Burns JD, Manno EM. Primary intracerebral hemorrhage: update on epidemiology, pathophysiology, and

treatment strategies. Compr Ther. 2008;34(3/4):183–195.

Ko SB, Choi HA, Lee K. Clinical syndromes and management of intracerebral hemorrhage. Curr Atheroscler

Rep. 2012;14(4):307–313.

Aksoy D et al. Magnetic resonance imaging profile of blood-brain barrier injury in patients with acute

intracerebral hemorrhage. J Am Heart Assoc. 2013;2(3):e000161.

Venkatasubramanian C et al. Natural history of perihematomal edema after intracerebral hemorrhage measured

by serial magnetic resonance imaging. Stroke. 2011;42(1):73–80.

Staykov D et al. Natural course of perihemorrhagic edema after intracerebral hemorrhage. Stroke.

2011;42(9):2625–2629.

Li N et al. Association of molecular markers with perihematomal edema and clinical outcome in intracerebral

hemorrhage. Stroke. 2013;44(3):658–663.

BalamiJS, Buchan AM. Complications of intracerebral haemorrhage. Lancet Neurol. 2012;11(1):101–118.

Barras CD et al. Density and shape as CT predictors of intracerebral hemorrhage growth. Stroke.

2009;40(4):1325–1331.

James PA et al. 2014 evidence-based guideline for the management of high blood pressure in adults: report from

the panel members appointed to the Eighth Joint National Committee (JNC 8). JAMA. 2014;311(5):507–520.

Erratum in:JAMA. 2014;311(17):1809.

Neal B et al. Effects of ACE inhibitors, calcium antagonists, and other blood-pressure-lowering drugs: results of

prospectively designed overviews of randomised trials. Lancet. 2000;356(9246):1955–1964.

Kannel WB, McGee DL. Diabetes and cardiovascular disease. The Framingham study. JAMA.

1979;241(19):2035–2038.

UK Prospective Diabetes Study (UKPDS) Group. Effect of intensive blood-glucose control with metformin on

complications in overweight patients with type 2 diabetes (UKPDS 34). Lancet. 1998;352(9131):854–865.

Erratum in: Lancet. 1998;352(9139):1558.

Lindholm LH et al. Cardiovascular morbidity and mortality in patients with diabetes in the Losartan Intervention

For Endpoint reduction in hypertension study (LIFE): a randomized trial against atenolol. Lancet. 2002;

359(9311):1004–1010.

Heart Outcomes Prevention Evaluation Study Investigators. Effects of ramipril on cardiovascular and

microvascular outcomes in people with diabetes mellitus: results of the HOPE study and MICRO-HOPE

substudy. Lancet. 2000;355(9200):253–259. Erratum in: Lancet. 2000;356(9232):860.

Collins R et al. MRC/BHF Heart Protection Study of cholesterol-lowering with simvastatin in 5963 people with

diabetes: a randomised placebo-controlled trial. Lancet. 2003;361(9374):2005–2016.

Colhoun HM et al. Primary prevention of cardiovascular disease with atorvastatin in type 2 diabetes in the

Collaborative Atorvastatin Diabetes Study (CARDS): multicentre randomised placebo-controlled trial. Lancet.

2004; 364(9435):685–696.

Blake GJ et al. Projected life-expectancy gains with statin therapy for individuals with elevated C-reactive protein

levels. J Am Coll Cardiol. 2002;40(1):49–55.

Vaughan CJ et al. Do statins afford neuroprotection in patients with cerebral ischaemia and stroke? CNS Drugs.

2001;15(8):589–596.

Gil-Núñez AC, Villanueva JA. Advantages of lipid-lowering therapy in cerebral ischemia: role of HMG-CoA

reductase inhibitors. Cerebrovasc Dis. 2001;11(Suppl 1):85–95.

Rexrode KM et al. A prospective study of body mass index, weight change, and risk of stroke in women. JAMA.

1997;277(19):1539–1545.

Kurth T et al. Body mass index and the risk of stroke in men. Arch Intern Med. 2002;162(22):2557–2562.

Neter JE et al. Influence of weight reduction on blood pressure: a meta-analysis of randomized controlled trials.

Hypertension. 2003;42(5):878–884.

He J et al. Dietary sodium intake and subsequent risk of cardiovascular disease in overweight adults. JAMA.

1999;282(21):2027–2034.

Khaw KT, Barrett-Connor E. Dietary potassium and stroke-associated mortality. A 12-year prospective

population study. N EnglJ Med. 1987;316(5):235–240.

Fletcher GF. Exercise in the prevention of stroke. Health Rep. 1994;6(1):106–110.

Lindenstrøm E et al. Lifestyle factors and risk of cerebrovascular disease in women. The Copenhagen City Heart

Study. Stroke. 1993;24(10):1468–1472.

Bonita R et al. Passive smoking as well as active smoking increases the risk of acute stroke. Tob Control.

1999;8(2):156–160.

Robbins AS et al. Cigarette smoking and stroke in a cohort of U.S. male physicians. Ann Intern Med.

1994;120(6):458–462.

Viscoli CM et al. A clinical trial of estrogen-replacement therapy after ischemic stroke. N Engl J Med.

2001;345(17):1243–1249.

Hulley S et al. Randomized trial of estrogen plus progestin for secondary prevention of coronary heart disease in

postmenopausal women. Heart and Estrogen/progestin Replacement Study (HERS) Research Group. JAMA.

1998; 280(7):605–613.

Simon JA et al. Postmenopausal hormone therapy and risk of stroke: the Heart and Estrogen-progestin

Replacement Study (HERS). Circulation. 2001;103(5):638–642.

Rossouw JE et al. Risks and benefits of estrogen plus progestin in healthy postmenopausal women: principal

results from the Women’s Health Initiative randomized controlled trial. JAMA. 2002;288(3):321–333.

Anderson GL et al. Effects of conjugated equine estrogen in postmenopausal women with hysterectomy: the

Women’s Health Initiative randomized controlled trial. JAMA. 2004;291(14):1701–1712.

Steering Committee of the Physicians’ Health Study Research Group. Final report on the aspirin component of

the ongoing Physicians’ Health Study. N EnglJ Med. 1989;321(3):129–135.

Chen ZM et al. Indications for early aspirin use in acute ischemic stroke: a combined analysis of 40,000

randomized patients from the chinese acute stroke trial and the internationalstroke trial. On behalf of the CAST

and IST collaborative groups. Stroke. 2000;31(6):1240–1249.

Iso H et al. Prospective study of aspirin use and risk of stroke in women. Stroke. 1999;30(9):1764–1771.

Ridker PM et al. A randomized trial of low-dose aspirin in the primary prevention of cardiovascular disease in

women. N EnglJ Med. 2005;352(13):1293–1304.

January CT et al. 2014 AHA/ACC/HRS guideline for the management of patients with atrial fibrillation: a report

of the American College of Cardiology/American Heart Association Task Force on practice guidelines and the

Heart Rhythm Society. Circulation. 2014;130(23):e199–e267. Erratum in: Circulation. 2014; 130(23):e272–e274.

Petersen P et al. Placebo-controlled, randomised trial of warfarin and aspirin for prevention of thromboembolic

complications in chronic atrial fibrillation. The Copenhagen AFASAK study. Lancet. 1989;1(8631):175–179.

The Boston Area Anticoagulation Trial for Atrial Fibrillation Investigators. The effect of low-dose warfarin on the

risk of stroke in patients with nonrheumatic atrial fibrillation. N EnglJ Med. 1990;323(22):1505–1511.

Connolly SJ et al. Canadian Atrial Fibrillation Anticoagulation (CAFA) Study. J Am Coll Cardiol. 1991;18(2):349–

355.

Stroke Prevention in Atrial Fibrillation Investigators. Warfarin versus aspirin for prevention of thromboembolism in

atrial fibrillation: Stroke Prevention in Atrial Fibrillation II Study. Lancet. 1994;343(8899):687–691.

Stroke Prevention in Atrial Fibrillation Investigators. Stroke Prevention in Atrial Fibrillation Study: final results.

Circulation.1991;84(2):527–539.

Jauch EC et al. Guidelines for the early management of patients with acute ischemic stroke: a guideline for

healthcare professionals from the American Heart Association/American Stroke Association. Stroke.

2013;44(3):870–947.

National Institute of Neurological Disorders and Stroke (NINDS). NIH Stroke Scale.

http://www.ninds.nih.gov/doctors/NIH_Stroke_Scale_Booklet.pdf Accessed June 4, 2015.

Solis OJ et al. Cerebral angiography in acute cerebral infarction. Rev Interam Radiol. 1977;2(1):19–25.

Symon L et al. The concepts of thresholds of ischaemia in relation to brain structure and function. J Clin Pathol

Suppl (R Coll Pathol). 1977;11:149–154.

Sharbrough FW et al. Correlation of continuous electroencephalograms with cerebral blood flow measurements

during carotid endarterectomy. Stroke. 1973;4(4):674–683.

Sundt TM Jr et al. Restoration of middle cerebral artery flow in experimental infarction. J Neurosurg.

1969;31(3):311–321.

The Multicenter Acute Stroke Trial—Europe Study Group. Thrombolytic therapy with streptokinase in acute

ischemic stroke. N EnglJ Med. 1996;335(3):145–150.

Donnan GA et al. Streptokinase for acute ischemic stroke with relationship to time of administration: Australian

Streptokinase (ASK) Trial Study Group. JAMA. 1996;276(12):961–966.

The National Institute of Neurological Disorders and Stroke rt-PA Stroke Study Group. Tissue plasminogen

activator for acute ischemic stroke. N EnglJ Med. 1995;333(24):1581–1587.

Hacke W et al. Intravenous thrombolysis with recombinant tissue plasminogen activator for acute hemispheric

stroke. The European Cooperative Acute Stroke Study (ECASS). JAMA. 1995;274(13):1017–1025.

Multicentre Acute Stroke Trial—Italy (MAST-I) Group. Randomised controlled trial of streptokinase, aspirin, and

combination of both in treatment of acute ischaemic stroke. Lancet. 1995;346(8989):1509–1514.

Hacke W et al. Randomised double-blind placebo-controlled trial of thrombolytic therapy with intravenous

alteplase in acute ischaemic stroke (ECASS II). Second European-Australasian Acute Stroke Study

Investigators. Lancet. 1998; 352(9136):1245–1251.

Hacke W et al. Association of outcome with early stroke treatment: pooled analysis of ATLANTIS, ECASS, and

NINDS rt-PA stroke trials. Lancet. 2004;363(9411):768–774.

Hacke W et al. Thrombolysis with alteplase 3 to 4.5 hours after acute ischemic stroke. N Engl J Med.

2008;359(13):1317–1329.

IST-3 Collaborative Group et al. The benefits and harms of intravenous thrombolysis with recombinant tissue

plasminogen activator within 6 h of acute ischaemic stroke (the third international stroke trial [IST-3]): a

randomised controlled trial. Lancet. 2012;379(9834):2352–2363. Erratum in: Lancet. 2012; 380(9843):730.

Kammersgaard LP et al. Admission body temperature predicts long-term mortality after acute stroke: the

Copenhagen Stroke Study. Stroke. 2002;33(7):1759–1762.

Zaremba J. Hyperthermia in ischemic stroke. Med Sci Monit. 2004;10(6):RA148–RA153.

Dippel DW et al. Effect of paracetamol (acetaminophen) and ibuprofen on body temperature in acute ischemic

stroke PISA, a phase II double-blind, randomized, placebo-controlled trial [ISRCTN98608690]. BMC

Cardiovasc Disord. 2003;3:2.

Kagansky N et al. The role of hyperglycemia in acute stroke. Arch Neurol. 2001;58(8):1209–1212.

Brott T et al. Hypertension and its treatment in the NINDS rt-PA Stroke Trial. Stroke. 1998;29(8):1504–1509.

Duke RJ et al. Intravenous heparin for the prevention of stroke progression in acute partial stable stroke. Ann

Intern Med. 1986;105(6):825–828.

International Stroke Trial Collaborative Group. The International Stroke Trial (IST): a randomised trial of aspirin,

subcutaneous heparin, both, or neither among 19435 patients with acute ischaemic stroke. Lancet.

1997;349(9065):1569–1581.

Rödén-Jüllig A, Britton M. Effectiveness of heparin treatment for progressing ischaemic stroke: before and after

study. J Intern Med. 2000;248(4):287–291.

Kay R et al. Low-molecular-weight heparin for the treatment of acute ischemic stroke. N Engl J Med.

1995;333(24):1588–1593.

The Publications Committee for the Trial of ORG 10172 in Acute Stroke Treatment (TOAST) Investigators. Low

molecular weight heparinoid, ORG 10172 (danaparoid), and outcome after acute ischemic stroke: a randomized

controlled trial. JAMA. 1998;279(16):1265–1272.

Berge E et al. Low molecular-weight heparin versus aspirin in patients with acute ischaemic stroke and atrial

fibrillation: a double-blind randomised study. HAEST Study Group. Heparin in Acute Embolic Stroke Trial.

Lancet. 2000;355(9211):1205–1210.

Diener HC et al. Treatment of acute ischemic stroke with the low-molecular-weight heparin certoparin: results of

the TOPAS trial. Therapy of Patients with Acute Stroke (TOPAS) Investigators. Stroke. 2001;32(1):22–29.

Coull BM et al. Anticoagulants and antiplatelet agents in acute ischemic stroke: report of the Joint Stroke

Guideline Development Committee of the American Academy of Neurology and the American Stroke

Association (a division of the American Heart Association). Neurology. 2002;59(1): 13–22.

CAST (Chinese Acute Stroke Trial) Collaborative Group. CAST: randomised placebo-controlled trial of early

aspirin use in 20,000 patients with acute ischaemic stroke. Lancet. 1997;349(9066):1641–1649.

Wang Y et al. Clopidogrel with aspirin in acute minor stroke or transient ischemic attack. N Engl J Med.

100.

101.

102.

103.

104.

105.

106.

107.

108.

109.

2013;369(1):11–19.

Abciximab in Ischemic Stroke Investigators. Abciximab in acute ischemics stroke: a randomized, double-blind,

placebo-controlled, dose-escalation study. Stroke. 2000;31(3):601–609.

Kobayashi S, TazakiY. Effect of the thrombin inhibitor argatroban in acute cerebral thrombosis. Semin Thromb

Hemost. 1997;23(6):531–534.

Furlan A et al. Intra-arterial prourokinase for acute ischemic stroke. The PROACT II study: a randomized

controlled trial. Prolyse in acute cerebral thromboembolism. JAMA. 1999;282(21):2003–2011.

Ogawa A et al. Randomized trial of intraarterial infusion of urokinase within 6 hours of middle cerebral artery

stroke: the middle cerebral artery embolism local fibrinolytic intervention trial (MELT) Japan. Stroke.

2007;38(10):2633–2639.

Saver JL et al. Stent-retriever thrombectomy after intravenous t-PA vs. t-PA alone in stroke. N Engl J Med.

2015;372(24):2285–2295.

Berkhemer OA et al. A randomized trial of intraarterial treatment for acute ischemic stroke. N Engl J Med.

2015;372(1):11–20. Erratum in: N EnglJ Med. 2015;372(4):394.

Goyal M et al. Randomized assessment of rapid endovascular treatment of ischemic stroke. N Engl J Med.

2015;372(11):1019–1030.

Campbell BC et al. Endovascular therapy for ischemic stroke with perfusion-imaging selection. N Engl J Med.

2015;372(11):1009–1018.

Jovin TG et al. Thrombectomy within 8 hours after symptom onset in ischemic stroke. N Engl J Med.

2015;372(24):2296–1306.

Smith WS et al. Mechanical thrombectomy for acute ischemic stroke: final results of the Multi MERCI trial.

Stroke. 2008;39(4):1205–1212.

Ciccone A et al. Endovascular treatment for acute ischemic stroke. N EnglJ Med. 2013;368(10):904–913.

Broderick JP et al. Endovascular therapy after intravenous t-PA versus t-PA alone for stroke. N Engl J Med.

2013;368(10):893–903. Erratum in: N EnglJ Med. 2013;368(13):1265.

Kidwell CS et al. A trial of imaging selection and endovascular treatment for ischemic stroke. N Engl J Med.

2013;368(10):914–923.

Robinson RG. Treatment issues in poststroke depression. Depress Anxiety. 1998; 8(Suppl 1):85–90.

Antithrombotic Trialists’ Collaboration. Collaborative meta-analysis of randomised trials of antiplatelet therapy for

prevention of death, myocardial infarction, and stroke in high risk patients. BMJ. 2002;324(7329):71–86.

Erratum in: BMJ 2002;324(7330):141.

Bousser MG et al. "AICLA" controlled trial of aspirin and dipyridamole in the secondary prevention of atherothrombotic cerebral ischemia. Stroke. 1983;14(1):5–14.

Sorensen PS et al. Acetylsalicylic acid in the prevention of stroke in patients with reversible cerebral ischemic

attacks. A Danish cooperative study. Stroke. 1983;14(1):15–22.

Antiplatelet Trialists’ Collaboration. Secondary prevention of vascular disease by prolonged antiplatelet

treatment. Br Med J (Clin Res Ed). 1988;296(6618):320–331.

A Swedish Cooperative Study. High-dose acetylsalicylic acid after cerebral infarction. Stroke.1987;18(2):325–

334.

Farrell B et al. The United Kingdom transient ischaemic attack (UK-TIA) aspirin trial: final results. J Neurol

Neurosurg Psychiatry. 1991;54(12):1044–1054.

Bernstein EF et al. Life expectancy and late stroke following carotid endarterectomy. Ann Surg.

1983;198(1):80–86.

The Dutch TIA Trial Study Group. A comparison of two doses of aspirin (30 mg vs. 283 mg a day) in patients

after a transient ischemic attack or minor ischemic stroke. N EnglJ Med. 1991;325(18):1261–1266.

The SALT Collaborative Group. Swedish Aspirin Low-Dose Trial (SALT) of 75 mg aspirin as secondary

prophylaxis after cerebrovascular ischaemic events. Lancet. 1991;338(8779):1345–1349.

Helgason CM, Tortorice KL, Winkler SR, et al. Aspirin response and failure in cerebral infarction. Stroke.

1993;24(3):345–350.

Hansson L et al. Effects of intensive blood-pressure lowering and low-dose aspirin in patients with hypertension:

principal results of the Hypertension Optimal Treatment (HOT) randomised trial. HOT Study Group. Lancet.

1998;351(9118):1755–1762.

Gent M et al. The Canadian American Ticlopidine Study (CATS) in thromboembolic stroke. Lancet.

1989;1(8649):1215–1220.

Hass WK et al. A randomized trial comparing ticlopidine hydrochloride with aspirin for the prevention of stroke

110.

111.

112.

113.

114.

115.

116.

117.

118.

119.

120.

121.

122.

123.

124.

125.

126.

127.

128.

129.

130.

131.

132.

133.

134.

135.

in high-risk patients. Ticlopidine Aspirin Stroke Study Group. N EnglJ Med. 1989;321(8):501–507.

CAPRIE Steering Committee. A randomised, blinded, trial of clopidogrel versus aspirin in patients at risk of

ischaemic events (CAPRIE). Lancet. 1996;348(9038):1329–1339.

Bennett CL et al. Thrombotic thrombocytopenic purpura associated with clopidogrel. N Engl J Med.

2000;342(24):1773–1777.

The ESPS Group. The European Stroke Prevention Study (ESPS). Principal end-points. Lancet.

1987;2(8572):1351–1354.

Diener HC et al. European Stroke Prevention Study. 2. Dipyridamole and acetylsalicylic acid in the secondary

prevention of stroke. J Neurol Sci. 1996;143(1/2):1–13.

ESPRIT Study Group et al. Aspirin plus dipyridamole versus aspirin alone after cerebral ischaemia of arterial

origin (ESPRIT): randomised controlled trial. Lancet. 2006;367(9523):1665–1673. Erratum in: Lancet.

2007;369(9558):274

Sacco RL et al. Aspirin and extended-release dipyridamole versus clopidogrel for recurrent stroke. N Engl J

Med. 2008;359(12):1238–1251.

Eikelboom JW et al. Antiplatelet drugs: antithrombotic therapy and prevention of thrombosis, 9th ed: American

College of Chest Physicians Evidence-Based Clinical Practice Guidelines. Chest. 2012;141 (2, Suppl):e89S–

e119S.

Huang Y et al. Cilostazol as an alternative to aspirin after ischaemic stroke: a randomised, double-blind, pilot

study. Lancet Neurol. 2008;7(6):494–499. Erratum in: Lancet Neurol. 2008;7(8):675.

Shinohara Y et al. Cilostazol for prevention of secondary stroke (CSPS 2): an aspirin-controlled, double-blind,

randomised non-inferiority trial. Lancet Neurol. 2010;9(10):959–968.

The Stroke Prevention in Reversible Ischemia Trial (SPIRIT) Study Group. A randomized trial of anticoagulants

versus aspirin after cerebral ischemia of presumed arterial origin. Ann Neurol. 1997;42(6):857–865.

Mohr JP et al. A comparison of warfarin and aspirin for the prevention of recurrent ischemic stroke. N Engl J

Med. 2001;345(20):1444–1451.

Chimowitz M et al. Warfarin-Aspirin Symptomatic Intracranial Disease (WASID) trial: final results. Stroke.

2004;35(1):235.

Diener HC et al. Aspirin and clopidogrel compared with clopidogrel alone after recent ischaemic stroke or

transient ischaemic attack in high-risk patients (MATCH): randomised, double-blind, placebo-controlled trial.

Lancet. 2004; 364(9431):331–337.

Patrono C et al. Platelet-active drugs: the relationships among dose, effectiveness, and side effects: the Seventh

ACCP Conference on antithrombotic and thrombolytic therapy. Chest. 2004;126(3, Suppl): 234S–264S.

Rocca B et al. Cyclooxygenase-2 expression is induced during human megakaryopoiesis and characterizes

newly formed platelets. Proc Natl Acad Sci USA. 2002;99(11):7634–7639.

Catella-Lawson F et al. Cyclooxygenase inhibitors and the antiplatelet effects of aspirin. N Engl J Med.

2001;345(25):1809–1817.

Eikelboom JW et al. Aspirin-resistant thromboxane biosynthesis and the risk of myocardial infarction, stroke, or

cardiovascular death in patients at high risk for cardiovascular events. Circulation. 2002;105(14):1650–1655.

North American Symptomatic Carotid Endarterectomy Trial Collaborators. Beneficial effect of carotid

endarterectomy in symptomatic patients with high-grade carotid stenosis. N EnglJ Med. 1991;325(7):445–453.

Barnett HJ et al. Evidence based cardiology: prevention of ischaemic stroke. BMJ. 1999;318(7197):1539–1543.

Biller J et al. Guidelines for carotid endarterectomy: a statement for healthcare professionals from a Special

Writing Group of the Stroke Council, American Heart Association. Circulation. 1998;97(5):501–509

Harker LA et al. Failure of aspirin plus dipyridamole to prevent restenosis after carotid endarterectomy. Ann

Intern Med. 1992;116(9):731–736.

Bhatt DL et al. Dual antiplatelet therapy with clopidogrel and aspirin after carotid artery stenting. J Invasive

Cardiol. 2001;13(12):767–771.

Alberts MJ. Results of a multicenter prospective randomized trial of carotid artery stenting vs. carotid

endarterectomy. Stroke. 2001;32(1):325.

CAVATAS Investigators. Endovascular versus surgical treatment in patients with carotid stenosis in the Carotid

and Vertebral Artery Transluminal Angioplasty Study (CAVATAS): a randomised trial. Lancet.

2001;357(9270):1729–1737.

Yadav JS et al. Protected carotid-artery stenting versus endarterectomy in high-risk patients. N Engl J Med.

2004;351(15):1493–1501.

Toyoda K et al. Blood pressure levels and bleeding events during antithrombotic therapy: the Bleeding with

136.

137.

138.

139.

140.

141.

142.

143.

144.

145.

146.

147.

148.

149.

150.

151.

152.

153.

154.

155.

156.

157.

158.

159.

160.

161.

162.

Antithrombotic Therapy (BAT) Study. Stroke. 2010;41(7):1440–1444.

Rosand J et al. The effect of warfarin and intensity of anticoagulation on outcome of intracerebral hemorrhage.

Arch Intern Med. 2004;164(8):880–884.

García-Rodríguez LA et al. Antithrombotic drugs and risk of hemorrhagic stroke in the general population.

Neurology. 2013;81(6):566–574.

Flaherty ML et al. Warfarin use leads to larger intracerebral hematomas. Neurology. 2008;71(14):1084–1089.

LoPresti MA et al. Hematoma volume as the major determinant of outcomes after intracerebral hemorrhage. J

Neurol Sci. 2014;345(1/2):3–7.

Hackam DG, Mrkobrada M. Selective serotonin reuptake inhibitors and brain hemorrhage: a meta-analysis.

Neurology. 2012;79(18):1862–1865.

He J et al. Aspirin and risk of hemorrhagic stroke: a meta-analysis of randomized controlled trials. JAMA.

1998;280(22):1930–1935.

Martin-Schild S et al. Intracerebral hemorrhage in cocaine users. Stroke. 2010;41(4):680–684.

Kernan WN et al. Phenylpropanolamine and the risk of hemorrhagic stroke. N EnglJ Med. 2000;343(25):1826–

1832.

Lee SM et al. Caffeine-containing medicines increase the risk of hemorrhagic stroke. Stroke. 2013;44(8):2139–

2143.

Ariesen MJ et al. Risk factors for intracerebral hemorrhage in the general population: a systematic review.

Stroke. 2003;34(8):2060–2065.

Sturgeon JD et al. Risk factors for intracerebral hemorrhage in a pooled prospective study. Stroke.

2007;38(10):2718–2725.

Huhtakangas J et al. Effect of increased warfarin use on warfarin-related cerebral hemorrhage: a longitudinal

population-based study. Stroke. 2011;42(9):2431–2435.

Flaherty ML et al. The increasing incidence of anticoagulant-associated intracerebral hemorrhage. Neurology.

2007;68(2):116–121.

Kuramatsu JB et al. Anticoagulant reversal, blood pressure levels, and anticoagulant resumption in patients with

anticoagulation-related intracerebral hemorrhage. JAMA. 2015;313(8):824–836.

Davis SM et al. Hematoma growth is a determinant of mortality and poor outcome after intracerebral

hemorrhage. Neurology. 2006;66(8):1175–1181.

Hemphill JC 3rd et al. Guidelines for the Management of spontaneous intracerebral hemorrhage: a guideline for

healthcare professionals from the American Heart Association/American Stroke Association. Stroke. 2015.

doi:10.1161/STR.0000000000000069

Holbrook A et al. Evidence-based management of anticoagulant therapy: antithrombotic therapy and prevention

of thrombosis, 9th ed: American College of Chest Physicians Evidence-Based Clinical Practice Guidelines.

Chest. 2012; 141(2, Suppl):e152S–e184S.

Huttner HB et al. Hematoma growth and outcome in treated neurocritical care patients with intracerebral

hemorrhage related to oral anticoagulant therapy: comparison of acute treatment strategies using vitamin K,

fresh frozen plasma, and prothrombin complex concentrates. Stroke. 2006;37(6):1465–1470.

Hanger HC et al. Warfarin-related intracerebral haemorrhage: better outcomes when reversal includes

prothrombin complex concentrates. Intern Med J. 2013;43(3):308–316.

Steiner T et al. Intracerebral hemorrhage associated with oral anticoagulant therapy: current practices and

unresolved questions. Stroke. 2006;37(1):256–262.

Connolly SJ et al. Dabigatran versus warfarin in patients with atrial fibrillation. N Engl J Med.

2009;361(12):1139–1151. Erratum in: N EnglJ Med. 2010;363(19):1877.

Patel MR et al. Rivaroxaban versus warfarin in nonvalvular atrial fibrillation. N Engl J Med. 2011;365(10):883–

891.

Granger CB et al. Apixaban versus warfarin in patients with atrial fibrillation. N EnglJ Med. 2011;365(11):981–

992.

Sakamoto Y et al. Systolic blood pressure after intravenous antihypertensive treatment and clinical outcomes in

hyperacute intracerebral hemorrhage: the stroke acute management with urgent risk-factor assessment and

improvement-intracerebral hemorrhage study. Stroke. 2013;44(7):1846–1851.

Rodriguez-Luna D et al. Impact of blood pressure changes and course on hematoma growth in acute

intracerebral hemorrhage. Eur J Neurol. 2013; 20(9):1277–1283.

Tikhonoff V et al. Blood pressure as a prognostic factor after acute stroke. Lancet Neurol. 2009;8(10):938–948.

Garg RK et al. Blood pressure reduction, decreased diffusion on MRI, and outcomes after intracerebral

163.

164.

165.

166.

167.

168.

169.

170.

hemorrhage. Stroke. 2012;43(1):67–71.

Anderson CS et al. Rapid blood-pressure lowering in patients with acute intracerebral hemorrhage. N Engl J

Med. 2013;368(25):2355–2365.

Anderson CS et al. Effects of early intensive blood pressure-lowering treatment on the growth of hematoma and

perihematomal edema in acute intracerebral hemorrhage: the Intensive Blood Pressure Reduction in Acute

Cerebral Haemorrhage Trial (INTERACT). Stroke. 2010;41(2):307–312.

Qureshi AI. Significance of lesions with decreased diffusion on MRI in patients with intracerebral hemorrhage.

Stroke. 2012;43(1):6–7.

Arima H et al. Lower treatment blood pressure is associated with greatest reduction in hematoma growth after

acute intracerebral hemorrhage. Hypertension. 2010;56(5):852–858.

Helbok R et al. Effect of mannitol on brain metabolism and tissue oxygenation in severe haemorrhagic stroke. J

Neurol Neurosurg Psychiatry. 2011;82(4): 378–383.

Greer DM et al. Impact of fever on outcome in patients with stroke and neurologic injury: a comprehensive

meta-analysis. Stroke. 2008;39(11): 3029–3035.

Broderick J et al. Guidelines for the management of spontaneous intracerebral hemorrhage in adults: 2007

update: a guideline from the American Heart Association/American Stroke Association Stroke Council, High

Blood Pressure Research Council, and the Quality of Care and Outcomes in Research Interdisciplinary

Working Group. Stroke. 2007;38(6): 2001–2023.

Messé SR et al. Prophylactic antiepileptic drug use is associated with poor outcome following ICH. Neurocrit

Care. 2009;11(1):38–44.

p. 1318

Although acute infection generally is associated with an increased white

blood cell count, fever, and localizing signs, these symptoms may be

absent in less severe disease. More severe infection, including sepsis,

may be associated with hypotension, disseminated intravascular

coagulation, and end-organ dysfunction.

Case 62-1 (Questions 1, 2)

Figure 62-1

Other disease states, particularly autoimmune disease and malignancy,

may mimic infectious diseases. Although it should be considered a

diagnosis of exclusion, drug-induced fever should be ruled out,

particularly in patients without other classic signs and symptoms of

infection.

Case 62-1 (Question 3)

Site-specific signs and symptoms and host factors generally predict the

most likely pathogens, and empirical antimicrobial therapy should be

directed against these organisms. Rapid detection tests improve the

efficiency identifying a pathogen; however, they are more costly than

traditional methodology.

Case 62-1 (Questions 4, 5,

Tables 62-1, 62-2)

Isolation of an organism may reflect infection; however, colonization and

contamination must be ruled out to avoid unnecessary antimicrobial

exposure. Once a pathogen is identified, susceptibility tests, particularly

disk diffusion or broth dilution, can demonstrate the most active

antimicrobial agents.

Case 62-1 (Questions 6, 7,

Tables 62-3 through 62-7)

Once the site of infection is confirmed and the likely pathogens are

identified, drug distribution to the site of infection, dosage, route of

administration, antimicrobial toxicity, side effects, and costs, must be

considered before selection of therapy.

Case 62-1 (Questions 8–10,

Table 62-8)

Antimicrobial dosing should reflect site of infection, route of elimination,

and pharmacokinetics and pharmacodynamics.

Case 62-1 (Questions 11–13,

Table 62-9)

Antimicrobial failure may be related to pharmacologic factors

(inadequate dosing, insufficient penetration to the site of infection, and

inadequate duration) and host factors (presence of prosthetic material,

undrained focus of infection, and immune status). Adjunct therapies,

including pressors and volume repletion, may improve outcomes in

critically ill patients.

Case 62-1 (Questions 14–17)

APPROACHING THE PROBLEM

The proper choice, dose, and duration of antimicrobial therapy are based on several

factors. Before initiating therapy, it is important first to confirm an infectious versus

noninfectious process. Once infection has been documented, the most likely site must

be identified, and signs and symptoms (e.g., erythema associated with cellulitis)

generally direct the clinician to the likely source. Because certain pathogens are

known to be associated with a specific site of infection, empirical therapy often can

be directed against these organisms. Additional laboratory tests, including the Gram

stain, serologic analysis, and antimicrobial susceptibility testing, generally identify

the primary pathogen and active agents. Spectrum of activity, established clinical

efficacy, adverse effect profile, pharmacokinetic disposition, and cost considerations

ultimately guide the choice of therapy. Once an agent has been selected, the dosage

and duration should be based on the size of the patient, site of infection, route of

elimination, and other factors.

p. 1319

p. 1320

ESTABLISHING THE PRESENCE OF AN

INFECTION

CASE 62-1

QUESTION 1: R.G., a 63-year-old, 70-kg man in the intensive care unit, underwent emergency resection of

his large bowel. He has been mechanically ventilated throughout his postoperative course. On day 20 of his

hospital stay, R.G. suddenly becomes confused; his blood pressure (BP) drops to 70/30 mm Hg, with a heart

rate of 130 beats/minute. His extremities are cold to the touch, and he presents with circumoral pallor. His

temperature increases to 40°C (axillary), and his respiratory rate is 24 breaths/minute. Copious amounts of

yellow–green secretions are suctioned from his endotracheal tube.

Physical examination reveals sinus tachycardia with no rubs or murmurs. Rhonchi with decreased breath

sounds are observed on auscultation. The abdomen is distended, and R.G. complains of new abdominal pain. No

bowel sounds can be heard, and the stool is guaiac positive. Urine output from the Foley catheter has been 10

mL/hour for the past 2 hours. Erythema is noted around the central venous catheter.

A chest radiograph demonstrates bilateral lower lobe infiltrates, and urinalysis reveals >50 white blood

cells/high-power field (WBC/HPF), few casts, and a specific gravity of 1.015. Blood, endotracheal aspirate, and

urine cultures are pending. Other laboratory values include the following:

Sodium (Na), 131 mEq/L (normal, 135–147)

Potassium (K), 4.1 mEq/L (normal, 3.5–5)

Chloride (Cl), 110 mEq/L (normal, 95–105)

CO2

, 16 mEq/L (normal, 20–29 mEq/L)

Blood urea nitrogen (BUN), 58 mg/dL (normal, 8–18)

Serum creatinine (SCr), 3.8 mg/dL (increased from 0.9 mg/dL at admission; normal, 0.6–1.2)

Glucose, 320 mg/dL (normal, 70–110)

Serum albumin, 2.1 g/dL (normal, 4–6)

Hemoglobin (Hgb), 10.3 g/dL (13.5–17.5 g/dL male patients)

Hematocrit (Hct), 33% (normal, 39%–49% [male patients])

WBC count, 15,600/μL (normal, 4,500–10,000/μL) with bands present

Platelets, 40,000/μL (normal, 130,000–400,000)

Prothrombin time (PT), 18 seconds (normal, 10–12)

Erythrocyte sedimentation rate (ESR), 65 mm/hour (normal, 0–20)

Procalcitonin, 1 mcg/L (normal <0.25mcg/L)

Which of R.G.’s signs and symptoms are consistent with infection?

R.G. has numerous signs and symptoms consistent with an infectious process. His

WBC count (15,600/μL) is increased, and a “shift to the left” (presence of bands,

i.e., immature neutrophils) is observed on the differential. An increased WBC count

is commonly observed with infection, particularly with bacterial pathogens. The

WBC differential in patients with a bacterial infection often demonstrates a shift to

the left owing to the bone marrow response to infection. Although infection is usually

associated with an increased WBC, overwhelming sepsis can also be associated

with a markedly decreased WBC count. In less acute infection (e.g., uncomplicated

urinary tract infection, localized abscess), the WBC count may remain within the

normal range because less bone marrow response would be anticipated.

R.G.’s temperature is 40°C by axillary measurement. Fever is a common

manifestation of infection, with oral temperatures generally greater than 38°C. Oral

and axillary temperatures tend to be approximately 0.4°C lower compared with

rectal measurement. As a result, R.G.’s temperature would be expected to be 40.4°C

if his temperature had been taken rectally. In general, rectal measurement of

temperature is a more reliable determination of fever. Some patients with

overwhelming infection, however, may present with hypothermia with temperatures

less than 36°C. Furthermore, patients with localized infections (e.g., uncomplicated

urinary tract infection, chronic abscesses) may be afebrile.

The bilateral lower lobe infiltrates on R.G.’s chest radiograph, the presence of

copious amounts of yellow–green secretions from his endotracheal tube, and the

erythema surrounding his central venous catheter is also compatible with one or more

infectious processes. Furthermore, R.G. has signs and symptoms consistent with

sepsis.

ESTABLISHING THE SEVERITY OF AN

INFECTION

CASE 62-1, QUESTION 2: What signs and symptoms manifested by R.G. are consistent with a serious

systemic infection?

The term sepsis, while a poorly defined syndrome, generally suggests a more

systemic infection with the presence of pathogenic microorganisms and/or their

toxins in the blood. A uniform system for defining the spectrum of disorders

associated with sepsis has been established, but it remains difficult to precisely

define.

1

The pathogenesis of sepsis is complex (Fig. 62-1) and only partially understood.

2

,

3

Gram-negative aerobes produce endotoxin that results in a cascade of endogenous

mediator release, including tumor necrosis factor (TNF), interleukin 1 (IL-1) and

interleukin 6 (IL-6), platelet-activating factor (PAF), and various other substances

from mononuclear phagocytes and other cells. Although this initial stimulus

commonly is associated with gram-negative endotoxin, other substances, including

gram-positive exotoxin and fungal cell wall constituents, may also be associated with

cytokine release. After release of TNF, IL-1, and PAF, arachidonic acid is

metabolized to form leukotrienes, thromboxane A2

, and prostaglandins, particularly

prostaglandin E2 and prostaglandin I2

. IL-1 and IL-6 activate the T cells to produce

interferon, IL-2, IL-4, and granulocyte-macrophage colony-stimulating factor (GMCSF). Increased endothelial permeability ensues. Subsequently, the endothelium

releases two hemodynamically active substances: endothelium-derived relaxing

factor (EDRF) and endothelin-l. Activation of the complement cascade (fragments

C3a and C5a) follows, with additional vascular abnormalities and neutrophil

activation. Other potentially important agents in this cascade include adhesion

molecules, kinins, thrombin, myocardial depressant substance, endorphins, and heat

shock protein. The net result of this cascade involves several hemodynamic, renal,

acid–base, and other disorders. Uncontrolled inflammation and coagulation have a

particularly important role in this sepsis cascade.

3

Hemodynamic Changes

Critically ill patients often have central intravenous (IV) lines in place for measuring

cardiac output and systemic vascular resistance (SVR). In other words, these lines

are placed in the pulmonary artery and allow for more precise measurement of

critical hemodynamics. A normal SVR of 800 to 1,200 dyne·s·cm–5 may fall to 500 to

600 dyne·s·cm–5

in septic shock as a result of extensive vasodilation. In response to

vasodilation the heart rate increases leading to increased cardiac output from a

normal 4 to 6 L/minute to up to 11 to 12 L/minute in septic patients; stroke volume

remains unchanged or decreased. In addition to reflex tachycardia causing a rise in

heart rate, stress-induced catecholamine release (norepinephrine, epinephrine) also

contributes. The initial increase in cardiac output generally is insufficient to

overcome the vasodilatory state, and hypotension ensues. In overwhelming septic

shock, myocardial depression may result, resulting in a decreased cardiac output.

The combination of decreased cardiac output and decreased SVR results in

hypotension often unresponsive to pressors and IV fluids. R.G. has hemodynamic

evidence of septic shock. He is hypotensive (BP, 70/30 mm Hg) and tachycardic

(130 beats/minute), presumably in response to vasodilation and catecholamine

release.

p. 1320

p. 1321

Figure 62-1 The sepsis cascade. ARDS, acute respiratory distress syndrome; ARF, acute renal failure; DIC,

disseminated intravascular coagulation; GM-CSF, granulocyte-macrophage colony-stimulating factor; IL-1,

interleukin-1; IL-2, interleukin-2; IL-6, interleukin-6; PAF, platelet-activating factor; TNF, tumor necrosis factor.

Although vasodilation commonly occurs in sepsis, this dilation is unequal and

chaotic. Some vascular beds constrict and others dilate, resulting in maldistribution

of blood flow. In sepsis, blood generally is shunted away from the kidneys,

mesentery, and extremities.

When sepsis has progressed to septic shock, blood flow to most major organs is

decreased. Normal urine output of approximately 0.5 to 1.0 mL/kg/hour (30–70

mL/hour for a 70-kg patient) can decrease to less than 20 mL/hour in sepsis. The

urine output for R.G. has decreased to 10 mL/hour, consistent with sepsis-induced

perfusion abnormalities. Decreased blood flow to the kidney as well as mediatorinduced microvascular failure can cause acute-tubular necrosis. R.G.’s uremia

(BUN, 58 mg/dL) and increased serum creatinine concentration (3.8 mg/dL) are

consistent with decreased renal perfusion secondary to sepsis. Decreased blood flow

to the liver may result in “shock liver,” in which liver function tests, including

alanine aminotransferase (ALT), aspartate aminotransferase (AST), and alkaline

phosphatase, become elevated. The liver function tests for R.G. are not available;

however, his serum albumin concentration is low (2.1 g/dL) and his PT of 18

seconds is prolonged. Decreased blood flow to the musculature classically is

characterized by cool extremities, and decreased blood flow to the brain can result in

decreased mentation. R.G. is confused, his extremities are cold, and the area around

his mouth appears pale. All these signs and symptoms provide strong evidence that

he is in septic shock.

Cellular Changes

The sepsis syndrome is associated with significant abnormalities in cellular

metabolism. Glucose intolerance commonly is observed in sepsis, and patients with

previously normal blood glucose levels may experience sudden increases in blood

sugar. In some cases, a rise in glucose is one of the first signs of an infectious

process. R.G.’s increased blood glucose concentration (320 mg/dL) is, therefore,

consistent with infection. Other sensitive indicators of sepsis-associated

inflammation include the ESR, C-reactive protein, and procalcitonin, nonspecific

tests that are commonly elevated in various inflammatory states, including infection.

The ESR, C-reactive protein, or procalcitonin can be used to follow the progression

of infection; currently, R.G.’s ESR is elevated at 65 mm/hour. With appropriate

management of infection, the ESR would be expected to decrease; inadequate

treatment would be associated with persistent elevation of the ESR and C-reactive

protein. Procalcitonin is a more specific indicator for infection than ESR or Creactive protein and has been used as a tool to discontinue antibiotics in patients with

noninfectious inflammation.

4 At present, R.G.’s procalcitonin is 1.0 mcg/L, which is

consistent with infection-associated inflammati




Respiratory Changes

Production of organic acids, such as lactate, increased glycolysis, decreased

fractional extraction of oxygen, and abnormal delivery-dependent oxygen

consumption are observed in sepsis.

3 This process leads to metabolic acidosis, with

accompanying decreased serum bicarbonate levels. The lungs normally respond to

metabolic acidosis in a compensatory manner with an increased respiratory rate

(tachypnea), resulting in an increased elimination of arterial carbon dioxide. R.G.’s

acid–base status is consistent with sepsis- associated metabolic acidosis (CO2

, 16

mEq/L) and compensatory respiratory alkalosis (respiratory rate, 24 breaths/minute).

A late complication of the above-mentioned sepsis cascade is acute respiratory

distress syndrome (ARDS). ARDS initially was described as noncardiogenic

pulmonary edema with severe hypoxemia caused by right-to-left intrapulmonary

shunting resulting from atelectasis and edema-filled alveoli. The primary

pathophysiology of ARDS is a breakdown in the natural integrity of the alveolar

capillary network in the lung.

5

In the early phase of ARDS, patients have severe

alveolar edema with large numbers of inflammatory cells, primarily neutrophils. The

chronic phase of ARDS (10–14 days after development of the syndrome) is

associated with significant lung destruction. Emphysema, pulmonary vascular

obliteration, and fibrosis commonly are observed. Severe ARDS is associated with

ratios of arterial oxygen level to fraction of inspired oxygen (Pao2

/FIO2

) of less than

100, low

p. 1321

p. 1322

Comments

Search This Blog

Archive

Show more

Popular posts from this blog

TRIPASS XR تري باس

CELEPHI 200 MG, Gélule

ZENOXIA 15 MG, Comprimé

VOXCIB 200 MG, Gélule

Kana Brax Laberax

فومي كايند

بعض الادويه نجد رموز عليها مثل IR ، MR, XR, CR, SR , DS ماذا تعني هذه الرموز

NIFLURIL 700 MG, Suppositoire adulte

Antifongiques مضادات الفطريات

Popular posts from this blog

علاقة البيبي بالفراولة بالالفا فيتو بروتين

التغيرات الخمس التي تحدث للجسم عند المشي

إحصائيات سنة 2020 | تعداد سكَان دول إفريقيا تنازليا :

ما هو الليمونير للأسنان ؟

ACUPAN 20 MG, Solution injectable

CELEPHI 200 MG, Gélule

الام الظهر

VOXCIB 200 MG, Gélule

ميبستان

Popular posts from this blog

TRIPASS XR تري باس

CELEPHI 200 MG, Gélule

Popular posts from this blog

TRIPASS XR تري باس

CELEPHI 200 MG, Gélule

ZENOXIA 15 MG, Comprimé

VOXCIB 200 MG, Gélule

Kana Brax Laberax

فومي كايند

بعض الادويه نجد رموز عليها مثل IR ، MR, XR, CR, SR , DS ماذا تعني هذه الرموز

NIFLURIL 700 MG, Suppositoire adulte

Antifongiques مضادات الفطريات

Popular posts from this blog

Kana Brax Laberax

TRIPASS XR تري باس

PARANTAL 100 MG, Suppositoire بارانتال 100 مجم تحاميل

الكبد الدهني Fatty Liver

الم اسفل الظهر (الحاد) الذي يظهر بشكل مفاجئ bal-agrisi

SEDALGIC 37.5 MG / 325 MG, Comprimé pelliculé [P] سيدالجيك 37.5 مجم / 325 مجم ، قرص مغلف [P]

نمـو الدمـاغ والتطـور العقـلي لـدى الطفـل

CELEPHI 200 MG, Gélule

أخطر أنواع المخدرات فى العالم و الشرق الاوسط

Archive

Show more