Skip to main content Accessibility help
×
Hostname: page-component-848d4c4894-xm8r8 Total loading time: 0 Render date: 2024-06-29T07:42:04.410Z Has data issue: false hasContentIssue false

Section 5 - Disorders Involving Abnormal Coagulation

Published online by Cambridge University Press:  15 June 2018

Louis Caplan
Affiliation:
Beth Israel-Deaconess Medical Center, Boston
José Biller
Affiliation:
Loyola University Stritch School of Medicine, Chicago
Get access

Summary

Image of the first page of this content. For PDF version, please use the ‘Save PDF’ preceeding this image.'
Type
Chapter
Information
Publisher: Cambridge University Press
Print publication year: 2018

Access options

Get access to the full version of this content by using one of the access options below. (Log in options will check for institutional or personal access. Content may require purchase if you do not have access.)

References

References

Agnelli, G., Buller, H. R, Cohen, A., et al. 2013 Oral apixaban for the treatment of acute venous thromboembolism. N Engl J Med, 368, 799808.CrossRefGoogle Scholar
Alarcon-Segovia, D., Deleze, M., and Oria, C. V. 1989. Antiphospholipid antibodies and the antiphospholipid syndrome in systemic lupus erythematosus: A prospective analysis of 500 consecutive patients. Medicine, 68, 353–65.Google Scholar
Ameri, A. and Bousser, M. G. 1992. Cerebral venous thrombosis. Neurol Clin, 10, 87111.CrossRefGoogle ScholarPubMed
Andreoli, L., Chighizola, C. B., Banzato, A., et al. 2013. Estimated frequency of antiphospholipid antibodies in patients with pregnancy morbidity, stroke, myocardial infarction, and deep vein thrombosis: A critical review of the literature. Arthritis Care Res, 65, 1869–73.CrossRefGoogle ScholarPubMed
Antiphospholipid Antibodies in Stroke Study (APASS) Group. 1990. Clinical and laboratory findings in patients with antiphospholipid antibodies and cerebral ischemia. Stroke, 21, 1268–73.Google Scholar
Arnout, I. 1996. The pathogenesis of antiphospholipid antibody syndrome: Hypothesis based on parallelisms with heparin-induced thrombocytopenia. Thromb Haemost, 75, 536–41.Google ScholarPubMed
Arnout, J. and Vermylen, I. 1998. Mechanism of action of beta-2-glycoprotein-1-dependent lupus anticoagulants. Lupus, 7, S238.Google ScholarPubMed
Arnaud, L., Mathian, A., Ruffatti, A., et al. 2014. Efficacy of aspirin for the primary prevention of thrombosis in patients with antiphospholipid antibodies: an international and collaborative meta-anaylsis. Autoimmunity Reviews, 13, 281–91.Google Scholar
Asherson, R.A. 1992. The catastrophic antiphospholipid syndrome. J Rheumatol, 19, 506–12.Google Scholar
Asherson, R. and Cervera, R. 2003. Catastrophic antiphospholipid syndrome. Rheumatol Rep, 5, 395400.CrossRefGoogle ScholarPubMed
Atsumi, T., Ieko, M., Bertolaccini, M. L., et al. 2000. Association of autoantibodies against the phosphatidylserine–prothrombin complex with manifestations of the antiphospholipid syndrome and with the presence of lupus anticoagulant. Arthritis Rheum, 43, 1982–93.Google Scholar
Bauer, K. A. 2013. Pros and cons of new oral anticoagulants. Hematology, 2013, 464–70.CrossRefGoogle ScholarPubMed
Bauersachs, R., Berkowitz, S. D., Brenner, B., et al. 2010. Oral rivaroxaban for symptomatic venous thromboembolism (and supplementary appendix). N Engl J Med, 363, 2499–510.Google Scholar
Belvís, R., Santamaría, A., Martí-Fàbregas, J., et al. 2007. Patent foramen ovale and prothrombotic markers in young stroke patients. Blood Coagul Fibrinolysis, 18, 537–42.Google Scholar
Bertolaccini, M. L., Amengual, O., Andreoli, L., et al. 2014. 14th International Congress on Antiphospholipid Antibodies Task Force. Report on antiphospholipid syndrome laboratory diagnostics and trends. Autoimmunity Rev, 13, 917–30.Google Scholar
Bottin, L., Frances, C., de Zuttere, D., et al. 2015. Strokes in Sneddon syndrome without antiphospholipid antibodies. Ann Neurol, 77, 817–29.Google Scholar
Boles, J. and Mackman, N. 2010. Role of tissue factor in thrombosis in antiphospholipid antibody syndrome. Lupus, 19, 370–78.Google Scholar
Brey, R. L. 2004. Management of the neurological manifestations of APS: What do the trials tell us? Thromb Res, 114, 489–99.Google Scholar
Brey, R. L., Hart, R. G., Sherman, D. G., and Tegeler, C. T. 1990. Antiphospholipid antibodies and cerebral ischemia in young people. Neurology, 40, 1190–6.Google Scholar
Büller, H. R., Décousus, H., Grosso, M. A., et al. 2013. Edoxaban versus warfarin for the treatment of symptomatic venous thromboembolism. N Engl J Med, 369, 1406–15.Google Scholar
Campbell, A. L., Pierangeli, S. S., Wellhausen, S., and Harris, E. N. 1995. Comparison of the effects of anticardiolipin antibodies from patients with the antiphospholipid antibody syndrome and with syphilis on platelet activation and aggregation. Thromb Haemost, 73, 529–34.Google ScholarPubMed
Carhuapoma, J. R., Mitsias, R., and Levine, S. R. 1997. Cerebral venous thrombosis and anticardiolipin antibodies. Neurology, 28, 2363–9.Google Scholar
Casciola-Rosen, L., Rosen, A., Petri, M., and Schlissel, M. 1996. Surface blebs on apoptotic cells are sites of enhanced procoagulant activity implications for coagulation events and antigenic spread in systemic lupus erythematosus. Proc Natl Acad Sci USA, 93, 1624–9.CrossRefGoogle ScholarPubMed
Cervera, R., Piette, J. C., Font, J., et al. 2002. Antiphospholipid syndrome: Clinical and immunologic manifestations and patterns of disease expression in a cohort of 1000 patients. Arthritis Rheum, 46, 1019–27.Google Scholar
Cervera, R., Khamashta, M. A., Font, J., et al. 2003. Morbidity and mortality in systemic lupus erythematosus during a 10-year period: A comparison of early and late manifestations in a cohort of 1000 patients. Medicine, 82, 299308.Google Scholar
Cervera, R., Serrano, R., Pons-Estel, G. J. et al. 2015. Morbidity and mortality in the antiphospholiid syndrome during a 10-year period: A multicenter prospective study of 1000 patients. Ann Rheum Dis, 74, 1011–18.Google Scholar
Cesbron, J. Y., Amouyel, R., and Masy, E. 1997. Anticardiolipin antibodies and physical disability in the elderly. Ann Intern Med, 126, 1003.Google Scholar
Chapman, J., Abu-Katash, M., Inzelberg, R., et al. 2002. Prevalence and clinical features of dementia associated with the antiphospholipid syndrome and circulating anticoagulants. J Neurol Sci, 203–204, 81–4.Google Scholar
Chaturvedi, S., Jin, Z., Homma, S., et al., on behalf of the PICSS–APASS investigators. 2009. Patent foramen ovale, cardiac valve thickening, and antiphospholipid antibodies as risk factors for subsequent vascular events. The PICSS–APASS study. Stroke, 40, 2337–42.Google Scholar
Chinnery, P. E, Shaw, P. I., Ince, P. G., Jackson, G. H., and Bishop, R. I. 1997. Fulminant encephalopathy due to the catastrophic primary antiphospholipid syndrome. J Neurol Neurosurg Psychiatry, 62, 300–1.Google Scholar
Ciocca, R. G., Choi, J., and Graham, A. M. 1995. Antiphospholipid antibodies lead to increased risk in cardiovascular surgery. Am J Surg, 170, 198200.Google Scholar
Cojocaru, I. M., Cojocaru, M., Burcin, C., et al. 2007. Evaluation of antiphospholipid antibodies in young women with ischemic stroke. Romanian J Int Med, 45, 201–4.Google Scholar
Crowther, M. A., Ginsberg, J. S., Julian, J., et al. 2003. A comparison of two intensities of warfarin for the prevention of recurrent thrombosis in patients with the antiphospholipid antibody syndrome. [See comment] [erratum appears in N Engl J Med, 2003 Dec 25; 349, 2577]. N Engl J Med, 349, 1133–8.Google Scholar
de Moerloose, P., Boehlen, E., and Reber, G. 1997. Prevalence of anticardiolipin and antinuclear antibodies in an elderly hospitalized population and mortality after a 6-year follow-up. Age Aging, 27, 319–21.Google Scholar
Del Papa, N., Guidali, L., Sala, A., et al. 1997a. Endothelial cells as target for antiphospholipid antibodies. Arthritis Rheum, 40, 551–61.Google ScholarPubMed
Del Papa, N., Raschi, E., Catelli, L., et al. 1997. Endothelial cells as a target for antiphospholipid antibodies: Role of anti-beta-2-glycoprotein 1 antibodies. Am J Reprod Immunol, 38, 212–7.Google Scholar
Deschiens, M. A., Conard, J., Horellou, M. H., et al. 1996. Coagulation studies, factor V Leiden, and anticardiolipin antibodies in 40 cases of cerebral venous sinus thrombosis. Stroke, 27, 1724–30.Google Scholar
Drouvalakis, K. A. and Buchanan, T. T. C. 1998. Phospholipid specificity of autoimmune and drug induced lupus anticoagulants: Association of phos-phatidylethanolamine reactivity with thrombosis in autoimmune disease. J Rheumatol, 25, 290–5.Google Scholar
Erkan, D., Lockshin, M. D., and APS ACTION members. 2012. APS ACTION–Antiphospholipid Syndrome Alliance for Clinical Trials and International Networking. Lupus, 21, 695–98.Google Scholar
Erkan, D., Aguiar, C. L., Andrade, D., et al., 2014. 14th International Congress on Antiphospholipid Antibodies Task Force. Report on antiphospholipid syndrome treatment trends. Autoimmunity Rev, 13, 685–96.Google Scholar
Filippidis, E., Kapsalaki, G., Patramani, K., et al. 2009. Cerebral venous sinus thrombosis: Review of the demographics, pathophysiology, current diagnosis, and treatment. Neurosurgical Focus, 27, E3.Google Scholar
Galli, M., Comfurius, P., Maassen, C, et al. 1990. Anticardiolipin antibodies (ACA) directed not to cardiolipin but to a plasma protein cofactor. Lancet, 335, 1544–7.Google Scholar
Gharavi, A. E. and Pierangeli, S. S. 1998. Origin of antiphospholipid antibodies: Induction of aPL by viral peptides. Lupus, 7, S524.Google Scholar
Gharavi, A. E., Tang, H., Gharavi, E. E., Wilson, W. A., and Espinoza, L. R. 1995. Induction of aPL by immunization with a 15 amino acid peptide. Arthritis Rheum, 38, S296.Google Scholar
Gharavi, A. E., Tang, H., Gharavi, E. E., Espinoza, L. R., and Wilson, W. A. 1996. Induction of antiphospholipid antibodies by immunization with a viral peptide. Arthritis Rheum, 39, S319.Google Scholar
Hachulla, E., et al. 1998. Cerebral magnetic resonance imaging in patients with or without antiphospholipid antibodies. Lupus, 7, 124–31.Google Scholar
Hudson, M., Herr, A. L., Rauch, J., et al. 2003. The presence of multiple prothrombotic risk factors is associated with a higher risk of thrombosis in individuals with anticardiolipin antibodies. J Rheumatol, 30, 2385–91.Google Scholar
Hughes, G. R. V. 1983. Thrombosis, abortion, cerebral disease and lupus anticoagulant. Br Med J, 187, 1088–91.Google Scholar
Jacobson, M. W., Rappaport, L. J., Keenan, R. A., et al. 1999. Neuropsychological deficits associated with antiphospholipid antibodies. J Clin Exp Neuropsychol, 21, 251–64.Google Scholar
Janardhan, V., Wolf, P. A., Kase, C. S., et al. 2004. Anticardiolipin antibodies and risk of ischemic stroke and transient ischemic attack: the Framingham cohort and offspring study. Stroke, 35, 736–41.Google Scholar
Kalashnikova, L. A., Nasonov, E. L., Kushekbaeva, A. E., and Gracheva, L. A. 1990. Anticardiolipin antibodies in Sneddon’s syndrome. Neurology, 40, 464–7.Google Scholar
Khamashta, M. A., Cuadrado, M. J., Mujic, F., et al. 1995. The management of thrombosis in the antiphospholipid antibody syndrome. N Engl J Med, 332, 993–7.Google Scholar
Klemp, P., Cooper, R. C, Strauss, F. J., et al. 1988. Anticardiolipin antibodies in ischemic heart disease. Clin Exp Immunol, 74, 254–7.Google Scholar
Krnic-Barrie, S., Reister, O., Connor, C., et al. 1997. A retrospective review of 61 patients with antiphospholipid syndrome. Arch Intern Med, 157, 2101–8.Google Scholar
Labarca, J. A., Rabaggliati, R. M., Radrigan, F. J., et al. 1997. Antiphospholipid syndrome associated with cytomegalovirus infection: case report and review of the literature. Clin Infect Dis 24, 197200.CrossRefGoogle Scholar
Law, G., Magder, L., Fang, H., et al., 2014. Hydroxychloroquine reduces thrombosis (both arterial and venous) in systemic lupus erythematosus, particularly in antiphospholipid positive patients. Annals of the Rheumatic Diseases, 71, 547.Google Scholar
Levine, S. R., Brey, R. L., Salowich-Palm, L., Sawaya, K. L., and Havstad, S. 1993. Antiphospholipid antibody associated stroke: Prospective assessment of recurrent event risk. Stroke, 24, 188.Google Scholar
Levine, S. R., Brey, R. L., Sawaya, K. L., et al. 1995. Recurrent stroke and thrombo-occlusive events in the antiphospholipid syndrome. Ann Neurol, 38, 119–24.Google Scholar
Levine, S. R., Brey, R. L., Tilley, B. C, et al. 2004. Antiphospholipid antibodies and subsequent thrombo-occlusive events in patients with ischemic stroke. JAMA, 291, 576–84.Google Scholar
Levy, R. A., Awad, E., Olivera, J., and Porto, L. C. 1998. Placental pathology in antiphospholipid syndrome. Lupus, 7, S815.CrossRefGoogle ScholarPubMed
Loizou, S., McCrea, J. D., Rudge, A. C, et al. 1985. Measurement of anticardiolipin antibodies by an enzyme linked immunosorbent assay (ELISA): Standardization and quantitation of results. Clin Exp Immunol, 62, 738–45.Google Scholar
Lopez, L. R., Dier, K. J., Lopez, D., Merrill, J. T., Fink, C. A. 2004. Beta-2-glycoprotein 1 and antiphosphatidylserine antibodies are predictors of arterial thrombosis in patients with antiphospholipid antibody syndrome. Am J Clin Pathol 121, 142–9.Google Scholar
Marlar, R. A. and Neumann, A. 1990. Neonatal purpura fulminans due to homozygous protein C or protein S deficiencies. Semin Thromb Hemost, 16, 299309.Google Scholar
McIntyre, J. A., Wagenknecht, D. R., and Waxman, D. W. 2003. Frequency and specificities of antiphospholipid antibodies in volunteer blood donors. Immunobiol, 207, 5963.Google Scholar
McNeil, H. P., Simpson, R. J., Chesterman, C. N., and Krilis, S. A. 1990. Antiphospholipid antibodies are directed to a complex antigen that includes a lipid-binding inhibitor of coagulation: β2-glycoprotein I (apolipoprotein H). Proc Natl Acad Sci USA, 87, 4120–4.CrossRefGoogle ScholarPubMed
Meroni, P. L., Del Papa, N., Raschi, E., et al. 1998. β2 glycoprotein 1 as a co-factor for antiphospholipid reactivity with endothelial cells. Lupus, 7, S447.Google Scholar
Miyakis, S., Lockshin, M. D., Atsumi, T., et al. 2006. International consensus statement on an update of the classification criteria for definite antiphospholipid syndrome (APS). J Thromb Haemost, 4, 295306.Google Scholar
Molad, Y., Sidi, Y., Gornish, M., et al. 1992. Lupus anticoagulant: Correlation with magnetic resonance imaging of brain lesions. J Rheumatol, 19, 556–61.Google Scholar
Monroe, D. M. and Key, N. S. 2007. The tissue factor–factor VIIa complex: Procoagulant activity, regulation, and multitasking. J Thromb Haemost, 5, 1097–105.Google Scholar
Mosek, A., Yust, L., Trêves, T. A., et al. 2000 Dementia and antiphospholipid antibodies. Dement Geriatr Cogn Disord, 11, 36–8.Google Scholar
Nencini, R., Baruffi, M. C., Abbate, R., et al. 1992. Lupus anticoagulant and anticardiolipin antibodies in young adults with cerebral ischemia. Stroke, 23, 189–93.Google Scholar
Neville, C., Ruach, J., Kassis, J., et al. 2003. Thromboembolic risk in patients with high titre anticardiolipin and multiple antiphospholipid antibodies. Thromb Haemost, 90, 108–15.Google Scholar
Nojima, J., Suehisa, E., Akita, N., et al. 1997. Risk of arterial thrombosis in patients with anticardiolipin antibodies and lupus anticoagulant. Br J Haematol, 96, 447–50.Google Scholar
Nojima, J., Masuda, Y., Iwatani, Y., et al. 2008. Tissue factor expression on monocytes induced by antiphospholipid antibodies as a strong risk factor for thromboembolic complications in SLE patients. Biochem Biophys Res Commun, 365, 195200.Google Scholar
Otomo, K., Atsumi, T., Amengual, O., et al. 2012. Efficacy of the antiphospholipid score for the diagnosis of antiphospholipid syndrome and its predictive value for thrombotic events. Arthritis Rheumatism, 64, 504–12.Google Scholar
Pengo, V., Ruffatti, A., Legnani, C., et al. 2011. Incidence of a first thromboembolic event in asymptomatic carriers of high-risk antiphospholipid antibody profile: A multicenter prospective study. Thrombosis Hemostasis, 118, 4717–18.Google Scholar
Pezzini, A., Grassi, M., Lodigiani, C., et al. 2014. Predictors of long-term recurrent vascular events after ischemic stroke at young age. The Italian project on stroke in young adults. Circulation, 129, 1668–76.Google Scholar
Pierangeli, S. S., Girardi, G., Vega-Ostertag, M., et al. 2005. Requirement of activation of complement C3 and C5 for antiphospholipid antibody-mediated thrombophilia. Arthritis Rheum, 52, 2120–4.Google Scholar
Provenzale, J. M., Heinz, E. R., Ortel, T. L., et al. 1994. Antiphospholipid antibodies in patients without systemic lupus erythematosus: Neuroradiologic findings. Radiology, 192, 531–7.Google Scholar
Rand, J. H., Wu, X. X., Andree, H. A., et al. 1997. Pregnancy loss in the antiphospholipid antibody syndrome: A possible thrombogenic mechanism. N Engl J Med, 337, 154–60.Google Scholar
Rand, J. H., Wu, X. X., Quinn, A. S., et al. 2008. Hydroxychloroquine directly reduces the binding of antiphospholipid antibody–beta2-glycoprotein I complexes to phospholipid bilayers. Blood, 112, 1687–95.Google Scholar
Rosove, M. H. and Brewer, P. M. C. 1992. Antiphospholipid thrombosis: Clinical course after the first thrombotic event in 70 patients. Ann Intern Med, 117, 303–8.Google Scholar
Roubey, R. A. S. 1998. Mechanisms of autoantibody-mediated thrombosis. Lupus, 7, S1149.Google Scholar
Saidi, S., Mahjoub, T., and Almawi, W. Y. 2009. Lupus anticoagulants and antiphospholipid antibodies as risk factors for a first episode of ischemic stroke. J Thromb Haemost, 7, 1075–80.Google Scholar
Sailer, M., Burchert, W., Ehrenheim, C., et al. 1997. Positron emission tomography and magnetic resonance imaging for cerebral involvement in patients with systemic lupus erythematosus. J Neurol, 244, 186–93.Google Scholar
Schmidt, R., Auer-Grumbach, P., Fazekas, F., Offenbacher, H., and Kapeller, R. 1995. Anticardiolipin antibodies in normal subjects. Neuropsychological correlates and MRI findings. Stroke, 26, 749–54.Google Scholar
Schmidt-Tanguy, A., Voswinkel, J., Henrion, D., Subra, J., Loufrani, L., et al. 2013. Anti- thrombotic effects of hydroxychloroquine in primary antiphospholipid syndrome patients. J Thromb Haemost, 11, 1927–9.Google Scholar
Schulman, S., Kearon, C., Kakkar, A. K., et al. 2009. Dabigatran versus warfarin for the treatment of acute venous thromboembolism. N Engl Med, 361, 2341–52.Google Scholar
Sciascia, S., Cosseddu, D., Montaruli, B. et al. 2011. Risk scale for the diagnosis of antiphospholipid syndrome. Ann Rheum Dis, 70, 1517–18.Google Scholar
Sciascia, S., Sanna, G., Murru, V., et al. 2013. GAPPS: The global anti-phospholipid syndrome score. Rheumatology, 52, 1397–403.Google Scholar
Sciascia, S., Sanna, G., Khamashta, M. A., et al. 2015. The estimated frequency of antiphospholipid antibodies in young adults with cerebrovascular events: A systematic review. Ann Theum Dis, 74, 2028–33.Google ScholarPubMed
Shah, N. M., Khamashta, M. A., Atsumi, T., and Hughes, G. R. V. 1998. Outcome of patients with anticardiolipin antibodies: A 10 year follow-up of 52 patients. Lupus, 7, 36.Google Scholar
Shi, T., Iverson, G. M., Qi, J. C., et al. 2004. Beta 2-glycoprotein I binds factor XI and inhibits its activation by thrombin and factor XIIa: Loss of inhibition by clipped beta 2-glycoprotein I. Proc Natl Acad Sci U S A, 101, 3939 –44.Google Scholar
Sneddon, I. B. 1965. Cerebral vascular lesions in livedo reticularis. Br J Dermatol, 77, 180–5.Google Scholar
Tietjen, G. E., Day, M., Norris, L., et al. 1998. Role of anticardiolipin antibodies in young persons with migraine and transient focal neurologic events. Neurology, 50, 1433–40.CrossRefGoogle ScholarPubMed
Toubi, E., Khamashta, M. A., Panarra, A., and Hughes, G. R. V. 1995. Association of antiphospholipid antibodies with central nervous system disease in systemic lupus erythematosus. Am J Med, 99, 397401.Google Scholar
Tourbah, A., Peitte, J. C, Iba-Zizen, M., et al. 1997. The natural course of cerebral lesions in Sneddon syndrome. Arch Neurol, 54, 5360.Google Scholar
Tuhrim, S., Rand, J. H., Wu, X., et al. 1999. Antiphosphatidylserine antibodies are independently associated with ischemic stroke. Neurology, 53, 1523–7.Google Scholar
Urbanus, R. T., Siegerink, B., Roest, M., et al. 2009. Antiphospholipid antibodies and risk of myocardial infarction and ischaemic stroke in young women in the RATIO study: A case-control study. Lancet Neuro, 8, 9981005.CrossRefGoogle ScholarPubMed
van Goor, M. R, Alblas, C. L., Leebeek, E W., Koudstaal, P. J., and Dippel, D. W. 2004. Do antiphospholipid antibodies increase the long-term risk of thrombotic complications in young patients with a recent TLA or ischemic stroke? Acta Neurol Scand, 109, 410–5.Google Scholar
Vega-Ostertag, M. E., Ferrara, D. E., Romay-Penabad, Z., et al. 2007. Role of p38 mitogen-activated protein kinase in antiphospholipid antibody-mediated thrombosis and endothelial cell activation. J Thromb Haemost, 5, 18281834.Google Scholar
Vermylen, J., Van Geet, C, and Arnout, J. 1998. Antibody-mediated thrombosis: Relation to the antiphospholipid syndrome. Lupus, 7, S636.Google Scholar
Vila, P., Hernandez, M. C., Lopez-Fernandez, M. E., and Baffle, J. 1994. Prevalence, follow-up and clinical significance of the anticardiolipin antibodies in normal subjects. Thromb Haemost, 72, 209–13.Google Scholar
Wahl, D. G., Guillemin, F., de Maistre, E., et al. 1998. Meta-analysis of the risk of venous thrombosis in individuals with antiphospholipid antibodies without underlying autoimmune disease or previous thrombosis. Lupus, 7, 1522.Google Scholar
Wang, L., Chen, H., Zhang, Y., et al. 2015. Clinical characteristics of cerebral venous sinus thrombosis in patients with systemic lupus erythematosus: A single-centre experience in China. J Immunol Res, 2015, 17.Google Scholar
Zeiger, B., Sepp, N., Stockhammer, G., et al. 1993. Sneddon’s syndrome: A long term follow-up of 21 patients. Arch Dermatol, 129, 437–44.Google Scholar
Ziporen, L. and Shoenfeld, Y. 1998. Antiphospholipid syndrome: From patient’s bedside to experimental animal models and back to the patient’s bedside. Hematol Cell Ther, 40, 175–82.Google Scholar
Zuily, S., Regnault, V., Selton-Sulty, C., et al. 2011. Increased risk for heart valve disease associated with antiphospholipid antibodies in patients with systemic lupus erythematosus: Meta-analysis of echocardiographic studies. Circulation, 124, 215–24.Google Scholar

References

Hardaway, R. M. Disseminated intravascular coagulation syndromes. Arch. Surg. Chic. Ill. 1960 83, 842–50 (1961).Google Scholar
Goodnight, S. H. et al. Defibrination after brain-tissue destruction: A serious complication of head injury. N. Engl. J. Med. 290, 1043–7 (1974).CrossRefGoogle ScholarPubMed
Gando, S., Nanzaki, S., Sasaki, S. & Kemmotsu, O. Significant correlations between tissue factor and thrombin markers in trauma and septic patients with disseminated intravascular coagulation. Thromb. Haemost. 79, 1111–15 (1998).Google Scholar
Gando, S. Disseminated intravascular coagulation in trauma patients. Semin. Thromb. Hemost. 27, 585–92 (2001).CrossRefGoogle ScholarPubMed
García-Avello, A. et al. Degree of hypercoagulability and hyperfibrinolysis is related to organ failure and prognosis after burn trauma. Thromb. Res. 89, 5964 (1998).Google Scholar
Barbui, T. & Falanga, A. Disseminated intravascular coagulation in acute leukemia. Semin. Thromb. Hemost. 27, 593604 (2001).Google Scholar
Arai, A. et al. Cardiac malignant pheochromocytoma with bone metastases. Intern. Med. Tokyo Jpn. 37, 940–4 (1998).Google Scholar
Letsky, E. A. Disseminated intravascular coagulation. Best Pract. Res. Clin. Obstet. Gynaecol. 15, 623–44 (2001).Google Scholar
Hossain, N. & Paidas, M. J. Disseminated intravascular coagulation. Semin. Perinatol. 37, 257–66 (2013).Google Scholar
Breakwell, L. M., Getty, C. J., & Austin, C. Disseminated intravascular coagulation in elective primary total hip replacement. J. Arthroplasty 14, 239–42 (1999).Google Scholar
Asherson, R. A. The catastrophic antiphospholipid syndrome, 1998. A review of the clinical features, possible pathogenesis and treatment. Lupus 7, S5562 (1998).Google Scholar
Katsaros, D. & Grundfest-Broniatowski, S. Successful management of visceral Klippel–Trenaunay–Weber syndrome with the antifibrinolytic agent tranexamic acid (cyclocapron): A case report. Am. Surg. 64, 302–4 (1998).Google Scholar
Persoons, M. C., Stals, F. S., van dam Mieras, M. C., & Bruggeman, C. A. Multiple organ involvement during experimental cytomegalovirus infection is associated with disseminated vascular pathology. J. Pathol. 184, 103–9 (1998).Google Scholar
Lifshitz, M., Kapelushnik, J., Ben-Harosh, M., & Sofer, S. Disseminated intravascular coagulation after Cerastes vipera envenomation in a 3-year-old child: A case report. Toxicon Off. J. Int. Soc. Toxinology 38, 1593–8 (2000).Google Scholar
Bey, T. A. et al. Loxosceles arizonica bite associated with shock. Ann. Emerg. Med. 30, 701–3 (1997).Google Scholar
Jung, J. W. et al. A fatal case of intravascular coagulation after bee sting acupuncture. Allergy Asthma Immunol. Res. 4, 107–9 (2012).Google Scholar
Dhanapriya, J. et al. Acute kidney injury and disseminated intravascular coagulation due to mercuric chloride poisoning. Indian J. Nephrol. 26, 206 (2016).Google Scholar
Felcher, A. et al. Disseminated intravascular coagulation and status epilepticus. Neurology 51, 629–31 (1998).Google Scholar
Bick, R. L. Disseminated intravascular coagulation. Hematol. Oncol. Clin. North Am. 6, 1259–85 (1992).Google Scholar
Gando, S., Levi, M., & Toh, C.-H. Disseminated intravascular coagulation. Nat. Rev. Dis. Primer 2, 16037 (2016).Google Scholar
Dhainaut, J.-F., Macias, W. L., & Nelson, D. R. Would patients with more subtle signs of coagulopathy have benefited from treatment with activated protein C? Crit. Care Med. 33, 1670–1 (2005).Google Scholar
Taylor, F. B. et al. Towards definition, clinical and laboratory criteria, and a scoring system for disseminated intravascular coagulation. Thromb. Haemost. 86, 1327–30 (2001).Google Scholar
Favaloro, E. J. & Adcock, D. M. Standardization of the INR: How good is your laboratory’s INR and can it be improved? Semin. Thromb. Hemost. 34, 593603 (2008).Google Scholar
Gando, S. et al. A multicenter, prospective validation of disseminated intravascular coagulation diagnostic criteria for critically ill patients: Comparing current criteria. Crit. Care Med. 34, 625–31 (2006).Google Scholar
Prechel, M. & Walenga, J. M. The laboratory diagnosis and clinical management of patients with heparin-induced thrombocytopenia: An update. Semin. Thromb. Hemost. 34, 8696 (2008).Google Scholar
Selleng, K., Selleng, S., & Greinacher, A. Heparin-induced thrombocytopenia in intensive care patients. Semin. Thromb. Hemost. 34, 425–38 (2008).Google Scholar
Schwartzman, R. J. & Hill, J. B. Neurologic complications of disseminated intravascular coagulation. Neurology 32, 791–7 (1982).Google Scholar
Shebuski, R. J. & Kilgore, K. S. Role of inflammatory mediators in thrombogenesis. J. Pharmacol. Exp. Ther. 300, 729–35 (2002).Google Scholar
Wada, H. et al. Hemostatic molecular markers before the onset of disseminated intravascular coagulation. Am. J. Hematol. 60, 273–8 (1999).Google Scholar
Favaloro, E. J. Laboratory testing in disseminated intravascular coagulation. Semin. Thromb. Hemost. 36, 458–67 (2010).Google Scholar
Levi, M. & van der Poll, T. The role of natural anticoagulants in the pathogenesis and management of systemic activation of coagulation and inflammation in critically ill patients. Semin. Thromb. Hemost. 34, 459–68 (2008).Google Scholar
de Jonge, E., van der Poll, T., Kesecioglu, J. & Levi, M. Anticoagulant factor concentrates in disseminated intravascular coagulation: Rationale for use and clinical experience. Semin. Thromb. Hemost. 27, 667–74 (2001).Google Scholar
Naidech, A. M. & Kumar, M. A., and the participants in the International Multidisciplinary Consensus Conference on Multimodality Monitoring. Monitoring of hematological and hemostatic parameters in neurocritical care patients. Neurocrit. Care 21, 168–76 (2014).Google Scholar
Johansson, P. I., Stensballe, J., Vindeløv, N., Perner, A., & Espersen, K. Hypocoagulability, as evaluated by thromboelastography, at admission to the ICU is associated with increased 30-day mortality. Blood Coagul. Fibrinolysis 21, 168–74 (2010).Google Scholar
Park, M. S. et al. Thromboelastography as a better indicator of hypercoagulable state after injury than prothrombin time or activated partial thromboplastin time. J. Trauma 67, 266–76 (2009).Google Scholar
Bakhtiari, K., Meijers, J. C. M., de Jonge, E., & Levi, M. Prospective validation of the International Society of Thrombosis and Haemostasis scoring system for disseminated intravascular coagulation. Crit. Care Med. 32, 2416–21 (2004).Google Scholar
Gando, S. et al. Evaluation of new Japanese diagnostic criteria for disseminated intravascular coagulation in critically ill patients. Clin. Appl. Thromb. Off. J. Int. Acad. Clin. Appl. Thromb. 11, 71–6 (2005).Google Scholar
Sivula, M., Tallgren, M., & Pettilä, V. Modified score for disseminated intravascular coagulation in the critically ill. Intensive Care Med. 31, 1209–14 (2005).Google Scholar
Cauchie, P. et al. Diagnosis and prognosis of overt disseminated intravascular coagulation in a general hospital – meaning of the ISTH score system, fibrin monomers, and lipoprotein-C-reactive protein complex formation. Am. J. Hematol. 81, 414–19 (2006).Google Scholar
Umemura, Y. et al. Design and evaluation of new unified criteria for disseminated intravascular coagulation based on the Japanese Association for Acute Medicine criteria. Clin. Appl. Thromb. Off. J. Int. Acad. Clin. Appl. Thromb. 22, 153–60 (2016).Google Scholar
Angstwurm, M. W. A., Dempfle, C.-E., & Spannagl, M. New disseminated intravascular coagulation score: A useful tool to predict mortality in comparison with Acute Physiology and Chronic Health Evaluation II and Logistic Organ Dysfunction scores. Crit. Care Med. 34, 314–20; quiz 328 (2006).Google Scholar
Toh, C. H. & Downey, C. Performance and prognostic importance of a new clinical and laboratory scoring system for identifying non-overt disseminated intravascular coagulation. Blood Coagul. Fibrinolysis Int. J. Haemost. Thromb. 16, 6974 (2005).Google Scholar
Cartin-Ceba, R. et al. Epidemiology of critical care syndromes, organ failures, and life-support interventions in a suburban US community. Chest 140, 1447–55 (2011).Google Scholar
Gando, S. et al. Natural history of disseminated intravascular coagulation diagnosed based on the newly established diagnostic criteria for critically ill patients: Results of a multicenter, prospective survey. Crit. Care Med. 36, 145–50 (2008).Google Scholar
Wheeler, A. P. & Bernard, G. R. Treating patients with severe sepsis. N. Engl. J. Med. 340, 207–14 (1999).Google Scholar
Levi, M., de Jonge, E., & van der Poll, T. Sepsis and disseminated intravascular coagulation. J. Thromb. Thrombolysis 16, 43–7 (2003).Google Scholar
Lavrentieva, A. et al. Early coagulation disorders after severe burn injury: Impact on mortality. Intensive Care Med. 34, 700–6 (2008).Google Scholar
Barret, J. P. & Gomez, P. A. Disseminated intravascular coagulation: A rare entity in burn injury. Burns J. Int. Soc. Burn Inj. 31, 354–7 (2005).Google Scholar
Singh, B. et al. Trends in the incidence and outcomes of disseminated intravascular coagulation in critically ill patients (2004–2010): A population-based study. Chest 143, 1235–42 (2013).Google Scholar
Ranieri, V. M. et al. Drotrecogin alfa (activated) in adults with septic shock. N. Engl. J. Med. 366, 2055–64 (2012).Google Scholar
Murata, A., Okamoto, K., Mayumi, T., Muramatsu, K. & Matsuda, S. The recent time trend of outcomes of disseminated intravascular coagulation in Japan: An observational study based on a national administrative database. J. Thromb. Thrombolysis 38, 364–71 (2014).Google Scholar
Hoffman, M. & Monroe, D. M. A cell-based model of hemostasis. Thromb. Haemost. 85, 958–65 (2001).Google Scholar
Franco, R. F. et al. The in vivo kinetics of tissue factor messenger RNA expression during human endotoxemia: Relationship with activation of coagulation. Blood 96, 554–9 (2000).Google Scholar
Levi, M. & van der Poll, T. Inflammation and coagulation. Crit. Care Med. 38, S2634 (2010).Google Scholar
Rauch, U. et al. Transfer of tissue factor from leukocytes to platelets is mediated by CD15 and tissue factor. Blood 96, 170–5 (2000).Google Scholar
Zimmerman, G. A., McIntyre, T. M., Prescott, S. M., & Stafforini, D. M. The platelet-activating factor signaling system and its regulators in syndromes of inflammation and thrombosis. Crit. Care Med. 30, S294301 (2002).Google Scholar
Bockmeyer, C. L. et al. Inflammation-associated ADAMTS 13 deficiency promotes formation of ultra-large von Willebrand factor. Haematologica 93, 137–40 (2008).Google Scholar
Osterud, B. & Bjørklid, E. The production and availability of tissue thromboplastin in cellular populations of whole blood exposed to various concentrations of endotoxin. An assay for detection of endotoxin. Scand. J. Haematol. 29, 175–84 (1982).Google Scholar
Engelmann, B. & Massberg, S. Thrombosis as an intravascular effector of innate immunity. Nat. Rev. Immunol. 13, 3445 (2013).Google Scholar
Müller, I. et al. Intravascular tissue factor initiates coagulation via circulating microvesicles and platelets. FASEB J. Off. Publ. Fed. Am. Soc. Exp. Biol. 17, 476–8 (2003).Google Scholar
Fuchs, T. A. et al. Extracellular DNA traps promote thrombosis. Proc. Natl. Acad. Sci. USA 107, 15880–5 (2010).Google Scholar
von Brühl, M.-L. et al. Monocytes, neutrophils, and platelets cooperate to initiate and propagate venous thrombosis in mice in vivo. J. Exp. Med. 209, 819–35 (2012).Google Scholar
Massberg, S. et al. Reciprocal coupling of coagulation and innate immunity via neutrophil serine proteases. Nat. Med. 16, 887–96 (2010).Google Scholar
Franchini, M. Pathophysiology, diagnosis and treatment of disseminated intravascular coagulation: An update. Clin. Lab. 51, 633–9 (2005).Google Scholar
Lineaweaver, W., Franzini, D., Dragonetti, D., McCarley, D., & Rumley, T. Haemophilus influenzae meningitis and Waterhouse–Friderichsen syndrome in an adult. South. Med. J. 79, 1034–6 (1986).Google Scholar
Ohashi, R. et al. Acute renal failure as the presenting sign of disseminated intravascular coagulation in a patient with metastatic prostate cancer. Int. J. Nephrol. Renov. Dis. 6, 4751 (2013).Google Scholar
Bick, R. L. Disseminated intravascular coagulation: Pathophysiological mechanisms and manifestations. Semin. Thromb. Hemost. 24, 318 (1998).Google Scholar
Collins, R. C., Al-Mondhiry, H., Chernik, N. L., & Posner, J. B. Neurologic manifestations of intravascular coagulation in patients with cancer. A clinicopathologic analysis of 12 cases. Neurology 25, 795806 (1975).Google Scholar
Graus, F., Rogers, L. R., & Posner, J. B. Cerebrovascular complications in patients with cancer. Medicine (Baltimore) 64, 1635 (1985).Google Scholar
Wijdicks, E. F. & Scott, J. P. Stroke in the medical intensive-care unit. Mayo Clin. Proc. 73, 642–6 (1998).Google Scholar
Navi, B. B. et al. Intracerebral and subarachnoid hemorrhage in patients with cancer. Neurology 74, 494501 (2010).Google Scholar
Goodnight, S. H. Bleeding and intravascular clotting in malignancy: A review. Ann. N. Y. Acad. Sci. 230, 271–88 (1974).Google Scholar
Pisoni, R., Ruggenenti, P. & Remuzzi, G. Drug-induced thrombotic microangiopathy: Incidence, prevention and management. Drug Saf. 24, 491501 (2001).Google Scholar
Nguyen, T. C. & Carcillo, J. A. Bench-to-bedside review: Thrombocytopenia-associated multiple organ failure – a newly appreciated syndrome in the critically ill. Crit. Care Lond. Engl. 10, 235 (2006).Google Scholar
Johnson, R. T. & Richardson, E. P. The neurological manifestations of systemic lupus erythematosus. Medicine (Baltimore) 47, 337–69 (1968).Google Scholar
Musio, F., Bohen, E. M., Yuan, C. M., & Welch, P. G. Review of thrombotic thrombocytopenic purpura in the setting of systemic lupus erythematosus. Semin. Arthritis Rheum. 28, 119 (1998).Google Scholar
Yasuda, S., Bohgaki, M., Atsumi, T. & Koike, T. Pathogenesis of antiphospholipid antibodies: Impairment of fibrinolysis and monocyte activation via the p38 mitogen-activated protein kinase pathway. Immunobiology 210, 775–80 (2005).Google Scholar
Galli, M. Treatment of the antiphospholipid syndrome. Auto- Immun. Highlights 5, 17 (2014).Google Scholar
Cervera, R. & Espinosa, G. Update on the catastrophic antiphospholipid syndrome and the ‘CAPS Registry’. Semin. Thromb. Hemost. 38, 333–8 (2012).Google ScholarPubMed
van Runnard Heimel, P. J. et al. A randomised placebo-controlled trial of prolonged prednisolone administration to patients with HELLP syndrome remote from term. Eur. J. Obstet. Gynecol. Reprod. Biol. 128, 187193 (2006).Google Scholar
Pijnenborg, R., Vercruysse, L., Hanssens, M. & Brosens, I. Endovascular trophoblast and preeclampsia: A reassessment. Pregnancy Hypertens. 1, 6671 (2011).Google Scholar
Warkentin, T. Heparin-induced thrombocytopenia in critically ill patients. Semin. Thromb. Hemost. 41, 049–60 (2015).Google Scholar
Warkentin, T. E., Sheppard, J.-A. I., Moore, J. C., Cook, R. J., & Kelton, J. G. Studies of the immune response in heparin-induced thrombocytopenia. Blood 113, 4963–9 (2009).Google Scholar
Hanly, E. J., Cohn, E. J., Johnson, J. L., & Peyton, B. D. DIC: Treatment frontiers. Curr. Surg. 59, 257–64 (2002).Google Scholar
Levi, M., Toh, C. H., Thachil, J., & Watson, H. G. Guidelines for the diagnosis and management of disseminated intravascular coagulation. British Committee for Standards in Haematology. Br. J. Haematol. 145, 2433 (2009).Google Scholar
Wada, H. et al. Expert consensus for the treatment of disseminated intravascular coagulation in Japan. Thromb. Res. 125, 611 (2010).Google Scholar
Kawasugi, K. et al. Prospective evaluation of hemostatic abnormalities in overt DIC due to various underlying diseases. Thromb. Res. 128, 186–90 (2011).Google Scholar
Alving, B. M., Spivak, J. L., DeLoughery, T. G. Consultative hematology: Hemostasis and transfusion issues in surgery and critical care medicine. Hematology Am. Soc. Hematol. Educ. Program 320–41, 1998.Google Scholar
Slichter, S. J. Evidence-based platelet transfusion guidelines. Hematol. Am. Soc. Hematol. Educ. Program 172–78 (2007). doi:10.1182/asheducation-2007.1.172Google Scholar
Wada, H. et al. Guidance for diagnosis and treatment of DIC from harmonization of the recommendations from three guidelines. J. Thromb. Haemost. (2013). doi:10.1111/jth.12155.Google Scholar
Sakuragawa, N., Hasegawa, H., Maki, M., Nakagawa, M. & Nakashima, M. Clinical evaluation of low-molecular-weight heparin (FR-860) on disseminated intravascular coagulation (DIC): A multicenter co-operative double-blind trial in comparison with heparin. Thromb. Res. 72, 475500 (1993).Google Scholar
Simpson, E. et al. Recombinant factor VIIa for the prevention and treatment of bleeding in patients without haemophilia. Cochrane Database Syst. Rev. CD005011 (2012). doi:10.1002/14651858.CD005011.pub4.Google Scholar
Liu, X.-L. et al. Low-dose heparin as treatment for early disseminated intravascular coagulation during sepsis: A prospective clinical study. Exp. Ther. Med. 7, 604–8 (2014).Google Scholar
Corrigan, J. J. & Jordan, C. M. Heparin therapy in septicemia with disseminated intravascular coagulation: Effect on mortality and on correction of hemostatic defects. N. Engl. J. Med. 283, 778–82 (1970).Google Scholar
Gando, S., Sawamura, A., & Hayakawa, M. Trauma, shock, and disseminated intravascular coagulation: Lessons from the classical literature. Ann. Surg. 254, 1019 (2011).Google Scholar
Vincent, J.-L. et al. A randomized, double-blind, placebo-controlled, phase 2b study to evaluate the safety and efficacy of recombinant human soluble thrombomodulin, ART-123, in patients with sepsis and suspected disseminated intravascular coagulation. Crit. Care Med. 41, 2069–79 (2013).Google Scholar
Jaimes, F. et al. Unfractioned heparin for treatment of sepsis: A randomized clinical trial (the HETRASE study). Crit. Care Med. 37, 1185–96 (2009).Google Scholar
Okajima, K. & Uchiba, M. The anti-inflammatory properties of antithrombin III: new therapeutic implications. Semin. Thromb. Hemost. 24, 2732 (1998).Google Scholar
Fourrier, F., Jourdain, M. & Tournoys, A. Clinical trial results with antithrombin III in sepsis. Crit. Care Med. 28, S3843 (2000).Google Scholar
Saito, H. et al. Efficacy and safety of recombinant human soluble thrombomodulin (ART-123) in disseminated intravascular coagulation: Results of a phase III, randomized, double-blind clinical trial. J. Thromb. Haemost. 5, 3141 (2007).Google Scholar
Yamakawa, K. et al. Treatment effects of recombinant human soluble thrombomodulin in patients with severe sepsis: A historical control study. Crit. Care Lond. Engl. 15, R123 (2011).Google Scholar
Abraham, E. et al. Drotrecogin alfa (activated) for adults with severe sepsis and a low risk of death. N. Engl. J. Med. 353, 1332–41 (2005).Google Scholar
Abraham, E. et al. Assessment of the safety of recombinant tissue factor pathway inhibitor in patients with severe sepsis: A multicenter, randomized, placebo-controlled, single-blind, dose escalation study. Crit. Care Med. 29, 2081–9 (2001).Google Scholar
CRASH-2 trial collaborators et al. Effects of tranexamic acid on death, vascular occlusive events, and blood transfusion in trauma patients with significant haemorrhage (CRASH-2): A randomised, placebo-controlled trial. Lancet Lond. Engl. 376, 2332 (2010).Google Scholar
CRASH-2 Collaborators, Intracranial Bleeding Study. Effect of tranexamic acid in traumatic brain injury: A nested randomised, placebo controlled trial (CRASH-2 Intracranial Bleeding Study). BMJ 343, d3795 (2011).Google Scholar

References

Ansell, JE. 2016. Universal, class-specific and drug-specific reversal agents for the new oral anticoagulants. J Thromb Thrombolyisis 41: 248–52.Google Scholar
Baharoglu, MI, Cordonnier, C, Al-Shai Salman, R, et al. 2016. Platelet transfusion versus standard care after acute stroke due to spontaneous cerebral hemorrhage associated with antiplatelet therapy (PATCH): A randomized, open-label, phase 3 trial. Lancet 387: 2605–13.Google Scholar
Casari, C, Bergmeier, W. 2016. Acquired platelet disorders. Thromb Res 141: S735.Google Scholar
Cervera, A, Amaro, S, Chamorro, A. 2012. Oral anticoagulant-associated intracerebral hemorrhage. J Neurol 259: 212–24.Google Scholar
Chen, C-Y, Tai, C-H, Tsay, W, et al. 2009. Prediction of fatal intracranial hemorrhage in patients with acute myeloid leukemia. Annals Oncol 20: 1100–4.Google Scholar
Chen, C-Y, Tai, C-H, Cheng, A et al. 2012. Intracranial hemorrhage in adult patients with hematologic malignancies. BMC Medicine 10: 97. doi: 10.1186/1741–7015–10–97.Google Scholar
Chern, JJ, Tsung, AJ, Humphries, W, et al. 2011. Clinical outcome of leukemia patients with intracranial hemorrhage. Clinical article. J Neurosurg 115: 268–72.Google Scholar
Coppola, A, Tagliaferri, A, Franchini, M. 2016. Controversies in inherited bleeding disorders. Semin Thromb Hemost 42: 459–62.Google Scholar
Diringer, MN, Skolnick, BE, Mayer, SA, et al. 2010. Thromboembolic events with recombinant activated factor VII in spontaneous intracerebral hemorrhage: results from the factor seven for acute hemorrhagic stroke (FAST) trial. Stroke 41: 4853.Google Scholar
Emiru, T, Bershad, EM, Zantek, ND, et al. 2012. Intracerebral hemorrhage: A review of coagulation function. Clin Appl Thromb Hemost. doi: 0.1177/1076029612454938.Google Scholar
Estcourt, LJ, Stanworth, SJ, Collett, D, et al. 2014. Intracranial hemorrhage in thrombocytopenic hematology patients – a nested case-control study: The InCiTe study protocol. BMJ Open 4: e004199.Google Scholar
Flaherty, ML, Tao, H, Haverbusch, M, et al. 2008. Warfarin use leads to larger intracerebral hematomas. Neurology 71: 1084–9.Google Scholar
Foerch, C, Sitzer, M, Steinmetz, H, et al. 2006 Pretreatment with antiplatelet agents is not independently associated with unfavorable outcome in intracerebral hemorrhage. Stroke 37: 2165–7.Google Scholar
Goldszmidt, AJ, Caplan, LR. 2010. Stroke Essentials, 2nd edn. Sudbury, MA: Jones & Bartlett Publishers.Google Scholar
Gonzalez-Duarte, A, Garcia-Ramos, GS, Valdes-Ferrer, SI, et al. 2008 Clinical description of intracranial hemorrhage associated with bleeding disorders. J Stroke Cerebrovasc Dis 17: 204–7.Google Scholar
Granger, CB, Alexander, JH, McMurray, JJ, et al. 2011. Apixaban versus warfarin in patients with atrial fibrillation. N Engl J Med 365: 981–92.Google Scholar
Gresele, P, Bury, L, Falcinelli, E. 2016. Inherited platelet function disorders: Algorithms for phenotypic and genetic investigation. Semin Thromb Hemost 42: 292305.Google Scholar
Lauer, A, Pfeilschifter, W, Schaffer, CB, et al. 2013. Intracerebral hemorrhage associated with antithrombotic treatment: Translational insights from experimental studies. Lancet Neurol 12: 394405.Google Scholar
Maas, MB, Forsenbery, NF, Kosteva, AR, et al. 2013. Coagulopathy disproportionately predisposes to lobar intracerebral hemorrhage. Neurocrit Care 18: 166–9.Google Scholar
Naidech, AM, Bernstein, RA, Levasseur, K, et al. 2009a. Platelet activity and outcome after intracerebral hemorrhage. Ann Neurol 65: 352–6.Google Scholar
Naidech, AM, Jovanovic, B, Liebling, S, et al. 2009b. Reduced platelet activity is associated with early clot growth and worse 3-month outcome after intracerebral hemorrhage. Stroke 40: 2398–401.Google Scholar
National Institute of Neurological Disorders and Stroke rt-PA Stroke Study Group. 1995. N Engl J Med 333: 1581–7.Google Scholar
O’Carroll, CB, Aguilar, MI. 2015. Management of post-thrombolysis hemorrhagic and orolingual angioedema complications. Neurohospitalist 5: 133–41.Google Scholar
Patel, MR, Mahaffey, KW, Garg, J, et al. 2011. Rivaroxaban versus warfarin in nonvalvular atrial fibrillation. N Engl J Med 365: 883–91.Google Scholar
Pezzini, A, Grassi, M, Paciaroni, M, et al. 2014. Antithrombotic medications and the etiology of intracerebral hemorrhage: MUCH-Italy. Neurology 82: 529–35.Google Scholar
Pollack, CV, Reilly, PA, Eikelborn, J, et al. 2015. Idarucizumab for dabigatran reversal. N Engl J Med 373: 511–20.Google Scholar
Purrucker, JC, Haas, K, Rizos, T, et al. 2016. Early clinical and radiological course, management, and outcome of intracerebral hemorrhage related to new oral anticoagulants. JAMA Neurol 73: 169–77.Google Scholar
Quiñones-Hinojosa, A, Gulati, M, Singh, V, et al. 2003. Spontaneous intracerebral hemorrhage due to coagulation disorders. Neurosurg Focus 15: E3.Google Scholar
Saloheimo, P, Ahonen, M, Juvela, S, et al. 2006. Regular aspirin-use preceding the onset of primary intracerebral hemorrhage is an independent predictor for death. Stroke 37: 129–33.Google Scholar
Sandrock-Lang, K, Wentzell, R, Santoso, S, et al. 2015. Inherited platelet disorders. Hamostaseologie 35: 2.Google Scholar
Sterrenberg, D, Nand, S. 2016. Congenital coagulation disorders. In Anticoagulation and Hemostasis in Neurosurgery, 1st edn, ed. Loftus, C. M.. Switzerland: Springer International Publishing, pp. 93114.Google Scholar
Wijman, CA. 2009. Is platelet activity important in acute intracerebral hemorrhage? Neurocrit Care 11: 305–6.Google Scholar
Wilson, D, Charidimou, A, Shakeshaft, C, et al. 2016. Volume and functional outcome of intracerebral hemorrhage according to oral anticoagulant type. Neurology 86: 360–6.Google Scholar
Yasaka, M, Sakata, T, Naritomi, H, et al. 2005. Optimal dose of prothrombin complex concentrate for acute reversal of oral anticoagulation. Thromb Res 115: 455–59.Google Scholar
Yates, S, Sarode, R. 2015. Reversal of anticoagulant effects in patients with intracerebral hemorrhage. Curr Neurol Neurosci Rep 15: 110.Google Scholar

References

Mannucci, PM. Laboratory detection of inherited thrombophilia: A historical perspective. Semin Thromb Hemost 2005; 32: 510.Google Scholar
Ma, K, Wells, P, Guzman, C, et al. A multicenter prospective study of risk factors and treatment of unusual site thrombosis. Thromb Res 2016; 144: 100–5.Google Scholar
Guria, K, Guria, GT. Spatial aspects of blood coagulation: Two decades of the self-sustained traveling wave of thrombin. Thromb Res 2015; 135: 423–33.Google Scholar
Boilard, E, Duchez, A-C, Brisson, A. The diversity of platelet microparticles. Curr Opin Hematol 2015; 22: 437–44.Google Scholar
van Roos, M-J, Pretorius, E. Metabolic syndrome, platelet activation and development of transient ischemic attack or thromboembolic stroke. Thromb Res 2015; 135: 434–42.Google Scholar
Yau, JW, Teoh, H, Verma, S. Endothelial control of thrombosis. BMC Cardiovasc Disord 2015; 15: 130.Google Scholar
van Hinsbergh, VWM. Endothelium: Role in regulation of coagulation and inflammation. Semin Immunopathol 2012; 34: 93106.Google Scholar
Tjarnlund-Wolf, A, Brogren, H, Lo, EH, Wang, X. Plasminogen activator inhibitor-1 and thrombotic cerebrovascular diseases. Stroke 2012; 43: 2833–9.CrossRefGoogle ScholarPubMed
Aleman, MM, Walton, BL, Byrnes, JR, Wolberg, AS. Fibrinogen and red blood cells in venous thrombosis. Thromb Res 2014; 133: S3840.Google Scholar
Davies, PF. Hemodynamic shear stress and the endothelium in cardiovascular pathophysiology. Nat Clin Pract Cardiovasc Med 2009; 6: 1626.Google Scholar
Wardlaw, JW, Smith, C, Dichgans, M. Mechanisms underlying sporadic cerebral small vessel disease: Insights from neuroimaging. Lancet Neurol 2013; 12: 483–97.Google Scholar
Fisher, CM. Cerebral military aneurysms in hypertension. Am J Pathol 1972; 66: 313–30.Google Scholar
Fisher, MJ. Brain regulation of thrombosis and hemostasis: From theory to practice. Stroke 2013; 44: 3275–85.Google Scholar
Szymanska, M, Alhenc-Gelas, M, Undas, A. Recurrent ischemic cerebrovascular events in a patient with type I antithrombin deficiency caused by 9788 G>A splice site mutation: A case report. Blood Coag Fibrinol 2013; 24: 213–15.Google Scholar
Kovac, M, Mitic, G, Mikovic, Z, et al. Pregnancy related stroke in the setting of homozygous type-II HBS antithrombin deficiency. Thromb Res 2016; 139: 111–13.Google Scholar
Kuhle, S, Lane, DA, Jochmanns, K, et al. Homozygous antithrombin deficiency type II (99 Leu to Phe mutation) and childhood thromboembolism. Thromb Haemost 2001; 86: 1007–11.Google Scholar
Knuiman, MW, Folsom, AR, Chambles, LE, Liao, D, Wu, KK. Association of hemostatic variables with MRI-detected cerebral abnormalities: The atherosclerosis risk in communities study. Neuroepidemiology 2001; 20: 96104.Google Scholar
de Lau, LML, Leebeek, FWG, de Maat, MPM, Koudstaal, PJ, Dippel, DWJ. A review of hereditary and acquired coagulation disorders in the aetiology of ischaemic stroke. Int J Stroke 2010; 5: 385–94.Google Scholar
Stam, J. Thrombosis of the cerebral veins and sinuses. N Engl J Med 2005; 352: 1791–98.Google Scholar
Kujovich, JC. Factor V Leiden thrombophilia. Genet Med 2011; 13: 116.Google Scholar
Mumford, AD, McVey, JH, Morse, CV, et al. Factor V I359T: A novel mutation associated with thrombosis and resistance to activated protein C. Br J Haematol 2003; 123: 496501.Google Scholar
Chan, WP, Lee, CK, Kwong, YL, Lam, CK, Liang, R. A novel mutation of Arg306 of factor V gene in Hong Kong Chinese. Blood 1998; 91: 1135–9.Google Scholar
Lalouschek, W, Schillinger, M, Hsieh, K, et al. Matched case–control study on factor V Leiden and the prothrombin G20210A mutation in patients with ischemic stroke/transient ischemic attack up to the age of 60 years. Stroke 2005; 36: 14051409.Google Scholar
Tofler, GH, Massaro, J, O’Donnell, CJ, et al. Plasminogen activator inhibitor and risk for cardiovascular disease: The Framingham Heart Study. Thromb Res 2016; 140: 30–5.Google Scholar
Nave, AH, Lange, KS, Leonards, CO, et al. Lipoprotein (a) as a risk factor for ischemic stroke: A meta-analysis. Atherosclerosis 2015; 242: 496503.CrossRefGoogle ScholarPubMed
Jiang, B, Ryan, KA, Hamedani, A, et al. Prothrombin G20210A mutation is associated with young-onset stroke. Stroke 2014; 45: 961–7.Google Scholar
Lonn, E, Yusuf, S, Arnold, MJO, et al. Homocysteine lowering with folic acid and B vitamins in vascular disease. N Engl J Med 2006; 354: 1567–77.Google Scholar
Nah, H-W, Kim, JS. Premature intracranial arterial calcification in a patient with hyperhomocysteinemia. Neurology 2010; 75: 2252.Google Scholar
Saposnik, G, Ray, JG, Sheridan, P, et al. Homocysteine-lowering therapy and stroke risk, severity, and disability: Additional findings from the HOPE 2 trial. Stroke 2009; 40: 1365–72.Google Scholar
Hankey, GJ, Eikelboom, JW, Yi, Q, et al. Antiplatelet therapy and the effects of B vitamins in patients with previous stroke or transient ischaemic attack: A post-hoc subanalysis of VITATOPS, a randomised, placebo-controlled trial. Lancet Neurol 2012; 11: 512–20.Google Scholar
George, JN, Nester, CM. Syndromes of thrombotic microangiopathy. New Eng J Med 2014; 371: 654–66.Google Scholar
Shen, YM. Clinical evaluation of thrombotic microangiopathy: Identification of patients with suspected atypical hemolytic uremic syndrome. Thromb J 2016; 14: 19.Google Scholar
Kavanagh, D, McGlasson, S, Jury, A, et al. Type I interferon causes thrombotic microangiopathy by a dose-dependent toxic effect on the microvasculature. Blood 2016; 128: 2824–33.Google Scholar
Al-Nouri, ZL, Reese, JA, Terrell, DR, et al. Drug-induced thrombotic microangiopathy: A systematic review of published reports. Blood 2015; 125: 616–18.Google Scholar
Luzzatto, L. Recent advances in the pathogenesis and treatment of paroxysmal nocturnal hemoglobinuria. F1000Research 2016, doi: 10.12688/f1000research.7288.1.Google Scholar
de Latour, RP, Mary, JY, Salanoubat, C, et al. Paroxysmal nocturnal hemoglobinuria: Natural history of disease subcategories. Blood 2008; 112: 3099–106.Google Scholar
Meppiel, E, Crassard, J, de Latour, RP, et al. Cerebral venous thrombosis in paroxysmal nocturnal hemoglobinuria. Medicine 2015; 94: 18.Google Scholar
Helley, D, de Latour, RP, Porcher, R, et al. Evaluation of hemostasis and endothelial function in patients with paroxysmal nocturnal hemoglobinuria receiving eculizumab. Haematologica 2010; 95: 574–81.Google Scholar
DeZern, AE, Brodsky, RA. Paroxysmal nocturnal hemoglobinuria. Hematolol Oncol Clin North Am 2015; 29: 479–94.Google Scholar
Artoni, A, Bucciarelli, P, Martinelli I. Cerebral thrombosis and myeloproliferative neoplasms. Curr Neurol Neurosci Rep 2014; 14: 496.Google Scholar
Crespo, AM, Abraira, L, Guanyabens, N, et al. Recurrent stroke with rapid development of intracranial stenosis in polycythemia vera. J Stroke Cerebrovasc Dis 2016; 25: e413.Google Scholar
Tanashyan, MM, Kuznetsova, PI, Shabalina, AA, et al. Clinical characteristics of cerebrovascular pathology with patients suffering from Ph-negative myeloproliferative disease. Cerebrovascular Diseases Extra 2016; 6: 6670.Google Scholar
Khandelwal, S, Arepally, GM. Immune pathogenesis of heparin-induced thrombocytopenia. Thromb Haemost 2016; 116: 792–8.Google Scholar
Itaabashi, R, Yazawa, Y, Kawata, K, et al. Paradoxical brain emboli related to heparin-induced thrombocytopenia after catheter ablation. J Stroke Cerebrovasc Dis 2015; 24: e21921.Google Scholar
Cundiff, DK. Anticoagulants for cerebral venous thrombosis. Stroke 2014; 45: 298304.Google Scholar
Warkentin, TE. Clinical picture of heparin-induced thrombocytopenia (HIT) and its differentiation from non-HIT thrombocytopenia. Thromb Haemost 2016; 116: 813–22.Google Scholar
Centers for Disease Control and Prevention. Reproductive health: Pregnancy mortality surveillance system. http://www.cdc.gov/reproductivehealth/maternalinfanthealth/PMSS.html. Published 2014. Accessed July 18, 2015.Google Scholar
Simcox, LE, Ormesher, L, Tower, C, Greer, IA. Thrombophilia and pregnancy complications. Int J Mol Sci 2015; 16: 28418–28.Google Scholar
Kamel, H, Navi, BB, Sriram, N, et al. Risk of a thrombotic event after the 6-week postpartum period. N Engl J Med 2014; 370: 1307–15.Google Scholar
Lockwood, C, Wendel, G. Practice bulletin no. 124: Inherited thrombophilias in pregnancy. Obstet Gynecol 2011; 118: 730–40.Google Scholar
Centers for Disease Control and Prevention. WISQARS Database. Twenty Leading Causes of Death by Age Group, United States 2010. Available online at: http://www.cdc.gov/Injury/wisqars/.Google Scholar
Roach, ES, Golomb, MR, Adams, R, et al. Management of stroke in infants and children: A scientific statement from a special writing group of the American Heart Association Stroke Council and the Council on Cardiovascular Disease in the Young. Stroke 2008; 39: 2644–91.Google Scholar
Amlie-Ledfond, C, Sebire, G, Fullerton, HJ. Recent developments in childhood arterial ischemic stroke. Lancet Neurol 2008; 7: 425.Google Scholar
Mackay, MT, Wiznitzer, M, Benedict, SL, et al. Arterial ischemic stroke risk factors: The International Pediatric Stroke Study. Ann Neurol 2011; 69: 130–40.Google Scholar
Schmidt, B, Andrew, M. Neonatal thrombosis: Report of a prospective Canadian and international registry. Pediatrics 1995; 96: 939–43.Google Scholar
Andrew, M, David, M, Adams, M, et al. Venous thromboembolic complications (VTE) in children: First analyses of the Canadian Registry of VTE. Blood 1994; 83: 1251–7.Google Scholar
Shroff, M, deVeber, G. Sinovenous thrombosis in children. Neuroimaging Clin N Am 2003; 13: 115–38.Google Scholar
deVeber, G, Andrew, M, Adams, C, et al. Cerebral sinovenous thrombosis in children. N Engl J Med 2001; 345: 417–23.Google Scholar
Mallick, AA, Ganesan, V, Kirkham, FJ, et al. Childhood arterial ischaemic stroke incidence, presenting features, and risk factors: A prospective population-based study. Lancet Neurol 2014; 13: 3543.Google Scholar
Kenet, Gili, Aronis, S, Berkun, Y, et al. Impact of thrombophilia on risk of arterial ischemic stroke or cerebral sinovenous thrombosis in neonates and children a systematic review and meta-analysis of observational studies. Circulation 2010; 121: 1838–47.Google Scholar
Yang, JYK, Chan, AKC. Pediatric thrombophilia. Pediatr Clin N Am 2013; 60: 1443–62.Google Scholar
Monagle, P, Adams, M, Mahoney, M, et al. Outcome of pediatric thromboembolic disease: A report from the Canadian Childhood Thrombophilia Registry. Pediatr Res 2000; 47: 763–6.Google Scholar
Chalmers, EA. Epidemiology of venous thromboembolism in neonates and children. Thromb Res 2006; 118: 312.Google Scholar

References

Allford, S. L., Hunt, B. J., Rose, P., and Machin, S. J. 2003. Guidelines on the diagnosis and management of the thrombotic microangiopathic haemolytic anaemias. Br J Haematol, 120, 556–73.Google Scholar
Alungal, J., Abdulla, M. C., Koya, J. M., and Jenner, P. F. 2014. Multiple hemorrhagic infarcts in thrombotic thrombocytopenic purpura. Neurol India, 62, 582–3.Google Scholar
Amorosi, E. L. and Ultmann, J. E. 1966. Thrombotic thrombocytopenic purpura: Report of 16 cases and review of the literature. Medicine (Baltimore), 45, 139–59.Google Scholar
Asada, Y., Sumiyoshi, A., Hayashi, T., Suzumiya, J., and Kaketani, K. 1985. Immunohistochemistry of vascular lesion in thrombotic thrombocytopenic purpura, with special reference to factor VIII related antigen. Thromb Res, 8, 469–79.Google Scholar
Bakshi, R., Shaikh, Z. A., Bates, V. E., and Kinkel, P. R. 1999. Thrombotic thrombocytopenic purpura: Brain CT and MRI findings in 12 patients. Neurology, 52, 1285–8.Google Scholar
Balduini, C.L., Gugliotta, L., Luppi, M., et al. 2010. High versus standard dose methylprednisolone in the acute phase of idiopathic thrombotic thrombocytopenic purpura: A randomized study. Ann Hematol, 89, 591–6.Google Scholar
Bellante, F., Redondo Saez, P., Springael, C., and Dethy, S. 2014. Stroke in thrombotic thrombocytopenic purpura induced by thyrotoxicosis: A case report. J Stroke Cerebrovasc Dis, 23, 1744–6.Google Scholar
Ben-Yehuda, D., Rose, M., Michaeli, T., and Eldor, A. 1998. Permanent neurological complications in patients with thrombotic thrombocytopenic purpura. Am J Hematol, 29, 74–8.Google Scholar
Bettoni, G., Palla, R., Valsecchi, C., et al. 2012. ADAMTS-13 activity and autoantibodies classes and subclasses as prognostic predictors in acquired thrombotic thrombocytopenic purpura. J Thromb Haemost, 10, 1556–65.Google Scholar
Blombery, P. and Scully, M. 2014. Management of thrombotic thrombocytopenic purpura: Current perspectives. J Blood Med, 5, 1523.Google Scholar
Cataland, S. R. and Wu, H. M. 2005. Immunotherapy for thrombotic thrombocytopenic purpura. Curr Opin Hematol, 12, 359–63.Google Scholar
Cataland, S. R. and Wu, H. M. 2015. Acquired thrombotic thrombocytopenic purpura: New therapeutic options and their optimal use. J Thromb Haemost, 13 (Suppl 1), S2239.Google Scholar
Crawley, J. T. B. and Scully, M. A. 2013 Thrombotic thrombocytopenic purpura: Basic pathophysiology and therapeutic strategies. Hematol Am Soc Hematol Educ Program 2013, 292–9.Google Scholar
D’Aprile, P., Farchi, G., Pagliarulo, R., and Carella, A. 1994. Thrombotic thrombocytopenic purpura: MR demonstration of reversible brain abnormalities. Am J Neuroradiol, 15, 1920.Google Scholar
Ellchuk, T. N., Shah, L. M., Hewlett, R. H., and Osborn, A. G. 2011. Suspicious neuroimaging pattern of thrombotic microangiopathy. Am J Neuroradiol, 32, 734–8.Google Scholar
Fagniez, O., Tertian, G., Dreyfus, M., et al. 2011. Hematological disorders related cerebral infarctions are mostly multifocal. J Neurol Sci, 304, 8792.Google Scholar
Froissart, A., Buffet, M., Veyradier, A., et al. 2012. Efficacy and safety of firstline rituximab in severe, acquired thrombotic thrombocytopenic purpura with a suboptimal response to plasma exchange. Experience of the French Thrombotic Microangiopathies Reference Center. Crit Care Med, 40, 104–11.Google Scholar
Fujikawa, K., Suzuki, H., McMullen, B., and Chung, D. 2001. Purification of human von Willebrand factor-cleaving protease and its identification as a new member of the metalloproteinase family. Blood, 98, 1662–6.Google Scholar
Fujimura, Y., Matsumoto, M., Isonishi, A., et al. 2011. Natural history of Upshaw–Schulman syndrome based on ADAMTS13 gene analysis in Japan. J Thromb Haemost, 9 (Suppl 1), 283301.Google Scholar
Furlan, M. and Lämmle, B. 2001. Aetiology and pathogenesis of thrombotic thrombocytopenic purpura and haemolytic uraemic syndrome: The role of von Willebrand factor-cleaving protease. Best Pract Res Clin Haematol, 14, 437–54.Google Scholar
Furlan, M., Robles, R., Galbusera, M., et al. 1998. Von Willebrand factor-cleaving protease in thrombotic thrombocytopenic purpura and the hemolytic uremic syndrome. N Engl J Med, 339, 1578–84.Google Scholar
George, J. N. 2010. How I treat patients with thrombocytopenic thrombotic purpura. Blood, 116, 4060–9Google Scholar
Griffin, D., Al-Nouri, Z. L., Muthurajah, D., et al. 2013 First symptoms in patients with thrombotic thrombocytopenic purpura (TTP): What are they and when do they occur? Transfusion, 53, 235–7.Google Scholar
Gurkan, E., Baslamisli, F., Guvenc, B., et al. 2005. Thrombotic thrombocytopenic purpura in southern Turkey: A single-center experience of 29 cases. Clin Lab Haematol, 27, 121–5.Google Scholar
Han, B., Page, E. E., Stewart, L. M., et al. 2015. Depression and cognitive impairment following recovery from thrombotic thrombocytopenic purpura. Am J Hematol, 90, 709–14.Google Scholar
Hoirisch-Clapauch, S. and Nardi, A. E. 2014. A role for tissue plasminogen activator in thrombotic thrombocytopenic purpura. Med Hypotheses, 83, 747–50.Google Scholar
Horton, T. M., Stone, J. D., Yee, D., et al. 2003. Case series of thrombotic thrombocytopenic purpura in children and adolescents. J Pediatr Hematol Oncol, 25, 336–9.Google Scholar
Imanirad, I., Rajasekhar, A., and Zumberg, M. 2012. A case series of atypical presentations of thrombotic thrombocytopenic purpura. J Clin Apher, 27, 221–6.Google Scholar
Kay, A. C., Solberg, L. A., Nichols, D. A., and Pettit, R. M. 1991. Prognostic significance of computed tomography of the brain in thrombotic thrombocytopenic purpura. Mayo Clin Proc, 66, 602–7.Google Scholar
Kelly, P. J., McDonald, C. T., O’Neill, G., et al. 1998. Middle cerebral artery main stem thrombosis in two siblings with familial thrombotic thrombocytopenic purpura. Neurology, 50, 1157–60.Google Scholar
Kondo, K., Yamawaki, T., Nagatsuka, K., Miyashita, K., and Naritomi, H. 2003. Reversible stenosis of major cerebral arteries demonstrated by MRA in thrombotic thrombocytopenic purpura. J Neurol, 250, 995–7.Google Scholar
Kremer Hovinga, J. A., Vesely, S. K., Terrell, D. R, Lämmle, B., and George, J. N. 2010. Survival and relapse in patients with thrombotic thrombocytopenic purpura. Blood, 115, 1500–11.Google Scholar
Kremer Hovinga, J. A. and Lämmle, B. 2012. Role of ADAMTS13 in the pathogenesis, diagnosis, and treatment of thrombotic thrombocytopenic purpura. Hematol Am Soc Hematol Educ Program 2012, 610–6.Google Scholar
Kroll, M. H., Hellums, J. D., McIntire, L. V., Schafer, A. I., and Moake, J. L. 1996. Platelets and shear stress. Blood, 88, 1525–41.Google Scholar
Kwaan, H. C. 1987. Clinicopathological features of thrombotic thrombocytopenic purpura. Semin Hematol, 24, 7481.Google Scholar
Kwaan, H. C. and Boggio, L. N. 2005. The clinical spectrum of thrombotic thrombocytopenic purpura. Semin Thromb Hemost, 31, 673–80.Google Scholar
Lämmle, B., Kremer Hovinga, J. A., and Alberio, L. 2005. Thrombotic thrombocytopenic purpura. J Thromb Haemost, 3, 1663–75.Google Scholar
Levy, G. A., Nichols, W. C., Lian, E. C., et al. 2001. Mutations in a member of the ADAMTS gene family cause thrombotic thrombocytopenic purpura. Nature, 413, 488–94.Google Scholar
Lotta, L. A., Garagiola, I., Palla, R., Cairo, A., and Peyvandi, F. 2010. ADAMTS13 mutations and polymorphisms in congenital thrombotic thrombocytopenic purpura. Hum Mutat, 31, 11–9.Google Scholar
Lucchesi, A., Fattori, P. P., Ronconi, S., et al. 2015. Atypical presentations of thrombotic thrombocytopenic purpura in middle-aged women with recurrent cerebral macrovascular thrombosis: A case report. Ann Hematol, 94, 1597–8.Google Scholar
Martin, J. N. Jr., Bailey, A. P., Rehberg, J. F., et al. 2008. Thrombotic thrombocytopenic purpura in 166 pregnancies: 1955–2006. Am J Obstet Gynecol, 199, 98104.Google Scholar
Meloni, G., Proia, A., Antonini, G., et al. 2001. Thrombotic thrombocytopenic purpura: Prospective neurologic neuroimaging and neurophysiologic evaluation. Haematologica, 86, 1194–9.Google Scholar
Moake, J. L. 2002. Thrombotic microangiopathies. N Engl J Med, 347, 589600.Google Scholar
Moake, J. L. and McPherson, P. D. 1989. Abnormalities of von Willebrand factor multimers in thrombotic thrombocytopenic purpura and the hemolytic uremic syndrome. Am J Med, 87, 915.Google Scholar
Moake, J. L., Rudy, C. K., Troll, J. H., et al. 1982. Unusually large plasma factor VIII: Von Willebrand factor multimers in chronic relapsing thrombotic thrombocytopenic purpura. N Engl J Med, 307, 1432–5.Google Scholar
Moschkowitz, E. 1924. Hyaline thrombosis of the terminal arterioles and capillaries: A hitherto undescribed disease. Proc N Y Pathol Soc, 24, 21–4.Google Scholar
Müller, J., Czinyéri, J., Sasvári, I., Garami, M., and Kovács, G. 2001. Thrombotic thrombocytopenic purpura, Moschkowitz syndrome. Int Pediatr, 16, 144–9.Google Scholar
Paliwal, P. R., Teoh, H. L., and Sharma, V. K. 2014. Association between reversible cerebral vasoconstriction syndrome and thrombotic thrombocytopenic purpura. J Neurol Sci, 339, 4751.Google Scholar
Park, S. A., Lee, T. K., Sung, K. B., and Park, S. K. 2005. Extensive brain stem lesions in thrombotic thrombocytopenic purpura: Repeat magnetic resonance findings. J Neuroimag, 15, 7981.Google Scholar
Petitt, R. M. 1980. Thrombotic thrombocytopenic purpura: A thirty-year review. Semin Thromb Hemost, 6, 350–5.Google Scholar
Peyvandi, F., Ferrari, S., Lavoretano, S., Canciani, M. T., and Mannucci, P. M. 2004. Von Willebrand factor cleaving protease (ADAMTS-13) and ADAMTS-13 neutralizing autoantibodies in 100 patients with thrombotic thrombocytopenic purpura. Br J Hematol, 127, 433–9.Google Scholar
Peyvandi, F., Lavoretano, S., Palla, R., et al. 2008. ADAMTS13 and anti-ADAMTS13 antibodies as markers for recurrence of acquired thrombotic thrombocytopenic purpura during remission. Haematologica, 93, 2329.Google Scholar
Piastra, M., Currò, V., Chiaretti, A., et al. 2001. Intracranial hemorrhage at the onset of thrombotic thrombocytopenic purpura in an infant: Therapeutic approach and intensive care management. Pediatr Emerg Care, 17, 42–5.Google Scholar
Reese, J. A., Muthurajah, D. S., Kremer Hovinga, J. A., et al. 2013. Children and adults with thrombotic thrombocytopenic purpura associated with severe, acquired ADAMTS 13 deficiency: Comparison of incidence, demographic and clinical features. Pediatr Blood Cancer, 60, 1676–82.Google Scholar
Ridolfi, R. L. and Bell, W. R. 1981. Thrombotic thrombocytopenic purpura: A report of 25 cases and a review of the literature. Medicine (Baltimore), 60, 413–28.Google Scholar
Rieger, M., Mannucci, P. M., Kremer Hovinga, J. A., et al. 2005. ADAMTS 13 autoantibodies in patients with thrombotic microangiopathies and other immunomediated diseases. Blood, 106, 1262–7.Google Scholar
Rinkel, G. J. E. and Wijdicks, E. F. M. 1991. Stroke in relapsing thrombotic thrombocytopenic purpura. Stroke, 28, 1087–9.Google Scholar
Rock, G. A., Shumak, K. H., Buskard, N. A., et al. 1991. Comparison of plasma exchange with plasma infusion in the treatment of thrombotic thrombocytopenic purpura. Canadian Apheresis Study Group. N Engl J Med, 325, 393–7.Google Scholar
Rock, G., Anderson, D., Clark, W., et al. 2005. Does cryosupernatant plasma improve outcome in thrombotic thrombocytopenic purpura? No answer yet. Br J Haematol, 129, 7986.Google Scholar
Rojas, J.C., Banerjee, C., Siddiqui, F., Nourbakhsh, B., and Powell, C. M. 2013. Acute ischemic stroke caused by atypical thrombotic thrombocytopenic purpura. Neurology, 80, e235–e38.Google Scholar
Ruggeri, Z. M., Dent, J. A., and Saldívar, E. 1999. Contribution of distinct adhesive interactions to platelet aggregation in flowing blood. Blood, 94, 172–8.Google Scholar
Sadler, J. E. 1998. Biochemistry and genetics of von Willebrand factor. Annu Rev Biochem, 67, 395424.Google Scholar
Sadler, J. E., Moake, J. L., Miyata, T., and George, J. M. 2004. Recent advances in thrombotic thrombocytopenic purpura. Hematol Am Soc Hematol Educ Prog, 407–23.Google Scholar
Sarode, R., Gottschall, J. L., Aster, R. H., and McFarland, J. G. 1997. Thrombotic thrombocytopenic purpura: Early and late responders. Am J Hematol, 54, 1027.Google Scholar
Sarode, R. 2009. Atypical presentations of thrombotic thrombocytopenic purpura: A review. J Clin Apher, 24, 4752.Google Scholar
Sarode, R., Bandarenko, N., Brecher, M.E., et al. 2014. Thrombotic thrombocytopenic purpura: 2012 American Society for Apheresis (ASFA) consensus conference on classification, diagnosis, management, and future research. J Clin Apher, 29, 148–67.Google Scholar
Sayani, F. A. and Abrams, C. S. 2015. How I treat refractory thrombotic thrombocytopenic purpura. Blood, 125, 3860–67.Google Scholar
Scheid, R., Hegenbart, U., Ballaschke, O., and von Cramon, D. Y. 2004. Major stroke in thrombotic-thrombocytopenic purpura (Moschkowitz syndrome). Cerebrovasc Dis, 18, 83–5.Google Scholar
Scheiflinger, F., Knobl, P., Trattner, B., et al. 2003. Nonneutralizing IgM and IgG antibodies to von Willebrand factor-cleaving protease (ADAMTS-13) in a patient with thrombotic thrombocytopenic purpura. Blood, 102, 3241–43.Google Scholar
Scully, M., Yarranton, H., Liesner, R., et al. 2008. Regional UK TTP Registry: Correlation with laboratory ADAMTS 13 analysis and clinical features. Br J Haematol, 142, 819–26.Google Scholar
Scully, M., McDonald, V., Cavenagh, J., et al. 2011. A phase 2 study of the safety and efficacy of rituximab with plasma exchange in acute acquired thrombotic thrombocytopenic purpura. Blood, 118, 1746–53.Google Scholar
Scully, M., Hunt, B.J., Benjamin, S., et al. 2012. Guidelines on the diagnosis and management of thrombotic thrombocytopenic purpura and other thrombotic microangiopathies. Br J Haematol, 158, 323–35.Google Scholar
Scully, M., Thomas, M., Underwood, M., et al. 2014. Thrombotic thrombocytopenic purpura and pregnancy: Presentation, management, and subsequent pregnancy outcomes. Blood, 124, 211–9.Google Scholar
Singer, K., Bornstein, F. P., and Wiles, S. A. 1947. Thrombotic thrombocytopenic purpura. Hemorrhagic diathesis with generalized platelet thrombosis. Blood, 2, 542–54.Google Scholar
Shekman, B. and Einav, Y. 2014. Thrombotic thrombocytopenic purpura and other thrombotic microangiopathic hemolytic anemias: Diagnosis and classification. Autoimmun Rev, 13, 584–6.Google Scholar
Sonneveld, M. A. H., De Maat, M. P. M., and Leebeek, F. W. G. 2014. Von Willebrand factor and ADAMTS13 in arterial thrombosis: A systematic review and meta-analysis. Blood Rev, 28, 167–78.Google Scholar
Sperati, C. J. and Moliterno, A. R. 2015. Thrombotic microangiopathy. Focus on atypical hemolytic uremic syndrome. Hematol Oncol Clin N Am, 29, 541–59.Google Scholar
Studt, J. D., Hovinga, J. A., Radonic, R., et al. 2004. Familial acquired thrombotic thrombocytopenic purpura: ADAMTS 13 inhibitory autoantibodies in identical twins. Blood, 103, 4195–7.Google Scholar
Tardy, B., Page, Y., Convers, P., et al. 1993. Thrombotic thrombocytopenic purpura: MR findings. Am J Neuroradiol, 14, 489–90.Google Scholar
Terrel, D. R., Williams, L. A., Vesely, S. K., et al. 2005. The incidence of thrombotic thrombocytopenic purpura–hemolytic uremic syndrome: All patients, idiopathic patients, and patients with severe ADAMTS-13 deficiency. J Thromb Haemost, 3, 1432–6.Google Scholar
Terrell, D. R., Vesely, S. K., Kremer Hovinga, J. A., Lämmle, B., and George, J. N., 2010. Different disparities of gender and race among the thrombotic thrombocytopenic purpura and hemolytic–uremic syndromes. Am J Hematol, 85, 844–7.Google Scholar
Tsai, H. M. 1996. Physiologic cleavage of von Willebrand factor by a plasma protease is dependent on its conformation and requires calcium ion. Blood, 87, 4235–44.Google Scholar
Tsai, H. M. 2000. High titers of inhibitors of von Willebrand factor-cleaving metalloproteinase in a fatal case of acute thrombotic thrombocytopenic purpura. Am J Hematol, 65, 251–5.Google Scholar
Tsai, H. M. 2002. Von Willebrand factor, ADAMTS 13, and thrombotic thrombocytopenic purpura. J Mol Med, 80, 639–47.Google Scholar
Tsai, H. M. 2003. Advances in the pathogenesis, diagnosis, and treatment of thrombotic thrombocytopenic purpura. J Am Soc Nephrol, 14, 1072–81.Google Scholar
Tsai, H. M. and Lian, E. C. 1998. Antibodies to von Willebrand factor-cleaving protease in acute thrombotic thrombocytopenic purpura. N Engl J Med, 339, 1585–94.Google Scholar
Turner, N., Sartain, S., and Moake, J. 2015. Ultralarge von Willebrand factor-induced platelet clumping and activation of the alternative complement pathway in thrombotic thrombocytopenic purpura and the hemolytic–uremic syndromes. Hematol Oncol Clin North Am, 29, 509–24.Google Scholar
Uğur Bilgin, A., Karaselek, M. A., and Camlı, K. 2014. Successful management of thrombotic thrombocytopenic purpura associated with pregnancy. Transfus Apher Sci, 50, 433–7.Google Scholar
Vesely, S. K. 2015. Life after acquired thrombotic thrombocytopenic purpura: Morbidity, mortality, and risks during pregnancy. J Thromb Haemost, 13 (Suppl. 1): S21622.Google Scholar
Veyradier, A., Obert, B., Houllier, A., Meyer, D., and Dima, J. P. 2001. Specific von Willebrand factor-cleaving protease in thrombotic microangiopathies: A study of 111 cases. Blood, 98, 1765–72.Google Scholar
Wada, H., Matsumoto, T., and Yamashita, Y. 2014. Natural history of thrombotic thrombocytopenic purpura and hemolytic uremic syndrome. Semin Thromb Hemost, 40, 866–73.Google Scholar
Westwood, J. P., Webster, H., McGuckin, S., et al. 2013. Rituximab for thrombotic thrombocytopenic purpura: Benefit of early administration during acute episodes and use of prophylaxis to prevent relapse. J Thromb Haemost, 11, 481–90.Google Scholar
Wijdicks, E. F. M. 1994. Silent brain infarct in thrombotic thrombocytopenic purpura. Stroke, 25, 1297–8.Google Scholar
Willis, M. S. and Bandarenko, N. 2005. Relapse of thrombotic thrombocytopenic purpura: Is it a continuum of disease? Semin Thromb Hemost, 31, 700–8.Google Scholar
Yu, W. L., Leung, T., Soo, Y., Lee, J., and Wong, K. S. 2015. Thrombotic thrombocytopenic purpura with concomitant small- and large-vessel thrombosis, atypical posterior reversible encephalopathy syndrome and cerebral microbleeds. Oxf Med Case Reports, 179182.Google Scholar
Zheng, X. L. 2015. ADAMTS13 and von Willebrand factor in thrombotic thrombocytopenic purpura. Annu Rev Med, 66, 211–25.Google Scholar
Zheng, X. L., Chung, D., Takayama, T. K., et al. 2001. Structure of von Willebrand factor-cleaving protease (ADAMTS13), a metalloprotease involved in thrombotic thrombocytopenic purpura. J Biol Chem, 276, 41059–63.Google Scholar

References

Abend, N. S., Licht, D. J., and Spencer, C. H. (2007). Lupus anticoagulant and thrombosis following Henoch–Schönlein purpura. Pediatr Neurol, 36, 345–7.Google Scholar
Belman, A. L., Leicher, C. R., Moshe, S. L., et al. (1985). Neurologic manifestations of Henoch–Schönlein purpura: Report of three cases and review of the literature. Pediatrics, 75, 687–92.Google Scholar
Berube, M. D., Blais, N., and Lanthier, S. (2014). Neurologic manifestations of Henoch–Schönlein purpura. In Neurologic Aspects of Systemic Disease. Part II. Biller, J. and Ferro, J. M., eds. Amsterdam: Elsevier, pp. 1101–11.Google Scholar
Calvino, M. C., Llorca, J., Garcia-Porrua, C., et al. (2001). Henoch–Schönlein purpura in children from northwest Spain: A 20 year epidemiologic and clinical study. Medicine, 80, 279–90.Google Scholar
Chen, C. L., Chiou, Y. H., Wu, C. Y., Lai, P. H., and Chung, H. M. (2000). Cerebral vasculitis in Henoch–Schönlein purpura: A case report with sequential magnetic resonance imaging changes and treated with plasmapharesis alone. Pediatr Nephrol, 15, 276–8.Google Scholar
Chiaretti, A., Caresta, E., Piastra, M., Pulitano, S., and Di Rocco, C. (2002). Cerebral hemorrhage in Henoch–Schönlein syndrome. Childs Nerv Syst, 18, 365–7.Google Scholar
Chuah, J. and Meaney, T. (2005). Anterior ischaemic optic neuropathy secondary to Henoch–Schönlein purpura. Eye (Lond), 19, 1028.Google Scholar
Clark, J. H. and Fitzgerald, J. F. (1985). Hemorrhagic complications of Henoch–Schönlein syndrome. J Pediatr Gastroenterol Nutr, 4, 311–15.Google Scholar
Elinson, P., Foster, K. W., and Kaufman, D. B. (1990). Case report: Magnetic resonance imaging of central system vasculitis. Acta Paediatr Scand, 70, 710–3.Google Scholar
Eun, S. H., Kim, S. J., Cho, D. S., et al. (2003). Cerebral vasculitis in Henoch–Schönlein purpura: MRI and MRA findings, treated with plasmapheresis alone. Pediatr Int, 45, 484–7.Google Scholar
Garzoni, L., Vanoni, F., Rizzi, M., et al. (2009). Nervous system dysfunction in Henoch–Schönlein syndrome: Systematic review of the literature. Rheumatology, 48, 1524–9.Google Scholar
Giangiacomo, J. and Tsai, C. C. (1977). Dermal and glomerular deposition of IgA in anaphylactoid purpura. Am J Dis Child, 131, 981–3.Google Scholar
Ha, T. S. and Cha, S. H. (1996). Cerebral vasculitis in Henoch–Schönlein purpura: A case report with sequential magnetic resonance imaging. Pediatr Nephrol, 10, 634–6.Google Scholar
Heberden, W. (1801). Commentari Di Morborium Historia et Curatione. London: Payne. Reprinted as “Commentaries on the history and cure of diseases,” in The Classics of Medicine Library, Birmingham, AL: Gryphon Editions Ltd, 1982, 395–7.Google Scholar
Henoch, E. H. (1874). Über eine eigenthumliche Form von Purpura. Berlin Klin Wochenschr, 11, 641–3.Google Scholar
Henoch, E. H. (1899). Vorlesungen über Kinderkrankheiten. In Volhesunger über Kinderkrankheiten, Hirschward, A., ed. Berlin: Aufli, p. 839.Google Scholar
Knight, J. F., Harada, T., Thomas, M. A. B., et al. (1988). IgA rheumatoid factor and other autoantibodies in acute Henoch–Schönlein purpura. Contrib Nephrol, 67, 117–20.Google Scholar
Levy, M., Broyer, M., Arsan, A., Levy-Bentolila, D., and Habib, R. (1976). Anaphylactoid purpura nephritis in childhood: Natural history and immunopathology. Adv Nephrol, 6, 183228.Google Scholar
Ng, C. C., Hunag, S. C., Huang, L. T. (1996) Henoch–Schönlein purpura with intracerebral hemorrhage, case report. Pediatr Radiol, 26, 276–7.Google Scholar
Osler, W. (1914). The visceral lesions of purpura and allied conditions. Br Med J Clin Res, 1, 517–25.Google Scholar
Paolini, S., Ciappetta, P., Piattella, M. C., and Domenicucci, M. (2003). Henoch– Schönlein syndrome and cerebellar hemorrhage: Report of an adolescent case and literature review. Surg Neurol, 60, 339–42.Google Scholar
Saulsbury, F. T. (1999). Henoch–Schönlein purpura in children: Report of 100 patients and review of the literature. Medicine (Baltimore), 78, 395409.Google Scholar
Saulsbury, F. T. (2007). Clinical update: Henoch–Schönlein purpura. Lancet, 369, 976–8.Google Scholar
Scattarella, V., Pannarale, R., D’Angelo, V., et al. (1983). Occipital hemorrhage in a child with Henoch–Schönlein syndrome. J Neurosurg Sci, 27, 37–9.Google Scholar
Schönlein, J. L. (1832). In Allgemeine und Spezielle Pathologie und Therapie. Würzburg: C. Etlinger, vol. 2, pp. 6870.Google Scholar
Shin, J., Lee, J. S., Kim, H. D., and Lee, Y. M. (2006). Neurologic manifestations and treatment of Henoch–Schönlein purpura. Brain Dev, 28, 547.Google Scholar
Sokol, D. K., McIntyre, J. A., Short, R. A., et al. (2000). Henoch–Schönlein purpura and stroke: Antiphosphatidyl-ethanolamine antibody in CSF and serum. Neurology, 55, 1379–81.Google Scholar
Stevenson, J. A., Leong, L. A., Cohen, A. H., and Border, W. A. (1982). Simultaneous demonstration of IgA deposits in involved skin, intestine, and kidney. Arch Pathol Lab Med, 106, 192–5.Google Scholar
Trapani, S., Miceli, A., Grisolia, F., et al. (2005). Henoch–Schönlein purpura in childhood: Epidemiological and clinical analysis of 150 cases over a 5-year period and review of the literature. Semin Arthritis Rheum, 35, 143–53.Google Scholar
Wen, Y.-K., Yang, Y., and Chang, C.-C. (2005). Cerebral vasculitis and intracerebral hemorrhage in Henoch–Schönlein purpura treated with plasmapheresis. Pediatr Nephrol, 20, 223–5.Google Scholar
Woolfenden, A. R., Hukin, J., Poskitt, K. J., et al. (1998). Encephalopathy complicating Henoch–Schönlein purpura; reversible MRI changes. Pediatr Neurol, 19, 74–7.Google Scholar
Wu, T. T., Sheu, S. J., and Chou, L. C. (2002). Henoch–Schönlein purpura with bilateral central retinal artery occlusion. Br J Ophthalmol, 86, 351–2.Google Scholar
Yilmaz, C., Caksen, H., Arslan, S., et al. (2006). Bilateral brachial plexopathy complicating Henoch–Schönlein purpura. Brain Dev, 28, 326–8.Google Scholar

Save book to Kindle

To save this book to your Kindle, first ensure coreplatform@cambridge.org is added to your Approved Personal Document E-mail List under your Personal Document Settings on the Manage Your Content and Devices page of your Amazon account. Then enter the ‘name’ part of your Kindle email address below. Find out more about saving to your Kindle.

Note you can select to save to either the @free.kindle.com or @kindle.com variations. ‘@free.kindle.com’ emails are free but can only be saved to your device when it is connected to wi-fi. ‘@kindle.com’ emails can be delivered even when you are not connected to wi-fi, but note that service fees apply.

Find out more about the Kindle Personal Document Service.

Available formats
×

Save book to Dropbox

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Dropbox.

Available formats
×

Save book to Google Drive

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Google Drive.

Available formats
×