years were dead or remained severely disabled compared with 32% of the patients ≤ 50 years.12 Moreover, quality of life may remain impaired especially in older patients.22,23 Data from a large uncontrolled series have suggested that outcome is substantially improved if treatment is initiated within 24 hours of stroke onset as compared with longer time windows for treatment.10 In the above-mentioned systematic review, the timing of surgery did not affect outcome. Similar observations were made in a recent series of patients, in which the mean interval from stroke onset to surgery was 47 hours.25 In the present study, given the limited patient numbers, no difference in outcome was found between patients treated on the first and those treated on the second day. In the majority of patients clinical sings of herniation appear after 2 days of stroke onset.5 Whether decompressive surgery is also beneficial if performed after the first 48 hours, is currently tested in HAMLET.13 The present study has limitations. First, as a result of slightly different eligibility criteria between the individual trials there were differences in baseline characteristics of the patients included. However, there was no statistically significant heterogeneity between the trials in the effect of surgery on any of the outcome measures. Second, 155 Chapter 11 as with most surgical trials, the nature of the treatment under study prevented a fully blinded outcome assessment. Although observer bias cannot be excluded, the consistency of results across the three trials, of which two used some form of blinding, argues against any major bias. Third, subgroup analyses on expected prognostic factors, such as age and the interval between the onset of symptoms and treatment were not powered to show quantitative differences in treatment effect between groups. However, surgery was significantly beneficial in all subgroups, suggesting that there are unlikely to be any qualitative subgroup-treatment effect interactions (i.e. harm in one group and benefit in another). Fourth, there were baseline incomparibilities between the treatment groups. However, analyses adjusted for baseline differences provided essentially the same results. In conclusion, decompressive surgery increases the probability of survival without increasing the number of very severely disabled survivors. Still, the decision to perform decompressive surgery should be made on an individual basis in every patient. References 1. 2. 3. 4. 5. 6. 7. 8. 9. 10. 11. 12. 156 Shaw C, Alvord Jr E, Berry E. Swelling of the brain following ischemic infarction with arterial occlusion. Arch Neurol 1959;1:161-77. Frank JI. Large hemispheric infarction, deterioration, and intracranial pressure. Neurology 1995;45:1286-1290. Sakai K, Iwahashi K, Terada K, Gohda Y, Sakurai M, Matsumoto Y. Outcome after external decompression for massive cerebral infarction. Neurol Med Chir Tokyo 1998;38:131-135. Qureshi AI, Suarez JI, Yahia AM. Timing of neurologic deterioration in massive middle cerebral artery infarction: a multicenter review. Crit Care Med 2003;31:272-277. Hacke W, Schwab S, Horn M, Spranger M, De Georgia M, von Kummer R. ‘Malignant’ middle cerebral artery territory infarction: clinical course and prognostic signs. Arch Neurol 1996;53:309-315. Berrouschot J, Sterker M, Bettin S, Koster J, Schneider D. Mortality of space-occupying (‘malignant’) middle cerebral artery infarction under conservative intensive care. Intensive Care Med 1998;24:620623. Hofmeijer J, van der Worp HB, Kappelle LJ. Treatment of space-occupying cerebral infarction. Crit Care Med 2003;31:617-625. Kasner SE, Demchuk AM, Berrouschot J. Predictors of fatal brain edema in massive hemispheric ischemic stroke. Stroke 2001;32:2117-2123. Rieke K, Schwab S, Krieger D. Decompressive surgery in space-occupying hemispheric infarction: results of an open, prospective trial. Crit Care Med 1995;23:1576-1587. Schwab S, Steiner T, Aschoff A. Early hemicraniectomy in patients with complete middle cerebral artery infarction. Stroke 1998;29:1888-1893. Morley NC, Berge E, Cruz-Flores S, Whittle IR. Surgical decompression for cerebral edema in acute ischaemic stroke. Cochrane Database Syst Rev 2002;CD003435. Gupta R, Connolly ES, Mayer S, Elkind MS. Hemicraniectomy for massive middle cerebral artery territory infarction: a systematic review. Stroke 2004;35:539-543. Decompressive surgery in MCA infarction: pooled analysis of 3 trials 13. 14. 15. 16. 17. 18. 19. 20. 21. 22. 23. 24. 25. Hofmeijer J, Amelink GJ, Algra A, van Gijn J, Macleod MR, Kappelle LJ, van der Worp HB. Hemicraniectomy After Middle cerebral artery infarction with Life-threatening Edema Trial (HAMLET). Protocol for a randomised controlled trial of decompressive surgery in space-occupying hemispheric infarction. Trials 2006;7:29. van Swieten JC, Koudstaal PJ, Visser MC, Schouten HJ, van GJ. Interobserver agreement for the assessment of handicap in stroke patients. Stroke 1988;19:604-607. Wirtz CR, Steiner T, Aschoff A. Hemicraniectomy with dural augmentation in medically uncontrollable hemispheric infarction. Neurosurg Focus 1997;2:E3. Hacke W, Kaste M, Bogousslavsky J. European Stroke Initiative Recommendations for Stroke Management-update 2003. Cerebrovasc Dis 2003;16:311-337. Adams HP, Jr., Adams RJ, Brott T. Guidelines for the early management of patients with ischemic stroke: A scientific statement from the Stroke Council of the American Stroke Association. Stroke 2003;34:1056-1083. Bereczki D, Liu M, do PG, Fekete I. Mannitol for acute stroke (Cochrane Review). Cochrane Database Syst Rev 2001 Jan;1:CD001153. Brott T, Adams HP, Jr., Olinger CP. Measurements of acute cerebral infarction: a clinical examination scale. Stroke 1989;20:864-870. McCullagh P. Regression Models for Ordinal Data. Journal of the Royal Statistical Society, Series B 1980;42:109-42. Woertgen C, Erban P, Rothoerl RD, Bein T, Horn M, Brawanski A. Quality of life after decompressive craniectomy in patients suffering from supratentorial brain ischemia. Acta Neurochir (Wien ) 2004;146:691-695. Foerch C, Lang JM, Krause J. Functional impairment, disability, and quality of life outcome after decompressive hemicraniectomy in malignant middle cerebral artery infarction. J Neurosurg 2004;101:248-254. Curry WT, Jr., Sethi MK, Ogilvy CS, Carter BS. Factors associated with outcome after hemicraniectomy for large middle cerebral artery territory infarction. Neurosurgery 2005;56:681-692. Kastrau F, Wolter M, Huber W, Block F. Recovery from aphasia after hemicraniectomy for infarction of the speech-dominant hemisphere. Stroke 2005;36:825-829. Rabinstein AA, Mueller-Kronast N, Maramattom BV. Factors predicting prognosis after decompressive hemicraniectomy for hemispheric infarction. Neurology 2006;67:891-893. 157 Discussion Chapter 12 General discussion 159 Chapter 12 T his thesis describes the preparation and preliminary results of a randomized clinical trial to study the effect of decompressive surgery in patients with middle cerebral artery (MCA) infarction who deteriorate as a result of spaceoccupying brain edema. The rationale of surgical decompression is to accommodate shift of brain tissue and to normalize intracranial pressure, thereby preserving cerebral blood flow and preventing secondary damage. This treatment has been proposed in the past, since patients with a hemispheric infarct and massive edema have a poor prognosis, with a case fatality rate of about 80%, despite maximal medical therapy in intensive care-based series.1,2 Moreover, there is no medical treatment of proven efficacy (chapter 2). Case reports and small patient series have suggested a substantial reduction in death rate after surgical decompression.3-7 As decompressive surgery may reduce brain tissue shifts and intracranial pressure, but is not expected to reduce tissue damage in the infarct itself, most patients will be left with a severe neurological deficit.8 Given this deficit, many neurologists are reluctant to recommend hemicraniectomy as a lifesaving treatment in patients who deteriorate as a result of edema formation. In 2001, an interview among Dutch neurologists disclosed that this intervention had been performed in only 3 of 15 hospitals with neurosurgical facilities, and only sporadically. However, other experts think decompressive surgery should be implemented as a standard treatment modality, because uncontrolled studies showed a large reduction in case fatality, without increasing the amount of severely disabled survivors.6,7 In agreement with the current standard of evidence-based medicine (EBM), we therefor initiated a randomized controlled trial (RCT).9 Expert opinion and evidence-based medicine EBM has replaced pathophysiological reasoning and expert opinion as the basis of clinical decision making.9 EBM relies on systematic, reproducible, unbiased clinical observation, and marks pathophysiological reasoning and expert opinion as insufficient, or even inaccurate, in guiding definite clinical decisions. The two pillars of EBM are RCTs and meta-analyses,9 which provide the best (Level 1) evidence. Observational studies, such as cohort and case-control studies are assigned a level of evidence of 2. Level 3, the lowest level, is assigned to consensus guidelines, expert opinion, and common clinical practice.9 The term EBM seems to imply that clinical medicine which is not based on RCTs or meta-analyses, is not based on evidence. Before the concept of EBM was introduced, experienced authority, manifested as the ‘expert opinion’, was assumed to be the highest source of knowledge in clinical medicine. Collecting knowledge depended on accumulating observations. The more observations were made (the greater the clinical experience), the more confidently knowledge could be extrapolated.10 This way of collecting knowledge may be referred to as ‘inductive logic’, which was well accepted 160 Discussion according to research traditions after the Enlightenment. The role of expert opinion in the context of EBM has been largely ignored. EBM assumes a reality, which is only approximately knowable, based on statistical probability. The basic mode of investigation involves hypothesis testing. Therefore, it would be tempting to describe EBM as based on ‘deductive logic’. The major problem with this interpretation is that the hypotheses tested in EBM are not axiomas, but ideas formulated by clinical experts, and thereby the result of induction. Moreover, if different RCTs on the same treatment would show conflicting results, and metaanalyses are not conclusive, consensus on the implementation of the treatment under study still has to be obtained from experts in the field. The logic involved in the process of implementing results of RCTs is inductive as well. The greater the accumulated evidence, the greater the confidence in the conclusion. Although EBM is built on the premise that expert opinion is not the best source for solving medical problems, it relies on expert opinion in its first stages (hypothesis formulation) and in its final stages (accepting the accumulated evidence as sufficient). Moreover, access to medical journals for the introduction of new treatment strategies may be easier for authors with authority in the field than for relative newcomers, which may create publication bias by expert opinion. Until very recently, the decision to perform hemicraniectomy in patients with space-occupying MCA infarction was based on ‘expert opinion’, extrapolated from uncontrolled patient series. Most uncontrolled and controlled trials on hemicraniectomy in patients with space-occupying infarction have been conducted by the same experts with belief in the benefit of the treatment. This influences the a priori probability of confirming the hypothesis that surgical decompression is of benefit in patients with space-occupying MCA infarction; thereby this belief also influences interpretation and presentation of trial data. The individual patient In an RCT groups of patients are analyzed, whereas in clinical practice the individual patient is the unit of observation. To implement the results of RCTs into clinical practice in a very strict way, each patient should be reduced to a set of prognostic factors and subsequently compared with similar patients treated in the trial. For RCTs with reasonably objective outcome measures (for example a new stroke), weighing the available evidence may be relatively easy. However, for trials with less objective outcome measures, such as functional outcome and quality of life, advantages and disadvantages are a matter of personal judgment. Interpretations become important in the process of implementing scientific evidence in patient care and therefore it is tempting to standardize treatment decisions. Scientific reasoning is an essential part of clinical decision making. However, 161 Chapter 12 clinical medicine not only consists of science, but also of humanitariarism.11 Scientific thinking is based on observation and deduction, whereas humanitarian thinking is based on empathy and ethical norms. Treatments that can be given are not always the ones that ought to be given. Clinical decisions are not solely based on the results of scientific research but also on value judgments. In a prospective study among nonselected consecutive patients admitted in a general hospital, approximately 25 percent of the treatment decisions was influenced by ethical problems or value judgments.12 Value judgments become more important if subjective measures, such as quality of life, are concerned. Ethical norms vary between doctors, patients, and societies, and so do ideas about quality of life. Despite the lack of Level 1 evidence, almost every neurologist believes that decompressive surgery reduces the risk of death in patients with space-occupying MCA infarction. However, since a substantial proportion of the surviving patients is left severely disabled, the answers to the question whether or not this treatment is ‘good’ are divergent. Some experts believe it is unethical to withhold decompressive surgery because of the reduction in death rate and the reputedly modest proportion of severely disabled survivors. Others think the neurological deficit in survivors is so severe that keeping patients alive is unethical. This disagreement is probably not only the result of different doctors seeing patients with different infarcts. Personal ideas about the value and quality of life with a severe neurological deficit also play a role. An RCT may help to assess handicaps of patients surviving after space-occupying MCA infarction with or without hemicraniectomy in a systematic fashion. However, it will not solve the divergence of opinion about the value of life of survivors. Still, the results of HAMLET and other RCTs described in this thesis will undoubtedly play an important role in the decision whether or not to perform hemicraniectomy in a future patient with a life-threatening MCA infarct. Utilitarian considerations play an important role in the process of these kinds of clinical decisions. These considerations require not only the valuation of the outcomes of the two treatment options, but also assessment of the probability that either of the outcomes will occur. The estimation of the probability of different outcomes requires empirical evidence; this aspect can certainly be provided by an RCT.11 Animal experiments Experimental stroke research has contributed much to the understanding of ischemic brain damage, but its contribution to the current management of stroke patients is poor.13 More than 500 neuroprotective agents have been shown to reduce damage in models of cerebral ischemia,14 but the only treatment for patients with acute ischemic stroke that has been proven effective in RCTs is trombolysis.15 Remarkably, the thrombolysis trials were not based on animal experiments. 162 Discussion There are several explanations for the discrepancies between experimental and clinical studies. Functional outcome is the most relevant outcome measure in clinical trials. The relationship between infarct volume and functional outcome of patients is marginal at best,16,17 let alone the relationship between infarct volume in animals and functional outcome in patients. Extrapolation from animal models to the clinic becomes even more difficult as the severity of the clinical condition increases.18 Therefore, stroke studies in animals are largely explanatory, and should be extrapolated to the clinic only with great care. Although animal research mainly is intended for explanatory purposes, its predictive validity is strongly dependent on the choice of the animal model. Previous studies on the temporal evolution of cerebral infarction in rats, and studies on hemicraniectomy in rats with space-occupying infarction, have been performed with models of variable or moderate focal cerebral ischemia and may therefore not reflect the syndrome of space-occupying infarction.19-22 In chapter 3 a more adequate model of spaceoccupying infarction is presented, with consistently large infarct volumes, substantial midline shift, and high death rates, reflecting this clinical syndrome in man. This model was used to study the course of MRI indices of cerebral perfusion and tissue damage before (chapters 4) and after (chapter 5) decompressive surgery. It was shown that in the presence of space-occupying edema, perfusion values were reduced not only in the core of the infarct, but also in ‘healthy’ tissue surrounding the infarct. After hemicraniectomy, cerebral blood flow increased in the cerebral tissue directly surrounding the infarct, potentially protecting this tissue from secondary ischemic damage. This is in line with the hypothesis formulated to explain a potential benefit of decompressive surgery on functional outcome7 and implies that patients without clinical signs of herniation might benefit from this treatment as well. However, despite a pseudo-normalization of apparent diffusion coefficient (ADC) values within the infarct core, histological examination revealed irreversible damage of brain tissue in the complete territory of the MCA, indicating severe permanent damage. Ethical aspects of animal experiments The recognition of the intrinsic value of animals is one of the main aspects of the legislation of animal experiments. The European Science Foundation acknowledges, among other rules, that infringement of the intrinsic value is acceptable only if the consequences for humanity of not performing the experiments are more important than the inconvenience caused to the animal (www.esf.org). Since most experimental research has an explanatory nature, the importance for mankind of many animal experiments can be doubted. A critical reappraisal of the acceptability of these experiments may reveal inconsistencies in the handling of this rule. Acknowledgement of the intrinsic value of animals leads to equality of animals and 163 Chapter 12 humans in many moral respects. In case of approximately moral equality, criteria used for the appraisal of experiments on animals and patients should be similar. However, in general, ethical committees are much more rigid in permitting clinical trials with patients than in permitting experiments on animals. Also, methodological flaws have been readily accepted in experimental research, whereas statistical rules of clinical trials are very strict.23 Moreover, it is almost impossible to publish negative results of animal experiments, whereas in clinical trials the finding that a treatment modality does not change outcome measures is often judged almost equally valuable as the opposite result. Publication bias caused by the impossibility to publish negative results undoubtedly results in unnecessarily repetition of animal experiments and unjustified initiation of clinical trials.23,24 Treatment of space-occupying MCA infarction Decompressive surgery has to be considered in every patient with a large hemispheric infarct deteriorating as a result of brain edema formation in the first 48 h after stroke onset, because in the pooled interim analysis of individual patient data of three recent randomized controlled clinical trials a favorable outcome was significantly more likely after hemicraniectomy than after conservative treatment (chapter 11). Several considerations have to be taken into account. Although favorable outcome was more likely after hemicraniectomy, a large proportion of surviving patients was left with severe disability. In the primary analysis, favorable outcome was defined as a score on the modified Rankin Scale (mRS)25 of 4 or better, which means that patients could be ‘moderately disabled’. By definition, these patients were unable to walk independently (with or without a cane or walker) and were unable to attend to their own bodily needs without assistance. The choice of performing decompressive surgery in an individual patient with space-occupying hemispheric infarction will therefore depend on the willingness to accept survival with a moderate to severe disability. Neuropsychological examination in a cohort of patients after decompressive surgery for space-occupying hemispheric infarction showed severe cognitive deficits in most patients (chapter 10). In patients with aphasia, language was compromised to such an extent that communication was very difficult or even impossible. Apart from severe focal deficits general cognitive decline was found in most patients. Cognitive impairment after stroke has been reported in several studies,26 and may limit the patient’s ability to achieve life satisfaction.27 Furthermore, neuropsychiatric disorders, such as depression, occur in 30% to 40% of stroke survivors.28 Not only motor function, but also depression, cognitive impairment, and incontinence were identified as major determinants of health-related quality of life after stroke.29 Information on quality of life of survivors is essential for treatment decisions in patients with space-occupying infarction and will be provided in future publications of the trials. 164 Discussion In controlled intervention studies on hemicraniectomy in patients with spaceoccupying infarction such as HAMLET30 and other trials, feasibility considerations play a role. Recruiting patients is difficult and laborious. For ethical reasons, the number of patients to be randomized should be kept to a minimum and if hemicraniectomy is of benefit for the group as a whole, trial results should be made public to implement this treatment in clinical practice. However, since impairments may differ between different subgroups of patients, e.g. those with aphasia, those of old age, and those with concomitant infarction in the posterior or anterior cerebral artery territory, it will remain questionable whether hemicraniectomy should be introduced as a standard treatment modality for every patient with impending herniation after cerebral infarction. Early termination of clinical trials may lead to exaggerated treatment effects, influencing decision making from the level of the individual patient to the level of international guidelines.31 Early termination may also prevent sufficient subgroup analyses and carries the risk that surgery will be performed on the basis of ‘level 1’ evidence in patients who will not truly benefit. Subgroup analyses should at least focus on age, interval to treatment, presence of aphasia, and involvement of other vascular territories besides that of the MCA. With the projected number of 112 inclusions, HAMLET is a relatively small trial. To prevent loss of power without introducing selection bias, an alternative method of randomizing patients is used (chapter 7). Because hemicraniectomy is supposed to reduce case fatality to a large extent, randomized treatment allocation may be considered unethical and unacceptable by the patients’ legal representatives. However, structured interviews showed no differences in trial appreciation between HAMLET participants and participants in a less dramatic trial, which investigates the effect of paracetamol in stroke patients (chapter 9). The results of the pooled analysis play an important role in the decision to perform early hemicraniectomy in patients with life-threatening MCA infarction. However, weighing the pros and cons will to a large extent remain an individual judgement. Moreover, quality of life and depression are important secondary outcome measures in the separate clinical trials; the full reports should be awaited before the treatment is implemented as standard. For individual patients, the ‘best clinical evidence’ derived from RCTs should be combined with individual expertise and personal value judgments. In case of the decision to withhold surgical treatment because of severe neurological damage, the indication for medical treatment can be doubted for the same reason. Future research Decompressive hemicraniectomy has to be considered on an individual basis in patients with a large hemispheric infarct who deteriorate as a result of progressive 165 Chapter 12 brain edema. Future research should focus on patient selection. It remains unclear which patients with MCA infarction are at risk for developing life-threatening edema (chapter 6) and studies on this topic are necessary. It is unlikely that trials will be repeated to answer the question which patients benefit most from hemicraniectomy, so data on subgroups should be largely extrapolated from ongoing RCTs, especially with regard to age, interval to treatment, presence of aphasia, and involvement of other vascular territories besides that of the MCA. Timing of the intervention is another subject of interest. Hemicraniectomy not only prevents death by herniation, but it may also prevent secondary ischemic damage by improving cerebral perfusion (chapter 5). This suggests that patients without clinical signs of herniation might benefit as well. Clinical trials on hemicraniectomy in patients with large MCA infarction at risk for massive edema formation may therefore be a next step. Ultimately, with the help of sufficiently sensitive and specific prognostic factors it should become possible to inform individual patients on their prognosis with or without hemicraniectomy. Conclusions • • • • A pooled analysis of individual patient data of ongoing randomized trials has demonstrated that decompressive surgery performed within 48 hours after the onset of symptoms in patients with space-occupying hemispheric infarction leads to a reduction in case fatality and an increased probability of a favorable outcome. Still, many patients are left severely disabled and it remains tempting to extrapolate these negative examples to future patients. Early termination of ongoing trials should be prevented, because subgroup analyses are necessary. Although the decision to perform decompressive surgery in patients with spaceoccupying hemispheric infarction involves ethical and subjective considerations, RCTs offer important information on outcome of these patients. For individual patients, the ‘best clinical evidence’ derived from RCTs should be judged in the context of individual values and desires. Future research should focus on prognostic factors to investigate which patients with MCA infarction are at risk for developing life-threatening edema and which patients benefit most from hemicraniectomy. Experimental evidence supports the notion that hemicraniectomy not only prevents death by herniation, but may also prevent secondary ischemic damage, by improving cerebral perfusion. This suggests that patients with MCA infarction at risk of massive edema formation, but without life-threatening brain shift, may benefit from this treatment as well. 166 Discussion References 1. 2. 3. 4. 5. 6. 7. 8. 9. 10. 11. 12. 13. 14. 15. 16. 17. 18. 19. 20. Hacke W, Schwab S, Horn M, Spranger M, De Georgia M, von Kummer R. ‘Malignant’ middle cerebral artery territory infarction: clinical course and prognostic signs. Arch Neurol 1996;53:309-315. Wijdicks EF, Diringer MN. Middle cerebral artery territory infarction and early brain swelling: progression and effect of age on outcome. Mayo Clin Proc 1998;73:829-836. Schwab S, Rieke K, Aschoff A, Albert F, von Kummer R, Hacke W. Hemicraniotomy in space-occupying hemispheric infarction: useful early intervention or desparate activism? Cerebrovasc Dis 1996;6:325329. Carter BS, Ogilvy CS, Candia GJ, Rosas HD, Buonanno F. One-year outcome after decompressive surgery for massive nondominant hemispheric infarction. Neurosurgery 1997;40:1168-1175. Koh MS, Goh KY, Tung MY, Chan C. Is decompressive craniectomy for acute cerebral infarction of any benefit? Surg Neurol 2000;53:225-230. Rieke K, Schwab S, Krieger D. Decompressive surgery in space-occupying hemispheric infarction: results of an open, prospective trial. Crit Care Med 1995;23:1576-1587. Schwab S, Steiner T, Aschoff A. Early hemicraniectomy in patients with complete middle cerebral artery infarction. Stroke 1998;29:1888-1893. Bamford J, Sandercock P, Dennis M, Burn J, Warlow C. Classification and natural history of clinically identifiable subtypes of cerebral infarction. Lancet 1991;337:1521-1526. Evidence-Based Medicine Working Group. Evidence-based medicine. A new approach to teaching the practice of medicine. JAMA 1992;268:2420-2425. Kulkarni AV. The challenges of evidence-based medicine: a philosophical perspective. Med Health Care Philos 2005;8:255-260. Wulff H, Gotzsche P. Medicine and the Humanities. In: Wulff H, Gotzsche P, eds. Rational Diagnosis and Treatment, third ed. Oxford: Blackwell Science, 2000:147-174. Kollemorten I, Strandberg C, Thomsen BM. Ethical aspects of clinical decision-making. J Med Ethics 1981;7:67-69. van der Worp HB. Treatment of acute ischaemic stroke with antioxidants. The gap between laboratory and practice. Utrecht University, doctorate thesis. 1999 O’Collins VE, Macleod MR, Donnan GA, Horky LL, van der Worp BH, Howells DW. 1,026 experimental treatments in acute stroke. Ann Neurol 2006;59:467-477. Tissue plasminogen activator for acute ischemic stroke. The National Institute of Neurological Disorders and Stroke rt-PA Stroke Study Group. N Engl J Med 1995;333:1581-1587. Saver JL, Johnston KC, Homer D. Infarct volume as a surrogate or auxiliary outcome measure in ischemic stroke clinical trials. The RANTTAS Investigators. Stroke 1999;30:293-298. Lyden PD, Zweifler R, Mahdavi Z, Lonzo L. A rapid, reliable, and valid method for measuring infarct and brain compartment volumes from computed tomographic scans. Stroke 1994;25:2421-2428. Beynen AC, Kort WJ. Animal models. In: van Zutphen LFM, Baumans V, Beynen AC, eds. Handboek proefdierkunde, 3 ed. Maarssen: Elsevier gezondheidszorg, 2001:202-211. Doerfler A, Forsting M, Reith W. Decompressive craniectomy in a rat model of “malignant” cerebral hemispheric stroke: experimental support for an aggressive therapeutic approach. J Neurosurg 1996;85:853-859. Forsting M, Reith W, Schabitz WR. Decompressive craniectomy for cerebral infarction. An experimental study in rats. Stroke 1995;26:259-264. 167 Chapter 12 21. 22. 23. 24. 25. 26. 27. 28. 29. 30. 31. 168 Engelhorn T, Doerfler A, Kastrup A. Decompressive craniectomy, reperfusion, or a combination for early treatment of acute “malignant” cerebral hemispheric stroke in rats? Potential mechanisms studied by MRI. Stroke 1999;30:1456-1463. Engelhorn T, von Kummer R, Reith W, Forsting M, Doerfler A. What is effective in malignant middle cerebral artery infarction: reperfusion, craniectomy, or both? An experimental study in rats. Stroke 2002;33:617-622. van der Worp HB, de Haan P, Morrema E, Kalkman CJ. Methodological quality of animal studies on neuroprotection in focal cerebral ischaemia. J Neurol 2005;252:1108-1114. Macleod MR, O’Collins T, Howells DW, Donnan GA. Pooling of animal experimental data reveals influence of study design and publication bias. Stroke 2004;35:1203-1208. van Swieten JC, Koudstaal PJ, Visser MC, Schouten HJ, van Gijn J. Interobserver agreement for the assessment of handicap in stroke patients. Stroke 1988;19:604-607. Sachdev PS, Brodaty H, Valenzuela MJ, Lorentz LM, Koschera A. Progression of cognitive impairment in stroke patients. Neurology 2004;63:1618-1623. Clarke P, Marshall V, Black SE, Colantonio A. Well-being after stroke in Canadian seniors: findings from the Canadian Study of Health and Aging. Stroke 2002;33:1016-1021. Ramasubbu R, Patten SB. Effect of depression on stroke morbidity and mortality. Can J Psychiatry 2003;48:250-257. Haacke C, Althaus A, Spottke A, Siebert U, Back T, Dodel R. Long-term outcome after stroke: evaluating health-related quality of life using utility measurements. Stroke 2006;37:193-198. Hofmeijer J, Amelink GJ, Algra A, van Gijn J, Macleod MR, Kappelle LJ, van der Worp HB; the HAMLET investigators. Hemicraniectomy After Middle cerebral artery infarction with Lifethreatening Edema Trial (HAMLET). Protocol for a randomised controlled trial of decompressive surgery in space-occupying hemispheric infarction. Trials 2006;7:29. Altman DG, Schulz KF, Moher D. The revised CONSORT statement for reporting randomized trials: explanation and elaboration. Ann Intern Med 2001;134:663-694. Appendix Appendix 169 Appendix The HAMLET investigators Executive Committee - A. Algra, G.J. Amelink, J. van Gijn, J. Hofmeijer (trial coordinator), L.J. Kappelle, M.R. Macleod (UK national coordinator), and H.B. van der Worp (principal investigator). The Steering Committee is constituted of the principal investigators of each actively randomizing centre (S.F.T.M. de Bruijn, G.J. Luijckx, R. van Oostenbrugge, J. Stam, and J.Th.J. Tans) and of the members of the executive committee. Data Monitoring Committee - Y. van der Graaf , P.J. Koudstaal, and A.I.R. Maas. Advisory Committee - G.W. van Dijk, W. Hacke (Chairman), C.J. Kalkman, C.A.F. Tulleken, and C.A.C. Wijman. Research nurse - M. van Buuren Participating centers and investigators (local principal investigators marked with *) University Medical Centre Utrecht: A. Algra, G.J. Amelink, J. van Gijn, J. Hofmeijer, L.J. Kappelle, H.B. van der Worp, Academic Medical Centre, Amsterdam: J. Stam,* G.J. Bouma, W.P. Vandertop, University Medical Centre Groningen: G.J. Luijckx,* J.J.A. Mooij, J.D.M. Metzemakers, Academic Hospital Maastricht: R. van Oostenbrugge,* J. Dings, Medical Centre Haaglanden, The Hague: J.Th.J. Tans,* J.A.L. Würzer, J. Boiten Haga Hospital, The Hague: S.F.T.M. de Bruijn,* C.F.E. Hoffmann. 170 Summary Summary 171 Summary Patients with a hemispheric infarct and massive space-occupying edema formation have a poor prognosis. Mortality rates of about 80% have been described, despite maximal medical therapy on an intensive care unit. The aim of the research presented in this thesis was to investigate the benefit of decompressive surgery in patients with middle cerebral artery (MCA) infarction who deteriorate as a result of space-occupying brain edema. In part 1, experimental studies on this subject are described. In part 2, preparations for and preliminary results of the ongoing randomized Hemicraniectomy After MCA infarction with Life-threatening Edema Trial (HAMLET) are presented. Chapter 2 describes the different treatment modalities for patients with MCA infarction, deteriorating as a result of edema formation. Various treatment strategies have been proposed to limit brain tissue shifts and to reduce intracranial pressure, such as osmotic therapy, hyperventilation, and sedation with barbiturates. However, there is no evidence of efficacy of any of these therapies, and several reports suggest that these are ineffective or even detrimental. It is concluded that there is no specific treatment modality of proven efficacy for patients with space-occupying hemispheric infarction. In chapter 3 a rat model of space-occupying cerebral infarction is presented, obtained by permanent intra-luminal MCA occlusion in Fisher rats. The time courses of ischemic damage and cerebral perfusion were studied by means of magnetic resonance imaging (MRI). At 24 hours of ischemia, midline shift peaked and perfusion of the tissue surrounding the infarct was maximally decreased, suggesting that this surrounding tissue was at risk for secondary damage as a result of hypoperfusion. It is concluded that therapies to reduce edema and to improve perfusion may be most efficacious if started before 24 hours of ischemia in this model. Studies of cerebral perfusion were performed in the above-mentioned model with two different MRI techniques (chapter 4). The commonly used dynamic susceptibility contrast (DSC)-MRI technique, which is dependent on intravenously injected contrast agent, was compared with non-invasive perfusion-weighted MRI (Flow-sensitive Alternating Inversion Recovery (FAIR)). FAIR correlated with DSC-MRI if perfusion was preserved (r2 = 0.719, P < 0.001), but provided lower perfusion values in areas with severe hypoperfusion (mean FAIR-aCBF ipsi- versus contralateral ratio and mean DSC-rCBF ipsi- versus contralateral ratio 0.39 ± 0.14 and 0.50 ± 0.14 respectively, P = 0.008). These areas were not only found within the infarct, but also in vascular territories other than that of the MCA in the same hemisphere. These territories are at risk for secondary damage and may be saved by effective treatment. In rats with space-occupying MCA infarction, perfusion improved in brain tissue surrounding the infarct after surgical decompression (25 ± 9 ml/min/100g tissue before 172 Summary and 38 ± 9 ml/min/100g tissue after surgical decompression, P = 0.035), indicating that secondary damage may be prevented by decompression (chapter 5). Apparent Diffusion Coefficient values seemed to normalize within the infarct, but, according to histological examination, this was a sham effect. Thus, secondary damage in tissue surrounding the infarct may be prevented by decompressive surgery, but the infarct itself remains permanently damaged. In chapter 6 the results of a meta-analysis of predictors of fatal edema formation in patients with MCA infarction are presented. It is concluded that infarct size is the major determinant. Unfortunately, single clinical and radiological variables, such as age, body temperature, infarct size, involvement of other vascular territories, or internal carotid artery occlusion lack sufficient predictive value to be useful in clinical practice. In chapter 7 a new method for randomizing patients to one of two treatments in a clinical trial is described. The method is based on minimization, to which an element of chance is added. The probability of allocating one of two treatments depends on the actual imbalance between the groups at the time of randomization. In trial simulations with different numbers of patients and prognostic factors, lower levels of imbalance between treatment groups and bias by predictability were obtained than with pure randomization or minimization, respectively, as well as compared with allocation according to a biased coin principle. The method may be of use in relatively small trials, when loss of power as a result of imbalance between treatment groups becomes important. The study protocol of HAMLET is presented in chapter 8. HAMLET is a multi-centre, randomized clinical trial aimed to compare the efficacy of decompressive surgery with best medical treatment on functional outcome in patients with supratentorial infarction with life-threatening space-occupying edema. One hundred and twelve patients will be included, aged between 18 and 60 years, with a large hemispheric infarct with space-occupying edema that leads to a decrease in consciousness. The primary outcome measure is functional outcome, as determined by the score on the modified Rankin Scale, at one year. HAMLET started recruiting in September 2002. As of January 1st, 58 patients had been included. Because of the life-threatening situation at the time of study inclusion and the suggested large reduction in mortality by surgical decompression, randomized treatment allocation in HAMLET may be considered unethical and unacceptable. Legal representatives of patients included in HAMLET were interviewed to study how they had experienced the informed consent procedure and which trial information they could recall after one year. The results are described in chapter 9. Most representatives considered trial participation and randomization acceptable, despite the serious 173 Summary illness of their relatives. Recall of trial information was better in representatives of HAMLET patients than in relatives of patients with less severe strokes participating in the Paracetamol (Acetaminophen) In Stroke (PAIS) study. The results of neuropsychological testing in an unselected cohort of 10 patients with space-occupying MCA infarction one year after decompressive surgery are presented in chapter 10. In addition to the expected severe focal deficits, such as aphasia and neglect, global cognitive decline was found in the majority of patients. In patients with infarcts in the dominant hemisphere, language was compromised to such an extent that communication was very difficult or even impossible. Despite similar scores on the modified Rankin Scale, cognitive functioning in patients with an infarct in the dominant hemisphere was worse than in patients with an infarct in the non-dominant hemisphere. To obtain sufficient data to reliably estimate the effects of early decompressive surgery in patients with space-occupying MCA infarction as soon as possible, individual data of patients from three ongoing European randomized controlled trials treated within 48 hours after stroke onset were pooled. The results of the analysis of the pooled data are summarized in chapter 11. Ninety-three patients were included. After decompressive surgery, more patients had an mRS≤4 (75% vs 24%; pooled absolute risk reduction (ARR) [95% confidence interval (95%CI)]: 51% [34-69]), an mRS≤3 (43% vs 21%; ARR: 23% [5-41]), and survived (78% vs 29%; ARR: 50% [33-67]). The effect of surgery was highly consistent across the three trials. Thus, decompressive surgery reduced mortality and increased the probability of a good functional outcome. However, the probability of surviving in a condition requiring assistance from others also increased substantially. It is concluded that the choice of performing decompressive surgery in an individual patient with space-occupying hemispheric infarction will depend on the willingness to accept survival with moderate disability. Information on quality of life of survivors is essential for guiding such decisions and will be provided in future publications of the trials. The implications of the studies reported in this thesis are discussed in chapter 12. It is concluded that, although the decision to perform decompressive surgery in patients with space-occupying hemispheric infarction comprises ethical and subjective aspects, randomized clinical trials offer important information on outcome of these patients. For individual patients, the ‘best clinical evidence’ derived from randomized controlled trials should be judged in the context of individual values and desires. Early termination of ongoing trials should be prevented, because subgroup analyses are necessary, at least with regard to age, interval to treatment, presence of aphasia, and involvement of other vascular territories besides that of the MCA. Future research should focus on prognostic factors to investigate which patients with MCA infarction are at risk of developing lifethreatening edema and which will benefit from hemicraniectomy most. 174 Samenvatting Samenvatting 175 Samenvatting Grote infarcten in het stroomgebied van de arteria cerebri media (ACM) kunnen gepaard gaan met oedeemvorming, die in ernstige gevallen kan leiden tot cerebrale inklemming. Patiënten met een dergelijk ruimte-innemend herseninfarct hebben hierdoor een slechte prognose; in prospectief onderzoek was de sterfte ongeveer 80%, ondanks maximale conservatieve therapie. Overlevenden zijn meestal ernstig gehandicapt. Omdat het effect van conservatieve therapie waarschijnlijk gering is, adviseren sommige neurologen en neurochirurgen om bij patiënten met een ruimte-innemend herseninfarct een chirurgische decompressie uit te voeren door een groot deel van het schedeldak en het harde hersenvlies weg te nemen. Door de operatie kan het beschadigde hersenweefsel buiten de schedel zwellen, waardoor binnen de schedel weefselverschuivingen en compressie van gezond hersenweefsel worden voorkomen. Het doel van deze behandeling is de cerebrale doorbloeding te verbeteren en secundaire hersenschade te voorkomen. De meeste in de literatuur beschreven ervaringen met chirurgische decompressie bij patiënten met ruimte-innemende herseninfarcten betreffen relatief kleine groepen patiënten, met in het algemeen gunstige resultaten. In twee grotere prospectieve onderzoeken was de sterfte bij conservatief behandelde patiënten ongeveer 78% en bij patiënten die geopereerd waren respectievelijk 34 en 16%. Gesuggereerd werd dat de kans op ernstige neurologische restverschijnselen niet toenam en dat de kans op een goed herstel beter was naarmate de operatie eerder plaatsvond. Deze twee prospectieve onderzoeken suggereren een groot gunstig effect van chirurgische behandeling. De onderzoeken kenden echter zoveel methodologische tekortkomingen dat de resultaten niet direct geëxtrapoleerd kunnen worden naar de praktijk. Ook is onduidelijk hoe het functionele herstel van de overlevende patiënten is. In dit proefschrift wordt onderzoek beschreven naar het effect van chirurgische decompressie bij patiënten met een herseninfarct in het stroomgebied van de ACM die neurologische achteruit gaan als gevolg van hersenoedeem. Deel 1 bestaat uit dierexperimenteel onderzoek en deel 2 uit de voorbereidingen en de eerste resultaten van een lopend multi-centrisch gerandomiseerd klinisch onderzoek naar het effect van chirurgische decompressie op het functionele herstel van patiënten met een ACM infarct die achteruit gaan als gevolg van oedeem: de Hemicraniectomy After MCA infarction with Life-threatening Edema Trial (HAMLET). In hoofdstuk 2 wordt samengevat welke behandelingsmogelijkheden bestaan voor patiënten met een ruimte-innemend herseninfarct. Er zijn diverse behandelingen beschreven om oedeemvorming en intracraniële drukverhoging tegen te gaan. Deze bestaan onder meer uit toediening van mannitol, glycerol, corticosteroïden of furosemide, hyperventilatie en sedatie met barbituraten. Van geen van deze behandelingen is in klinische trials een gunstig effect aangetoond. Er wordt door sommige onderzoekers zelfs gesuggereerd dat deze weinig effectief of schadelijk zijn. 176 Samenvatting Er bestaat geen bewezen effectieve behandeling voor patiënten met een ACM infarct die neurologisch verslechteren als gevolg van hersenoedeem. In hoofdstuk 3 wordt een rattenmodel beschreven van het ruimte-innemend herseninfarct. De infarcten zijn geïnduceerd door intraluminale occlusie van de ACM bij Fisher ratten. Het beloop van de weefselschade en de doorbloeding zijn bestudeerd met behulp van Magnetic Resonance Imaging (MRI). Na 24 uur was de zwelling maximaal en was de doorbloeding van nog niet geïnfarceerd hersenweefsel rond het infarct het sterkst verminderd. Dit suggereert dat het nog gezonde hersenweefsel rond het infarct is bedreigd en dat therapieën om dit weefsel te behouden in dit model binnen 24 uur na ontstaan van het infarct gestart moeten worden. De doorbloeding van de hersenen in het model is uitgebreid bestudeerd (hoofdstuk 4). Daarvoor is gebruik gemaakt van twee MRI technieken, namelijk de standaard techniek, waarbij contrastvloeistof wordt ingespoten (dynamic susceptibility contrast magnetic resonance imaging (DSC-MRI)), en een nieuwere techniek, zonder contrast vloeistof (flow-sensitive alternating inversion recovery (FAIR)). De doorbloeding met FAIR kwam grotendeels overeen met die volgens DSC-MRI, behalve in gebieden waar de doorbloeding ernstig was afgenomen; hierin werd met FAIR een lagere doorbloeding gemeten. Gebieden met een afgenomen doorbloeding werden niet alleen gevonden in het infarct zelf, maar ook in hersenweefsel om het infarct heen. Voor dit hersenweefsel bestaat het gevaar van secundaire beschadiging, die voorkomen zou kunnen worden door de juiste behandeling. In het hierboven beschreven model verbeterde de doorbloeding van hersenweefsel rond het infarct na chirurgische decompressie (hoofdstuk 5). Dit betekent dat deze behandeling secundaire schade zou kunnen voorkomen. Er was ook een verbetering van de ‘Apparent Diffusion Coefficient’ in het infarct zelf, die een maat is voor beschadiging van hersenweefsel. Bij microscopisch onderzoek bleek deze verbetering echter niet te berusten op daadwerkelijk herstel, maar waarschijnlijk op een toename van oedeem. De schade in de herseninfarcten zelf, met de bijbehorende ernstige neurologische uitval, werd door chirurgische decompressie dus niet verbeterd. De resultaten van een meta-analyse van in de literatuur beschreven voorspellers van levensbedreigend hersenoedeem bij patiënten met ACM infarcten zijn beschreven in hoofdstuk 6. Infarctgrootte blijkt de belangrijkste determinant te zijn. Helaas werd er geen klinische of radiologische variabele gevonden die levensbedreigend oedeem voorspelt in een vroeg stadium, voordat patiënten daadwerkelijk achteruit gaan. In hoofstuk 7 is een nieuwe methode beschreven om één van twee behandelingen toe te wijzen in klinische trials. De methode is gebaseerd op minimisatie, waarbij de behandeling wordt toegewezen louter op grond van de toewijzingen bij eerdere patiënten, om ongelijkheid tussen de behandelingsgroepen zoveel mogelijk te 177 Samenvatting beperken. Bij de nieuwe methode wordt een kanselement toegevoegd aan de toewijzingsbeslissing op grond van minimisatie. De grootte van deze kans hangt af van de ongelijkheid tussen de behandelingsgroepen. Met simulatietrials is aangetoond dat met deze nieuwe methode selectiebias kleiner is dan bij minimisatie en de ongelijkheid tussen de behandelingsgroepen kleiner dan bij pure randomisatie. Minder ongelijkheid tussen de behandelingsgroepen zonder grote toename van selectie bias kan in trials met weinig te includeren patiënten van belang kan zijn. In Hoofdstuk 8 staat het onderzoeksprotocol van HAMLET. HAMLET is een open gerandomiseerde klinische trial, waarin bij 112 patiënten met een ruimte-innemend supratentorieel herseninfarct het herstel na chirurgische decompressie vergeleken wordt met dat na conservatieve behandeling. Patiënten tot en met 60 jaar met een ruimte-innemend infarct in het stroomgebied van de ACM kunnen binnen 96 h na het ontstaan van de symptomen worden geïncludeerd, als de behandelend neuroloog de indruk heeft dat er achteruitgang bestaat op basis van oedeemvorming. Bij deze patiënten moeten ernstige neurologische uitval, geleidelijke achteruitgang van het bewustzijn en ruimte-innemend oedeem op de CT- of MRI-scan van de hersenen bestaan. De primaire uitkomstmaat is het functionele herstel na 1 jaar volgens de gemodificeerde Rankin Schaal (mRS). Daarnaast zal onder meer de kwaliteit van leven onderzocht worden. De eerste patiënt werd geïncludeerd in september 2002 en 1 januari 2007 waren 58 patiënten geïncludeerd. De vraag om toestemming voor deelname aan HAMLET wordt gesteld aan wettelijke vertegenwoordigers van patiënten, omdat de patiënten zelf door een gedaald bewustzijn niet wilsbekwaam zijn. De vraag om toestemming zou onacceptabel gevonden kunnen worden door wettelijke vertegenwoordigers, gezien de levensbedreigende toestand en het vermoede grote effect van chirurgische decompressie op de kans van overleven. Wettelijke vertegenwoordigers die toestemming gaven voor deelname aan HAMLET van een familielid werden geïnterviewd, met als doel te onderzoeken hoe de vraag om toestemming beleefd was en hoeveel informatie onthouden was (hoofdstuk 9). De meeste wettelijke vertegenwoordigers vonden de vraag om toestemming acceptabel. Er was na een jaar meer onderzoeksinformatie onthouden door wettelijke vertegenwoordigers die toestemming gaven voor HAMLET dan door wettelijke vertegenwoordigers die toestemming gaven voor een trial waarin een minder invasieve behandeling werd onderzocht, namelijk het Paracetamol (Acetaminophen) In Stroke (PAIS) onderzoek. In hoofdstuk 10 staan de resultaten van neuropsychologisch onderzoek van 10 patiënten met een ruimte-innemend ACM infarct een jaar na chirurgische decompressie. Naast de ernstige focale cognitieve uitvalsverschijnselen, die verwacht waren op grond van de plaats en de grootte van het herseninfarct, werd bij 8 van de 10 patiënten globaal cognitief verval vastgesteld. Bij patiënten met een infarct in de dominante hemisfeer was de taalfunctie zo slecht dat communicatie vrijwel niet mogelijk was. Patiënten met 178 Samenvatting een infarct in de dominante hemisfeer deden het bij neuropsychologisch onderzoek slechter dan patiënten met een infarct in de niet-dominante hemisfeer, ondanks gelijke uitkomst scores volgens de mRS. Voordat chirurgische decompressie wordt ingevoerd als standaard behandeling bij patiënten met een ruimte-innemend herseninfarct, moeten de resultaten van neuropsychologisch onderzoek van HAMLET worden afgewacht. Om het effect van vroege chirurgische decompressie (binnen 48 uur na ontstaan van het herseninfarct) bij patiënten met een ruimte-innemend herseninfarct eerder te kunnen beoordelen dan op basis van kleine individuele trials mogelijk zou zijn werden data van drie lopende gerandomiseerde Europese onderzoeken samengevoegd. De resultaten van de analyse van deze data staan in hoofdstuk 11. Er werden 93 patiënten geïncludeerd. Na chirurgische decompressie hadden meer patiënten een mRS≤4 (75% vs 24%; gepoolde absolute risico reductie (ARR) [95% betrouwbaarheids interval (95%CI)]: 51% [34-69]), een mRS≤3 (43% vs 21%; ARR: 23% [5-41]) en overleefden meer patiënten (78% vs 29%; ARR: 50% [33-67]) dan na conservatieve behandeling. Dit betekent ‘numbers needed to treat’ van 2 voor overleven met een mRS≤4, 4 voor overleven met een mRS≤3 en 2 voor overleven ongeacht de uitkomst. Dat houdt in dat chirurgische decompressie de mortaliteit reduceert met 50%, met een mRS≤3 bij 55% van de overlevenden. Het effect van chirurgische decompressie was consistent tussen de drie onderzoeken. De eindconclusies van het onderzoek worden bediscussieerd in hoofdstuk 12. De beslissing om chirurgische decompressie te verrichten moet hoge mate individueel genomen worden. Toch zijn gerandomiseerde onderzoeken van belang. De informatie uit het onderzoek over de kans op herstel zal voor iedere patiënt in het licht van persoonlijke ideeën over kwaliteit van leven beoordeeld moeten worden. Voortijdige onderbreking van trials moet voorkomen worden, mede omdat subgroepanalyses gewenst zijn, om te bepalen of bijvoorbeeld leeftijd, afasie, tijdstip van operatie en infarcering van andere stroomgebieden naast dat van de ACM van belang zijn voor het functionele herstel. Toekomstig onderzoek moet zich richten op prognostische factoren, om erachter te komen welke patiënten met een ACM infarct het risico lopen op levensbedreigend oedeem en welke patiënten het meest gebaat zijn bij chirurgische decompressie. 179 hoofdstuk 2 Dankwoord Dankwoord 181 Dankwoord Velen wil ik dankzeggen voor hun bijdragen aan dit proefschrift. Dr. H.B. van der Worp, beste Bart, je stond aan de basis van dit onderzoek. Je handtekening staat in ieder hoofdstuk van dit proefschrift. Ieder idee, experiment of artikel werd beter als het door jouw handen ging. Ik heb onze samenwerking plezierig en leerzaam gevonden. Je relativeringsvermogen was een steun bij frustraties. Aan de HAMLET promotie tour, in jouw mintgroene Volvo 360, bewaar ik goede herinneringen. Je buitensporige controle behoefte en je onstuitbare neiging dezelfde flauwe grappen steeds te herhalen zijn je vergeven. Prof. Dr. L.J. Kappelle, beste Jaap, jij bent een voorbeeld voor me op wetenschappelijk, klinisch-neurologisch en (niet in de laatste plaats) menselijk vlak. Bedankt voor al je hulp. Prof. Dr. A. Algra, beste Ale, ik heb grote bewondering voor de prettige manier waarop jij leermeester bent voor velen. Je methodologische bijdragen aan HAMLET en aan de gepoolde analyse waren onmisbaar. Enorm bedankt. Dr. G.J. Amelink, beste Hans, naast de nachtelijke operaties ad hoc waren je ongenuanceerde relativerende relazen onontbeerlijk. Bedankt. Prof. Dr. J. van Gijn, het is een eer door u opgeleid te zijn tot neuroloog. Ik dank u in het bijzonder voor uw bijdrage aan mijn manuscripten, waardoor geschreven staat wat bedoeld wordt. Prof. Dr. K. Nicolay, beste Klaas, bedankt voor je hartelijke ontvangst en leerzame begeleiding op de afdeling ‘experimentele in vivo NMR’ . Marrit van Buuren, bedankt voor je bijdragen aan de data verzameling en de patiënten follow up voor HAMLET, die groter zijn dan menigeen denkt. Patiënten en hun familieleden die aan HAMLET hebben deelgenomen dank ik zeer voor alle inspanningen. Prof. Dr. J. Stam, Dr. G.J. Bouma, Prof. Dr. W.P. Vandertop, Dr. G.J. Luijckx, Prof. Dr. J.J.A. Mooij, Dr. J.D.M. Metzemakers, Dr. R. van Oostenbrugge, Prof. Dr. J. Dings, Dr. J.Th.J. Tans, Dr. J.A.L. Würzer, Dr. S.F.T.M. de Bruijn, Dr. C.F.E. Hoffmann en alle andere neurologen, neurochirurgen en intensivisten uit alle deelnemende ziekenhuizen die een bijdrage hebben geleverd aan HAMLET ben ik zeer erkentelijk. Alle Nederlandse neurologen die patiënten voor deelname aan HAMLET naar één van de deelnemende 182 Dankwoord ziekenhuizen hebben verwezen en verpleegkundigen die zich voor HAMLET hebben ingezet bedank ik hiervoor. Prof. Dr. Y. van der Graaf, Dr. A.I.R. Maas en Prof. Dr. P.J. Koudstaal, bedankt voor uw inspanningen als Data Monitoring and Safety Committee. Malcolm Macleod, thank you for your efforts for HAMLET in the UK. Prof. Dr. W. Hacke, dear Werner, thank you for giving advice in the early stages of the trial and for chairing the advisory committee. The DECIMAL and DESTINY investigators and Peter Rothwell, thank you for sharing and analyzing data in a pooled analysis of decompressive surgery trials. Mijn collega’s in de kliniek bedank ik voor hun amicale collegialiteit. De vriendschap met Floor, de vele kopjes koffie en thee met Ynte en Marieke, de humoristische kleine vakanties voor en na congressen met Walter, Geert-Jan, Bart, Floor, Ynte, Marieke, en Wouter droegen in hoge mate bij aan plezier op de werkvloer. Gert van Dijk, bedankt voor je steun. De zeiltochtjes behoren tot de hoogtepunten van mijn assistententijd. Dop Bär maakte mijn eerste schreden op experimenteel onderzoeksgebied mogelijk. In het laboratorium van de afdeling ‘experimentele in vivo NMR’ werd ik met raad en daad bijgestaan door Gerard van Vliet, Wouter Veldhuis en Janneke Schepers. Bedankt! Ivonne en Esther, paranimfen en makkers vanaf de eerste dag, wat ben ik blij met jullie! Mijn ouders bedank ik voor hun onvoorwaardelijke steun en vertrouwen, mijn broer voor zijn vriendschap en mijn zus, mijn beste vriendin en ware paranimf, voor alles wat ze is en mij laat zijn. Tenslotte Erwin, bedankt dat je het mogelijk maakt ‘werk’ en ‘thuis’ probleemloos te combineren. Bedankt voor je geduld en al het goede dat wij samen delen. 183 Dankwoord 184 Curriculum Vitae Curriculum Vitae 185 Curriculum Vitae Jeannette Hofmeijer was born on December 20th, 1971. She finished secondary education in 1991 (Corderius College, Amersfoort). She studied medicine and philosophy at the Radboud University Nijmegen from 1991 to 1999. As a student, she studied neuropsychological functioning of patients with myotonic dystrophy (prof. dr. B.G.M. van Engelen). In 1999 she did internships in Tanzania at the Sengerema District Designated Hospital (dr. M. J. Voeten). After obtaining her medical degree (cum laude), she started working as a resident at the Department of Neurology of the University Medical Center Utrecht (prof. dr. J. van Gijn) in 2000. In 2001 she worked at the department of Experimental In Vivo NMR of the Utrecht University, where she investigated tissue damage and perfusion in an animal model of space-occupying cerebral infarction by means of MRI (prof. dr. K. Nicolay). From 2002 she coordinates HAMLET (Hemicraniectomy After MCA-infarction with Life-threatening Edema Trial), sponsored by the Netherlands Heart Foundation. In February 2007 she qualified as a neurologist and started as a fellow at the Intensive Care Center of the University Medical Center Utrecht (prof. dr. J. Keseçioglu). She lives together with Erwin Blezer. They have a daughter, Fee, and are expecting a second baby in June 2007. 186 Publications Publications 187 Publications Hofmeijer J, Klijn CJ, Kappelle LJ, van Huffelen AC, van Gijn J. Collateral circulation via the ophthalmic artery or leptomeningeal vessels is associated with impaired cerebral vasoreactivity in patients with symptomatic carotid artery occlusion. Cerebrovasc Dis 2002;14:22-26. Hoogendam A, Hofmeijer J, Frijns CJ, Heeringa M, Schouten T, Jansen PA. [Severe parkinsonism due to metoclopramide in a patient with polypharmacy]. Ned Tijdschr Geneeskd 2002;146:175-177. Hofmeijer J, van der Worp HB, Amelink GJ, Algra A, van Gijn J, Kappelle LJ. [Surgical decompression in space-occupying cerebral infarction; notification of a randomized trial]. Ned Tijdschr Geneeskd 2003;147:2594-2596. Hofmeijer J, van der Worp HB, Kappelle LJ. Treatment of space-occupying cerebral infarction. Crit Care Med 2003;31:617-625. Hofmeijer J, Veldhuis WB, Schepers J, Nicolay K, Kappelle LJ, Bär PR, van der Worp HB. The time course of ischemic damage and cerebral perfusion in a rat model of space-occupying cerebral infarction. Brain Res 2004;1013:74-82. Hofmeijer J, Schepers J, Veldhuis WB, Nicolay K, Kappelle LJ, Bär PR, van der Worp HB. Delayed decompressive surgery increases apparent diffusion coefficient and improves peri-infarct perfusion in rats with space-occupying cerebral infarction. Stroke 2004;35:1476-1481. Hofmeijer J, Schepers J, van der Worp HB, Kappelle LJ, Nicolay K. Perfusion MRI with FAIR and DSC-MRI. A qualitative comparison in rats with permanent middle cerebral artery occlusion. NMR Biomed 2005;18(6):390-4. Hofmeijer J, Amelink GJ, Algra A, van Gijn J, Macleod MR, Kappelle LJ, van der Worp HB. Hemicraniectomy after middle cerebral artery infarction with life-threatening Edema trial (HAMLET). Protocol for a randomised controlled trial of decompressive surgery in space-occupying hemispheric infarction. Trials 2006;7:29. Hofmeijer J, van der Worp HB, Kappelle LJ. Complications of acute ischemic stroke and their management. In: Adams-HP J, ed. Handbook of cerebrovascular diseases, 2 ed. New York: Marcel Dekker, 2006:183-204. 188 Publications Vahedi K,* Hofmeijer J,* Juettler E,* Vicaut E, George B, Algra A, Amelink GJ, Schmiedeck P, Schwab S, Rothwell PM, Bousser MG, van der Worp HB, Hacke W for the DECIMAL, DESTINY, and HAMLET investigators. Early decompressive surgery in malignant middle cerebral artery infarction: a pooled analysis of three randomized controlled trials. Lancet Neurol 2007;6:215-222. Hofmeijer J, Anema PC van der Tweel I. A new algorithm for sequential allocation of two treatments in small clinical trials. J Clin Epidemiol Under revision. Hofmeijer J, Amelink GJ, den Hertog H, Algra A, Kappelle LJ, van der Worp HB, on behalf of the HAMLET and the PAIS investigators. Appreciation of the informed consent procedure in a randomized trial of decompressive surgery for space-occupying hemispheric infarction. JNNP Under revision. Hofmeijer J, Algra A, Kappelle LJ, van der Worp HB. Predictors of life-threatening brain edema in middle cerebral artery infarction. A systematic review. Submitted. Hofmeijer J, van der Worp HB, Amelink GJ, Kappelle LJ, Nys GMS, van Zandvoort MJE. Long-term cognitive outcome after decompressive surgery for space-occupying hemispheric infarction. In preparation. 189
© Copyright 2024