HHS Public Access Author manuscript Author Manuscript

Crit Care Med. Author manuscript; available in PMC 2017 April 01. Published in final edited form as: Crit Care Med. 2016 April ; 44(4): 790–797. doi:10.1097/CCM.0000000000001553.

Rate of Peri-Hematomal Edema Expansion Predicts Outcome After Intracerebral Hemorrhage Sebastian Urday, MD, MPhil1,+, Lauren A. Beslow, MD, MSCE1,+, Feng Dai, PhD1, Fan Zhang, BS1, Thomas W.K. Battey, BS2, Anastasia Vashkevich, BA2, Alison M. Ayres, BA2, Audrey C. Leasure, BS1, Magdy H. Selim, MD, PhD4, J. Marc Simard, MD, PhD3, Jonathan Rosand, MD, MSc2, W. Taylor Kimberly, MD, PhD2, and Kevin N. Sheth, MD1

Author Manuscript

1Department

of Neurology, Yale University School of Medicine, 333 Cedar Street, New Haven, CT

06510, USA 2Department

of Neurology, Massachusetts General Hospital, 55 Fruit Street, Boston, MA 02114,

USA 3Department

of Neurosurgery, University of Maryland School of Medicine, 655 West Baltimore Street, Baltimore, MD 21201-1559, USA

4Department

of Neurology, Beth Israel Deaconess Medical Center, 330 Brookline Avenue, Boston, MA 02215, USA

Abstract Author Manuscript

Objective—Intracerebral hemorrhage (ICH) is a devastating disorder with no current treatment. Whether peri-hematomal edema (PHE) is an independent predictor of neurologic outcome is controversial. We sought to determine whether PHE expansion rate predicts outcome after ICH. Design—Retrospective cohort study Setting—Tertiary medical center Patients—139 consecutive supratentorial spontaneous ICH patients >18 years admitted between 2000–2013. Interventions—None

Author Manuscript

Measurements and Main Results—ICH, intraventricular hemorrhage (IVH), and PHE volumes were measured from computed tomography (CT) scans obtained at presentation, 24 hours, and 72 hours post-ICH. PHE expansion rate was the difference between initial and followup PHE volumes divided by the time interval. Logistic regression was performed to evaluate the relationship between 1) PHE expansion rate at 24 hours and 90-day mortality and 2) PHE *

Address for reprints: Reprints will not be ordered, Corresponding author. Sebastian Urday, MD, MPhil, Division of Neurocritical Care and Emergency Neurology, Department of Neurology, Yale Medical School, 15 York St, LCI 1003, New Haven, CT 06510. Phone: 203 737 8051, Fax: (203) 785-4580, [email protected]. +These two authors contributed equally to the paper. Institution where work was performed: Yale University School of Medicine Copyright form disclosures: The other authors declare no competing interests. The remaining authors have disclosed that they do not have any potential conflicts of interest.

Urday et al.

Page 2

Author Manuscript

expansion rate at 24 hours and 90-day modified Rankin Scale score (mRS). PHE expansion rate between admission and 24-hours post-ICH was a significant predictor of 90-day mortality (OR 2.97, 95%CI 1.48–5.99, p=0.002). This association persisted after adjusting for all components of the ICH score (OR 2.21, 95% CI 1.05–4.64, p=0.04). Similarly, higher 24-hour PHE expansion rate was associated with poorer mRS in an ordinal shift analysis (OR 2.40, 95% CI 1.37–4.21, p=. 002), even after adjustment for all ICH score components (OR 2.07, 95% CI 1.12–3.83, p=0.02). Conclusions—Faster PHE expansion rate 24 hours post-ICH is associated with worse outcome. PHE may represent an attractive translational target for secondary injury after ICH. Keywords cerebral hemorrhage; brain edema

Author Manuscript

INTRODUCTION Intracerebral hemorrhage (ICH) accounts for 10–15% of all strokes and is the deadliest stroke subtype. (1) A better understanding of ICH pathophysiology has led to interest in therapies that ameliorate secondary injury such as peri-hematomal edema (PHE). (2–4)

Author Manuscript

PHE results from peri-hemorrhagic inflammation, the toxicity of blood breakdown products, and other secondary processes. (4, 5) Therefore, PHE may cause neurological deterioration due to both biological injury (direct neurotoxicity, perturbations to water/solute homeostasis) and to additional mass effect beyond that of the hematoma. (4, 5) In animal models of ICH, PHE is consistently associated with worse neurological outcome. (6) Currently, however, the relationship between PHE and outcome in ICH patients is controversial, since studies examining this question have shown conflicting results. (7, 8) This critical gap in knowledge severely impedes translation of novel therapies for ICH. It is widely appreciated that the rate at which a mass expands is critical in determining neurological injury. For example, tissue injury and displacement in a patient with a rapidly evolving acute subdural hematoma often leads to coma and death, whereas a patient with a brain tumor that has developed gradually may have few symptoms. Our aim was to understand the impact of edema formation over time on outcome after ICH. The main hypothesis of the study was that an increased rate of PHE expansion at 24 hours is independently associated with increased mortality and worse functional outcome. We also explored the potential association between PHE formation at 72 hours and functional outcome.

Author Manuscript

METHODS Study Design Subjects were retrospectively identified from an Institutional Review Board-approved prospective cohort study of ICH between 2000 and 2013 at Massachusetts General Hospital. Written informed consent was obtained from participants or their surrogates.

Crit Care Med. Author manuscript; available in PMC 2017 April 01.

Urday et al.

Page 3

Author Manuscript

Eligible subjects for the prospective cohort (n = 497) were aged over 18 years and had a diagnosis of primary spontaneous ICH with a known time of ICH onset. (9) When an exact time of onset could not be determined, the time when the subject was last seen normal was substituted for the time of symptom onset. Subjects were admitted within the first 48 hours after symptom onset. Clinical Data Demographics and clinical characteristics were obtained by interviewing patients (or their families or surrogates) and reviewing hospital records. Clinical outcomes were 90-day mortality (assessed by telephone by trained study staff and supplemented by regular surveillance of the Social Security Death Index) and 90-day modified Rankin Scale (mRS) score (scale 0–6). (10) When the latter was not obtainable (n = 55), discharge mRS was used.

Author Manuscript

Primary Analysis

Author Manuscript

We created a primary analysis population (n = 139) by excluding infratentorial hemorrhages (n = 63), primary/isolated intraventricular hemorrhages (n = 35), and subjects who underwent subsequent surgery or ventriculostomy placement (n = 198) (Figure 1). Our goal was to exclude confounding variables that could mask the effects of PHE. We additionally excluded warfarin-related hemorrhages (n = 94) because the coagulation pathway may be involved in PHE formation. (11) Of 139 subjects, 110 had computed tomography (CT) scans on admission and ~24 hours post-ICH, 58 had scans on admission and ~72 hours post-ICH, and 71 had scans ~72 hours post-ICH (Table 1). We examined the following PHE variables: absolute PHE, relative PHE (absolute PHE/ICH volume) and PHE expansion rate (calculated as the difference between the initial and the follow-up PHE volumes, divided by the interval of time between the two CT scans). Specifically, we examined PHE expansion rate between admission and 24 hours post-ICH (primary analyses), PHE expansion rate between admission and 72 hours post-ICH, and absolute and relative PHE at 72 hours post-ICH. Imaging Analysis

Author Manuscript

ICH, PHE, and intraventricular hemorrhage (IVH) volumes were determined by a single rater (SU) from CT scans obtained closest to the relevant time-points, using Analyze 11.0 (AnalyzeDirect, Overland Park, KS, USA), as previously described. (12) Briefly, boundaries of the ICH, PHE, and IVH when present were outlined using the edge-detection tool and were adjusted after inspection of the volumes in each of the three orthogonal planes. PHE was defined as areas of hypodensity adjacent to the hemorrhage with the most hypodense region immediately adjacent to the hemorrhage. Additionally, the region of PHE was more hypodense than the same area in the contralateral hemisphere (Figure 2). We previously established excellent interrater reliability for CT-based PHE measurements in 20 subjects from this cohort and determined that measurements were comparable to those obtained on MRI. (12) A second rater (AL) performed PHE measurements 10 additional subjects. The intraclass correlation coefficient for the PHE measurements for these 10 subjects was 0.84 (95% CI 0.66–1.00, p=0.003) which indicates excellent interrater reliability.

Crit Care Med. Author manuscript; available in PMC 2017 April 01.

Urday et al.

Page 4

Statistical Analysis

Author Manuscript

Continuous variables were summarized using means [standard deviations (SDs)] or medians [interquartile ranges (IQRs)] when appropriate. Discrete variables were summarized using counts (percentages). Poor functional outcome was defined as an mRS score > 2. (13)

Author Manuscript

We examined associations between PHE variables and clinical outcomes in logistic regression models (binary for mortality and poor versus good outcome and ordinal for raw mRS score which assumed a natural ordinal level between each score from 0 to 6). We adjusted for known predictors of outcome following ICH in the following models: 1) All components of the ICH score (14) (age, ICH volume, IVH volume, GCS score, each as a continuous variable); 2) various definitions of hematoma expansion (HE) used in the literature (any HE, HE > 6 mL, > 12.5 mL, > 33% of initial hematoma volume, each as a dichotomous variable); 3) time from onset to first CT scan; and 4) do-not-resuscitate (DNR) status. DNR was defined as any subject who had a DNR order placed during the hospitalization; only one subject had a DNR order prior to the hospitalization for ICH. All the statistical analyses were performed using SAS version 9.3 (SAS institute, Cary, NC). P-values ≤ 0.05 were considered statistically significant.

RESULTS Characteristics of the 139 subjects in the primary analysis cohort, those with both admission and 24 hours post-ICH scans (n = 110), those with admission and 72 hours post-ICH scans (n = 58), and those with 72 hours post-ICH scans (n = 71) are shown in Table 1.

Author Manuscript

PHE expansion rate at 24 hours and absolute PHE expansion were weakly correlated to absolute hemorrhage expansion at 24 hours. (Spearman rank correlation coefficient = 0.22 and 0.34, respectively). Subjects with discharge mRS scores (n=55) were younger and had smaller baseline ICH and PHE volumes than subjects who had 90-day mRS scores (n=84) [67.1 years versus 75.6 years, p=0.01; 12.4 mL versus 23.4 mL, p=0.0004; 9.9 mL versus 20.2 mL, p=0.002; Wilcoxon rank-sum]. Among the 110 subjects in whom PHE expansion rate at 24 hours was calculable, median PHE expansion rate was not statistically different between those with discharge versus 90-day mRS scores. PHE expansion rate between admission and 24 hours post-ICH

Author Manuscript

Subjects who died had an average PHE expansion rate of 0.74 mL/h (SE 0.2) compared to 0.17 mL/h (SE 0.1) in survivors (p = 0.0002) (Figure 3A). There was a strong independent association between PHE expansion rate and 90-day mortality (Table 2). Similarly, PHE expansion rate was an independent predictor of higher (worse) mRS score (Table 2). The association for both mortality and functional outcome persisted even after adjustment for baseline ICH severity, as captured in the ICH score (Table 2) PHE expansion rate between admission and 72 hours post-ICH At 72 hours, an exploratory analysis was performed that evaluated the relationship between PHE expansion rate and functional outcome. Using a binary outcome, there was an independent association between PHE expansion rate and poor functional outcome (Table Crit Care Med. Author manuscript; available in PMC 2017 April 01.

Urday et al.

Page 5

Author Manuscript

3). Subjects with a poor functional outcome had an average PHE expansion rate of 0.22 mL/h (SE 0.05) compared to 0.02 mL/h (SE 0.04) in functionally-independent subjects (p = 0.07) (Figure 3B). The relationship was not statistically significant after adjustment for components of the ICH score in the ordinal analysis of mRS. PHE volume at 72 hours post-ICH In univariable analysis, absolute PHE volume was significantly associated with 90-day mortality (Table 4). Similarly, absolute PHE was an independent predictor of higher (worse) mRSscore in unadjusted analysis and when adjusting for time to first scan or DNR status (Table 4). There was no significant association between relative PHE and clinical outcomes (data not shown).

Author Manuscript

DISCUSSION

Author Manuscript

There is robust evidence that secondary brain injury, in the form of PHE, leads to worse outcome in experimental models of ICH. (15, 16) However, whether PHE is an independent predictor of neurologic outcome in human ICH remains controversial, since studies have produced contradictory results. (7, 8, 17) Here we have shown that the rate of PHE expansion predicts outcome after ICH. To our knowledge, this index of edema formation has not been investigated previously. (4) Prior studies have traditionally focused on absolute PHE, relative PHE (absolute PHE/ICH volume), and absolute and relative PHE growth. (7, 8) The main findings of our study are that 1) the rate of PHE expansion between admission and 24 hours post-ICH predicts 90-day mortality, and 2) the rate of PHE expansion between admission and 24 hours post-ICH predicts poor functional outcome, after controlling for known predictors of outcome after ICH.

Author Manuscript

We investigated the rate of PHE expansion because there are other examples in clinical neurology in which the rate at which a mass expands influences patient outcome. Romero et al. showed that the rate of ICH expansion is an independent risk factor for mortality. (18) Mechanistically, PHE may cause neurological deterioration by exerting both biological injury (e.g. perturbations to water/solute homeostasis) and brain-tissue shifts that compress vital structures. (4, 19) Our results suggest that some studies may not have found an association between PHE and outcome because this parameter was not explored. (4) In support of this, Arima et al. conducted an analysis of 270 patients and showed that absolute growth of edema between admission and 72 hours post-ICH did not independently predict poor outcome (defined as death or dependency at 90 days). (8) However, the investigators subsequently showed that absolute edema growth does independently predict poor outcome in an analysis of 1138 patients. (17) In view of our results, the rate of PHE expansion may be a more robust parameter in ICH studies, capable of discerning the relationship between PHE and clinical outcome in smaller datasets. The contribution of PHE to patient outcome may depend on the rate of expansion, with PHE that expands more rapidly producing more stress on the system than PHE that accumulates gradually. (20) We found that acutely (i.e. up to 24 hours after the ictus), rapid PHE

Crit Care Med. Author manuscript; available in PMC 2017 April 01.

Urday et al.

Page 6

Author Manuscript

expansion predicts death. In this analysis, we observed a consistent relationship between PHE rate and outcome at 24 hours but not at 72 hours; however, the number of follow-up scans available at the 72 hour time were limited, and we may have been underpowered to detect an association. The optimal time point for PHE assessment must be further investigated in future studies since this may have implications for potential therapeutic time windows and clinical trial endpoint ascertainment.

Author Manuscript

Importantly, different biological mechanisms underlie PHE formation during these time frames. (4) Acutely, the blood-brain barrier (BBB) is still intact and ionic edema accumulates due to the transcapillary flux of electrolytes and water from the intravascular to the interstitial compartment. (4) The inflammatory response to ICH then leads to the progressive breakdown of the BBB, resulting in vasogenic edema. (4) Peri-hematomal inflammation peaks by two days after ICH. (21) As has been previously described, edema formation after ICH is likely a multistage process in terms of the pathophysiology. (4) The principle stages of edema formation include ionic edema and progressive vasogenic edema, which includes clot retraction. (4) The current analysis captures the potential effect of edema across all stages but does not identify the relative contributions of rate at each individual stage. Prospective studies should use advanced imaging techniques that provide insights regarding the source of edema formation (ionic versus vasogenic versus cytotoxic).

Author Manuscript

The findings that a faster PHE expansion rate between admission and 24 hours post-ICH is detrimental is significant from a translational perspective, because it provides a relatively wide window for therapeutic intervention. Most ICH clinical trials conducted to date have focused on reducing hemorrhage expansion, since it is strongly associated with poor outcome. (22) However, most hemorrhage expansion occurs within the first 24 hours after the ictus, limiting the therapeutic window. (23) Targeting PHE post-ICH could be a valuable adjunct therapeutic approach to improve clinical outcome in a larger pool of patients. We confirmed the findings of a previous investigation that showed that absolute PHE volume at 72 hours post-ICH was independently associated with worse clinical outcome in an ordinal shift analysis of the mRS. (24) Additionally, we showed an independent association between absolute PHE volume at 72 hours post-ICH and mortality. The small sample size in our study likely limits our power to detect an association with poor functional outcome (mRS 3–6).

Author Manuscript

Our results are particularly relevant now that a better understanding of the pathophysiology of PHE has led to the emergence of potential molecular targets and therapies to ameliorate edema. (4) For example, an early-phase clinical trial showed that an anti-inflammatory agent, fingolimod, given for 72 hours after ICH onset, reduced the progression of PHE and was associated with an increased likelihood of functional independence at 90 days compared with controls. (2) In addition, there is preliminary evidence that the SUR1-TRPM4 (sulfonylurea receptor 1 – transient receptor potential M4 cation) channel is up-regulated after human ICH. (4) This channel contributes to edema formation in various animal models of acute brain injury (e.g. cerebral ischemia) and it is inhibited glyburide, a secondgeneration sulfonylurea. (25)

Crit Care Med. Author manuscript; available in PMC 2017 April 01.

Urday et al.

Page 7

Author Manuscript

A distinctive feature of our study is the selection of an ICH population to evaluate the clinical impact of PHE. We believe that our primary analysis population represents the best cohort in which to reconcile conflicting reports in the literature, because it excludes patients in which the independent contribution of PHE on outcome may be masked due to uniformly high mortality. (4) For example, we excluded patients who underwent ventriculostomy placement because it could represent development of significant IVH leading to acute obstructive hydrocephalus, which could be a confounder. A crucial next step is to determine whether the observed associations are generalizable to other ICH populations (e.g. warfarinassociated hemorrhages), and if there are variations by topography (e.g. basal ganglia versus cortical ICHs) or underlying etiology (e.g. hypertensive, amyloid angiopathy). (4)

Author Manuscript Author Manuscript

Our study has a number of limitations. Given the retrospective design, our imaging analysis was limited to scans available at admission, and closest to 24 and 72 hours after ICH. While this time frame includes the contribution of key pathways implicated in PHE formation (e.g. inflammatory pathways), there is evidence that edema continues to accumulate for 2–3 weeks after ICH. (26, 27) In particular, hemoglobin-degradation products promote delayed vasogenic edema starting three days after ICH. (4, 28) Also, CT scans were obtained at different times in all patients, although we did include models adjusting for the time to scan. Lack of a pre-specified imaging protocol also means there is a possibility of selection bias (e.g. subjects that had CTs at 72 hours post-ICH might have been deteriorating). Additionally, while many retrospective and prospective stroke studies use discharge outcome as the 90 day outcome when 90 day outcome is not available (29), this method may misclassify subjects because a particular subject’s functional status could either improve or get worse. This limitation is further highlighted by the fact that subjects in whom only discharge mRS was available were younger and had smaller hemorrhages with less PHE. Prospective studies, with sufficient power to examine multivariable models and that have careful follow-up will be needed to replicate our results, particularly with regard to functional outcome.

Author Manuscript

Although our analysis adjusted for key factors implicated in ICH outcome (i.e. the ICH score, various definitions of hemorrhage expansion, DNR orders), important variables were not available due to the retrospective nature of the study. For example, we lacked data on use of osmotherapy, which may modify PHE volume and affect patient outcome. (30, 31,) However, a previous study found that a small number of ICH patients develop high intracranial pressure, suggesting osmotic agents may not be used as widely as in the management of other neurocritical care conditions. (2) DNR status is a known mediator of a potential “self-fulfilling prophecy” in ICH outcomes. In this analysis, even when adjusting for DNR, rate of edema was still associated with poor outcome. Prospective studies will need to perform a survivors’ only analysis of functional outcome and record administration of osmotic therapies in order to understand any potential effects further.

CONCLUSIONS In conclusion, this study provides evidence that the rate of PHE expansion independently predicts clinical outcomes after ICH. PHE may represent an attractive translational target for

Crit Care Med. Author manuscript; available in PMC 2017 April 01.

Urday et al.

Page 8

Author Manuscript

this disorder. Future prospective studies with neuroimaging acquired systematically at various time-points post-ICH are warranted to validate these findings.

ACKNOWLEDGEMENTS S.U. is supported by the Leon Rosenberg, MD Medical Student Research Fund in Genetics (Yale University School of Medicine) and a 2014 Student Scholarship in Cerebrovascular Disease and Stroke (American Heart Association Stroke Council). L.A.B. is supported by the National Institute of Neurological Disorders and Stroke (NINDS; K12NS049453). W.T.K is supported by NINDS K23NS076597. J.M.S. is supported by grants from the Department of Veterans Affairs (Baltimore; BX001629), the NINDS (NS060801, NS061808), and the National Heart, Lung and Blood Institute (HL082517).

Author Manuscript

Dr. Urday received support for article research from the National Institutes of Health (NIH). He received funding from Leon Rosenberg, MD Medical Student Research Fund in Genetics (Yale University School of Medicine) and a 2014 Student Scholarship in Cerebrovascular Disease and Stroke (American Heart Association Stroke Council). He disclosed that W.T.K., L.A.B. and K.N.S. are investigators in GAMES-RP, a phase II study of an investigational compound aimed at preventing swelling after large stroke. J.M.S. holds a US patent (7,285,574, Methods for treating neural cell swelling). J.R. has a consulting relationship with Boehringer Ingelheim. Dr. Dai received support for article research from the NIH. His institution received funding from the NIH. Dr. Zhang received support for article research and received funding from the NIH. Dr. Vashkevich received support for article research from the NIH. Dr. Ayres received support for article research from the NIH. Dr. Selim received support for article research from the NIH. Dr. Rosand received support for article research from the NIH. His institution received funding from the NIH. Dr. Kimberly received support for article research from the NIH. Her institution received funding from the American Heart Association; NIH; and Remedy Pharmaceuticals, Inc.

REFERENCES

Author Manuscript Author Manuscript

1. van Asch CJ, Luitse MJ, Rinkel GJ, van der Tweel I, Algra A, Klijn CJ. Incidence, case fatality, and functional outcome of intracerebral haemorrhage over time, according to age, sex, and ethnic origin: A systematic review and meta-analysis. Lancet Neurol. 2010; 9:167–176. [PubMed: 20056489] 2. Fu Y, Hao J, Zhang N, Ren L, Sun N, Li YJ, Yan Y, Huang D, Yu C, Shi FD. Fingolimod for the treatment of intracerebral hemorrhage: A 2-arm proof-of-concept study. JAMA Neurol. 2014; 71:1092–1101. [PubMed: 25003359] 3. Sheth KN, Rosand J. Targeting the immune system in intracerebral hemorrhage. JAMA Neurol. 2014; 71:1083–1084. [PubMed: 25004375] 4. Urday S, Kimberly WT, Beslow LA, Vortmeyer AO, Selim MH, Rosand J, Simard JM, Sheth KN. Targeting secondary injury in intracerebral haemorrhage-perihaematomal oedema. Nat Rev Neurol. 2015; 11:111–122. [PubMed: 25623787] 5. Keep RF, Hua Y, Xi G. Intracerebral haemorrhage: Mechanisms of injury and therapeutic targets. Lancet Neurol. 2012; 11:720–731. [PubMed: 22698888] 6. Nakamura T, Keep RF, Hua Y, Schallert T, Hoff JT, Xi G. Deferoxamine-induced attenuation of brain edema and neurological deficits in a rat model of intracerebral hemorrhage. J Neurosurg. 2004; 100:672–678. [PubMed: 15070122] 7. Appelboom G, Bruce SS, Hickman ZL, Zacharia BE, Carpenter AM, Vaughan KA, Duren A, Hwang RY, Piazza M, Lee K, Claassen J, Mayer S, Badjatia N, Connolly ES Jr. Volume-dependent effect of perihaematomal oedema on outcome for spontaneous intracerebral haemorrhages. J Neurol Neurosurg Psychiatry. 2013; 84:488–493. [PubMed: 23345281] 8. Arima H, Wang JG, Huang Y, Heeley E, Skulina C, Parsons MW, Peng B, Li Q, Su S, Tao QL, Li YC, Jiang JD, Tai LW, Zhang JL, Xu E, Cheng Y, Morgenstern LB, Chalmers J, Anderson CS. Investigators I. Significance of perihematomal edema in acute intracerebral hemorrhage: The interact trial. Neurology. 2009; 73:1963–1968. [PubMed: 19996072] 9. Falcone GJ, Biffi A, Brouwers HB, Anderson CD, Battey TW, Ayres AM, Vashkevich A, Schwab K, Rost NS, Goldstein JN, Viswanathan A, Greenberg SM, Rosand J. Predictors of hematoma volume in deep and lobar supratentorial intracerebral hemorrhage. JAMA Neurol. 2013; 70:988–994. [PubMed: 23733000]

Crit Care Med. Author manuscript; available in PMC 2017 April 01.

Urday et al.

Page 9

Author Manuscript Author Manuscript Author Manuscript Author Manuscript

10. Wojcik NC, Huebner WW, Jorgensen G. Strategies for using the national death index and the social security administration for death ascertainment in large occupational cohort mortality studies. Am J Epidemiol. 2010; 172:469–477. [PubMed: 20643697] 11. Levine JM, Snider R, Finkelstein D, Gurol ME, Chanderraj R, Smith EE, Greenberg SM, Rosand J. Early edema in warfarin-related intracerebral hemorrhage. Neurocrit Care. 2007; 7:58–63. [PubMed: 17657657] 12. Urday S, Beslow LA, Goldstein DW, Vashkevich A, Ayres AM, Battey TW, Selim MH, Kimberly WT, Rosand J, Sheth KN. Measurement of perihematomal edema in intracerebral hemorrhage. Stroke. 2015; 46:1116–1119. [PubMed: 25721012] 13. Anderson CS, Heeley E, Huang Y, Wang J, Stapf C, Delcourt C, Lindley R, Robinson T, Lavados P, Neal B, Hata J, Arima H, Parsons M, Li Y, Wang J, Heritier S, Li Q, Woodward M, Simes RJ, Davis SM, Chalmers J. Investigators I. Rapid blood-pressure lowering in patients with acute intracerebral hemorrhage. N Engl J Med. 2013; 368:2355–2365. [PubMed: 23713578] 14. Hemphill JC 3rd, Bonovich DC, Besmertis L, Manley GT, Johnston SC. The ich score: A simple, reliable grading scale for intracerebral hemorrhage. Stroke. 2001; 32:891–897. [PubMed: 11283388] 15. Wang J, Tsirka SE. Neuroprotection by inhibition of matrix metalloproteinases in a mouse model of intracerebral haemorrhage. Brain. 2005; 128:1622–1633. [PubMed: 15800021] 16. Li G, Fan RM, Chen JL, Wang CM, Zeng YC, Han C, Jiao S, Xia XP, Chen W, Yao ST. Neuroprotective effects of argatroban and c5a receptor antagonist (pmx53) following intracerebral haemorrhage. Clin Exp Immunol. 2014; 175:285–295. [PubMed: 24117111] 17. Yang J, Arima H, Wu G, Heeley E, Delcourt C, Zhou J, Chen G, Wang X, Zhang S, Yu S, Chalmers J, Anderson CS. Investigators I. Prognostic significance of perihematomal edema in acute intracerebral hemorrhage: Pooled analysis from the intensive blood pressure reduction in acute cerebral hemorrhage trial studies. Stroke. 2015; 46:1009–1013. [PubMed: 25712944] 18. Romero JM, Heit JJ, Delgado Almandoz JE, Goldstein JN, Lu J, Halpern E, Greenberg SM, Rosand J, Gonzalez RG. Spot sign score predicts rapid bleeding in spontaneous intracerebral hemorrhage. Emerg Radiol. 2012; 19:195–202. [PubMed: 22271362] 19. Ropper AH. Lateral displacement of the brain and level of consciousness in patients with an acute hemispheral mass. N Engl J Med. 1986; 314:953–958. [PubMed: 3960059] 20. Caplan, LR. Caplan's stroke : A clinical approach. Philadelphia: Saunders/Elsevier; 2009. 21. Wu H, Zhang Z, Li Y, Zhao R, Li H, Song Y, Qi J, Wang J. Time course of upregulation of inflammatory mediators in the hemorrhagic brain in rats: Correlation with brain edema. Neurochem Int. 2010; 57:248–253. [PubMed: 20541575] 22. Brouwers HB, Greenberg SM. Hematoma expansion following acute intracerebral hemorrhage. Cerebrovasc Dis. 2013; 35:195–201. [PubMed: 23466430] 23. Xi GH, Keep RF, Hoff JT. Mechanisms of brain injury after intracerebral haemorrhage. Lancet Neurology. 2006; 5:53–63. [PubMed: 16361023] 24. Sansing LH, Messe SR, Cucchiara BL, Lyden PD, Kasner SE. Anti-adrenergic medications and edema development after intracerebral hemorrhage. Neurocrit Care. 2011; 14:395–400. [PubMed: 21264527] 25. Simard JM, Woo SK, Schwartzbauer GT, Gerzanich V. Sulfonylurea receptor 1 in central nervous system injury: A focused review. J Cereb Blood Flow Metab. 2012; 32:1699–1717. [PubMed: 22714048] 26. Venkatasubramanian C, Mlynash M, Finley-Caulfield A, Eyngorn I, Kalimuthu R, Snider RW, Wijman CA. Natural history of perihematomal edema after intracerebral hemorrhage measured by serial magnetic resonance imaging. Stroke. 2011; 42:73–80. [PubMed: 21164136] 27. Zazulia AR, Diringer MN, Derdeyn CP, Powers WJ. Progression of mass effect after intracerebral hemorrhage. Stroke. 1999; 30:1167–1173. [PubMed: 10356094] 28. Mehdiratta M, Kumar S, Hackney D, Schlaug G, Selim M. Association between serum ferritin level and perihematoma edema volume in patients with spontaneous intracerebral hemorrhage. Stroke. 2008; 39:1165–1170. [PubMed: 18292378]

Crit Care Med. Author manuscript; available in PMC 2017 April 01.

Urday et al.

Page 10

Author Manuscript

29. Qureshi AI, Chaudhry SA, Sapkota BL, Rodriquez GJ, Suri MF. Discharge destination as a surrogate for modified Rankin Scale defined outcomes at 3- and 12-months poststroke among survivors. Arch Phys Med Rehabil. 2012; 93:1408–1413. [PubMed: 22446290] 30. Wagner I, Hauer EM, Staykov D, Volbers B, Dorfler A, Schwab S, Bardutzky J. Effects of continuous hypertonic saline infusion on perihemorrhagic edema evolution. Stroke. 2011; 42:1540–1545. [PubMed: 21512173] 31. Ryu JH, Walcott BP, Kahle KT, Sheth SA, Peterson RT, Nahed BV, Coumans JV, Simard JM. Induced and sustained hypernatremia for the prevention and treatment of cerebral edema following brain injury. Neurocrit Care. 2013; 19:222–231. [PubMed: 23468135] 32. Kamel H, Hemphill JC 3rd. Characteristics and sequelae of intracranial hypertension after intracerebral hemorrhage. Neurocrit Care. 2012; 17:172–176. [PubMed: 22833445]

Author Manuscript Author Manuscript Author Manuscript Crit Care Med. Author manuscript; available in PMC 2017 April 01.

Urday et al.

Page 11

Author Manuscript Author Manuscript Figure 1.

Flow chart of primary analysis population selection. Abbreviation: ICH, intracerebral hemorrhage.

Author Manuscript Author Manuscript Crit Care Med. Author manuscript; available in PMC 2017 April 01.

Urday et al.

Page 12

Author Manuscript Author Manuscript

Figure 2.

Representative Peri-hematomal Edema Measurement. A) Left thalamic hemorrhage. B) ICH (red) and PHE (cyan) outlined with an edge detection method. (12)

Author Manuscript Author Manuscript Crit Care Med. Author manuscript; available in PMC 2017 April 01.

Urday et al.

Page 13

Author Manuscript Author Manuscript Author Manuscript Author Manuscript

Figure 3.

Average peri-hematomal edema expansion rates. A) Comparison of the average rate of perihematomal edema expansion (mL/h) between admission and 24 hours post-intracerebral hemorrhage of subjects who died and subjects who survived. B) Comparison of the average rate of peri-hematomal edema expansion (mL/h) between admission and 72 hours postintracerebral hemorrhage of subjects with a poor functional outcome and functionally

Crit Care Med. Author manuscript; available in PMC 2017 April 01.

Urday et al.

Page 14

Author Manuscript

independent subjects. Abbreviations: mRS, modified Rankin Scale; PHE, peri-hematomal edema

Author Manuscript Author Manuscript Author Manuscript Crit Care Med. Author manuscript; available in PMC 2017 April 01.

Urday et al.

Page 15

Table 1

Author Manuscript

Subject Characteristics Primary Analysis Cohort (N = 139)

Admission and 24 hours postICH scans (n = 110)

Admission and 72 hours post-ICH scans (n = 58)

72 hours postICH scan (n = 71)

71.1 (12.8)

71.8 (12.7)

72.5 (12.6)

72.6 (12.5)

81 (58.3)

63 (57.3)

29 (50)

39 (54.9)

117 (84.2)

94 (85.5)

46 (79.3)

59 (83.1)

10 (7.2)

7 (6.4)

6 (10.3)

6 (8.5)

Hypertension

107 (77)

88 (80)

47 (81)

55 (77.5)

Diabetes

32 (23)

26 (23.6)

13 (22.4)

14 (19.7)

Anti-hypertensive

86 (61.9)

72 (65.5)

39 (67.2)

41 (57.7)

Statin

44 (31.7)

37 (33.6)

17 (29.3)

20 (28.2)

179 (34.3)

182 (35.2)

182 (33.9)

180 (33.8)

Age, y, mean (SD) Male, n (%) Race, n (%) Caucasian African American Medical History, n (%)

Author Manuscript

Medication History, n (%)

Clinical Features Systolic BP, mm Hg, mean (SD) GCS score, median (IQR)

14 (11–15)

14 (10–15)

15 (10.8–15)

15 (11.8–15)

130 (112–154)

132 (113–155)

130 (114–161)

131 (109–154)

30 (21.6)

30 (27.3)

15 (25.9)

14 (19.7)

Lobar

69 (49.6)

51 (46.4)

26 (44.8)

34 (47.9)

Deep

70 (50.4)

59 (53.6)

32 (55.2)

37 (52.1)

Intraventricular extension

52 (37.4)

50 (45.5)

31 (53.4)

37 (52.1)

Admission ICH

19 (9.0–40.7)

16.2 (7.9–50.2)

NA

Follow-up ICH

19.9 (8.9–47.9)

16.2 (7.6–46.8)

15.5 (7.4–45.4)

Admission PHE

14.1 (7.3–34.3)

12.8 (6.9–36.6)

NA

Follow-up PHE

18.4 (8.3–39.5)

21.2 (11.8–54.4)

21.2 (11.3–53.2)

NA

NA

1.6 (0.8)

Onset to admission scan

3.3 (1.8–6.1)

3 (1.6–5.7)

NA

Onset to follow-up scan

22.5 (15.5–27.1)

66.9 (60.3–75.8)

74.3 (63.6–78.6)

Glucose, median (IQR) DNR, n (%) Location of Hematoma, n (%)

Author Manuscript

Volume, mL, median (IQR)

Follow-up relative PHE, mean (SD) Time to CT scan, h, median (IQR)

Clinical outcomes, n (%)

Author Manuscript

90-day mortality

30 (21.6)

29 (26.4)

16 (27.6)

15 (21.1)

90-day mRS > 2

107 (77)

91 (82.7)

48 (82.8)

53 (74.6)

BP indicates blood pressure; DNR, do-not-resuscitate order; GCS, Glasgow Coma Scale; ICH, intracerebral hemorrhage; IQR, interquartile range; mRS, modified Rankin Scale; PHE, peri-hematomal edema

Crit Care Med. Author manuscript; available in PMC 2017 April 01.

Author Manuscript

Author Manuscript

Author Manuscript 2.98 (1.40–6.33) 2.78 (1.38–5.61) 2.59 (1.33–5.06)

PHE rate, HE > 6 mL adjusted

PHE rate, HE >12.5 mL adjusted

PHE rate, HE > 33% adjusted

PHE rate, DNR status adjusted

3.06 (1.32–7.09)

0.009

0.003

0.005

0.004

0.04

0.002

0.003

2.24 (0.69–7.28)

2.63 (0.82–8.33)

3.33 (0.91–12.5)

2.78 (0.82–9.09)

2.70 (0.81–9.09)

2.50 (0.80–7.69)

3.13 (0.68–14.3)

2.63 (0.84–8.33)

2.78 (0.89–8.33)

2.86 (0.64–12.5)

2.56 (0.83–8.33)

2.63 (0.85–8.33)

Odd Ratio (95% CI)

0.18

0.10

0.07

0.10

0.10

0.12

0.14

0.10

0.08

0.17

0.10

0.09

P-value

2.03 (1.10–3.74)

2.33 (1.33–4.08)

2.20 (1.26–3.85)

2.29 (1.30–4.05)

2.33 (1.30–4.20)

2.34 (1.34–4.10)

2.07 (1.12–3.83)

2.36 (1.36–4.11)

2.59 (1.43–4.68)

1.94 (1.06–3.55)

2.32 (1.33–4.04)

2.40 (1.37–4.21)

Odd Ratio (95% CI)

0.02

0.003

0.006

0.004

0.005

0.003

0.02

0.002

0.002

0.03

0.003

0.002

P-value

Higher (worse) mRS score

Ordinal Outcome

DNR indicates do-not-resuscitate order; GCS, Glasgow Coma Scale; HE, hemorrhage expansion; ICH, intracerebral hemorrhage; IVH, intraventricular hemorrhage; mRS, modified Rankin Scale; OR, odds ratio; PHE, peri-hematomal edema

ICH score components: Age, ICH volume, IVH volume, GCS

a

2.97 (1.46–6.02)

PHE rate, Any HE adjusted

2.91 (1.44–5.87)

2.21 (1.05–4.64)

PHE rate, All ICH score componentsa adjusted

scan adjusted

3.05 (1.51–6.15)

PHE rate, GCS adjusted

PHE rate, Time to

0.005

3.10 (1.48–6.49)

PHE rate, IVH vol adjusted

1st

0.003

2.23 (1.09–4.57)

0.03

0.003

2.86 (1.44–5.69)

PHE rate, Age adjusted

PHE rate, ICH vol adjusted

P-value 0.002

Odd Ratio (95% CI) 2.97 (1.48–5.99)

PHE rate

Variable(s)

Mortality

Poor functional outcome (mRS >2)

Binary outcome

Effect of peri-hematomal edema expansion rate between admission and 24 hours post-intracerebral hemorrhage on clinical outcome determined using logistic regression analysis

Author Manuscript

Table 2 Urday et al. Page 16

Crit Care Med. Author manuscript; available in PMC 2017 April 01.

Author Manuscript

Author Manuscript

Author Manuscript 1.07 (1.00–1.16) 1.01 (0.92–1.11) 1.09 (0.99–1.19) 1.08 (0.99–1.18) 1.05 (0.95–1.16) 1.08 (0.99–1.17)

PHE rate, GCS adjusted

PHE rate, All ICH score componentsa adjusted

PHE rate, Any HE adjusted

PHE rate, HE > 6 mL adjusted

PHE rate, HE >12.5 mL adjusted

PHE rate, HE > 33% adjusted

1.09 (0.98–1.21)

Crit Care Med. Author manuscript; available in PMC 2017 April 01. 0.13

0.09

0.07

0.35

0.10

0.07

0.85

0.14

0.05

0.38

0.10

0.07

P-value

1.60 (1.01–2.53)

1.28 (1.02–1.64)

1.28 (1.00–1.61)

1.28 (1.01–1.64)

1.28 (1.01–1.64)

1.30 (1.02–1.64)

1.54 (1.04–2.22)

1.30 (1.01–1.67)

1.28 (1.01–1.61)

1.43 (1.02–2.04)

1.28 (1.01–1.64)

1.28 (1.01–1.64)

Odd Ratio (95% CI)

0.04

0.03

0.05

0.05

0.04

0.04

0.03

0.04

0.04

0.04

0.04

0.04

P-value

1.09 (1.00–1.19)

1.10 (1.01–1.19)

1.10 (1.01–1.19)

1.08 (0.99–1.18)

1.10 (1.01–1.20)

1.10 (1.01–1.20)

1.06 (0.97–1.15)

1.09 (1.00–1.19)

1.12 (1.03–1.23)

1.07 (0.98–1.17)

1.09 (1.01–1.19)

1.11 (1.01–1.20)

Odd Ratio (95% CI)

0.05

0.04

0.03

0.10

0.03

0.02

0.21

0.04

0.01

0.16

0.04

0.02

P-value

Higher (worse) mRS score

Ordinal Outcome

DNR indicates do-not-resuscitate order; GCS, Glasgow Coma Scale; HE, hemorrhage expansion; ICH, intracerebral hemorrhage; IVH, intraventricular hemorrhage; mRS, modified Rankin Scale; OR, odds ratio; PHE, peri-hematomal edema

ICH score components: Age, ICH volume, IVH volume, GCS

a

ORs are per 0.04 mL/h increase

PHE rate, DNR status adjusted

1.08 (0.99–1.17)

1.09 (1.00–1.20)

PHE rate, IVH vol adjusted

scan adjusted

1.04 (0.95–1.14)

PHE rate, ICH vol adjusted

PHE rate, Time to

1.07 (0.99–1.17)

PHE rate, Age adjusted

1st

1.08 (1.00–1.18)

Odd Ratio (95% CI)

PHE rate

Variable(s)

Mortality

Poor functional outcome (mRS >2)

Binary outcome

Effect of peri-hematomal edema expansion rate between admission and 72 hours post-intracerebral hemorrhage on clinical outcome determined using logistic regression analysis

Author Manuscript

Table 3 Urday et al. Page 17

Author Manuscript

Author Manuscript

Author Manuscript 1.03 (1.01–1.05) 1.02 (1.00–1.05) 1.01 (0.97–1.05) 1.03 (1.01–1.05) 1.03 (1.01–1.06)

Absolute PHE volume, IVH vol adjusted

Absolute PHE volume, GCS adjusted

Absolute PHE volume, All ICH score componentsa adjusted

Absolute PHE volume, Time to 72 h scan

Absolute PHE volume, DNR status

0.02

0.008

0.61

0.02

0.01

1.01 (0.99–1.03)

1.01 (0.99–1.04)

1.00 (0.95–1.05)

1.02 (1.00–1.05)

1.01 (0.99–1.03)

0.98 (0.94–1.02)

1.01 (0.99–1.03)

1.01 (0.99–1.03)

Odd Ratio (95% CI)

0.33

0.16

0.91

0.08

0.23

0.39

0.19

0.19

P-value

1.02 (1.00–1.04)

1.02 (1.01–1.04)

1.01 (0.98–1.04)

1.02 (1.01–1.04)

1.02 (1.01–1.04)

1.00 (0.98–1.03)

1.02 (1.01–1.04)

1.02 (1.01–1.04)

Odd Ratio (95% CI)

0.01

0.004

0.52

0.005

0.004

0.81

0.007

0.005

P-value

Higher (worse) mRS score

Ordinal Outcome

DNR indicates do-not-resuscitate order; GCS, Glasgow Coma Scale; ICH, intracerebral hemorrhage; IVH, intraventricular hemorrhage; mRS, modified Rankin Scale; OR, odds ratio; PHE, peri-hematomal edema

ICH score components: Age, ICH volume, IVH volume, GCS

a

1.01 (0.98–1.04)

0.51

0.01

1.03 (1.01–1.05)

Absolute PHE volume, Age adjusted

Absolute PHE volume, ICH vol adjusted

P-value 0.008

Odd Ratio (95% CI) 1.03 (1.01–1.05)

Absolute PHE volume

Variable(s)

Mortality

Poor functional outcome mRS >2)

Binary outcome

Effect of absolute peri-hematomal edema volume at 72 hours post-intracerebral hemorrhage on clinical outcome determined using logistic regression analysis

Author Manuscript

Table 4 Urday et al. Page 18

Crit Care Med. Author manuscript; available in PMC 2017 April 01.

Rate of Perihematomal Edema Expansion Predicts Outcome After Intracerebral Hemorrhage.

Intracerebral hemorrhage is a devastating disorder with no current treatment. Whether perihematomal edema is an independent predictor of neurologic ou...
NAN Sizes 0 Downloads 11 Views