Skip to main content Accessibility help
×
Hostname: page-component-848d4c4894-pjpqr Total loading time: 0 Render date: 2024-06-15T07:58:11.916Z Has data issue: false hasContentIssue false

T

Published online by Cambridge University Press:  05 May 2023

J. F. Cade
Affiliation:
University of Melbourne
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
Critical Care Compendium
1001 Topics in Intensive Care & Acute Medicine
, pp. 456 - 478
Publisher: Cambridge University Press
Print publication year: 2023

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

Bibliography

Boland, TA, Lee, VH, Bleck, TP. Stress-induced cardiomyopathy. Crit Care Med 2015; 43: 686.Google Scholar
Connelly, KA, MacIsaac, AI, Jelinek, VM. Stress, myocardial infarction, and the ‘tako-tsubo’ phenomenon. Heart 2004; 90: e52.Google Scholar
Dote, K, Sato, H, Tateishi, H, et al. [Myocardial stunning due to simultaneous multivessel coronary spasms. [Japanese] J Cardiol 1991; 21: 203.Google Scholar
Park, JH, Kang, SJ, Song, JK, et al. Left ventricular apical ballooning due to severe physical stress in patients admitted to the medical ICU. Chest 2005; 128: 296.Google Scholar
Pernicova, I, Garg, S, Bourantas, CV, et al. Takotsubo cardiomyopathy: a review of the literature. Angiology 2010; 61: 166.CrossRefGoogle ScholarPubMed
Samardhi, H, Raffel, OC, Savage, M, et al. Takotsubo cardiomyopathy: an Australian single centre experience with medium term follow-up. Intern Med J 2012; 42: 35.Google Scholar
Samuels, MA, The brain-heart connection. Circulation 2007; 116: 77.Google Scholar
Sharkey, SW, Windenburg, DC, Lesser, JR, et al. Natural history and expansive clinical profile of stress (tako-tsubo) cardiomyopathy. J Am Coll Cardiol 2010; 55: 333.Google Scholar

Bibliography

Edsall, G. Problems in the immunology and control of tetanus. Med J Aust 1976; 2: 216.Google Scholar
Gaber, T A-Z K, Mannemela, S. Botulinum toxin for muscle spasm after tetanus. J R Soc Med 2005; 98: 63.Google Scholar
Tidyman, M, Prichard, JG, Deamer, RL, et al. Adjunctive use of dantrolene in severe tetanus. Anesth Analg 1985; 64: 538.Google Scholar

Bibliography

Nora, JJ. Causes of congenital heart disease: old and new modes, mechanisms, and models. Am Heart J 1993; 125: 1409.Google Scholar
Wilson, NJ, Neutze, JM. Adult congenital heart disease: principles and management guidelines. Aust NZ J Med 1993; 23: 498 & 697.Google Scholar

Bibliography

Akca, S, Haji-Michael, P, de Mendonca, A, et al. Time course of platelet counts in critically ill patients. Crit Care Med 2002; 30: 753.Google Scholar
Aster, RH, Bougie, DW. Drug-induced immune thrombocytopenia. N Engl J Med 2007; 357: 904.Google Scholar
Aster, RH, Curtis, BR, McFarland, JG, et al. Drug-induced immune thrombocytopenia: pathogenesis, diagnosis, and management. J Thromb Haemost 2009; 7: 911.Google Scholar
Balduini, CL, Savoia, A, Seri, M. Inherited thrombocytopenias frequently diagnosed in adults. J Thromb Haemost 2013; 11: 1006.Google Scholar
Beutler, E. Platelet transfusions: the 20,000/μL trigger. Blood 1993; 81: 1411.Google Scholar
Chong, BH. Heparin-induced thrombocytopenia. J Thromb Haemost 2003; 1: 1471.Google Scholar
Cines, DB, Blanchette, VS. Immune thrombocytopenic purpura. N Engl J Med 2002; 346: 995.Google Scholar
Ferrara, JLM. The febrile platelet transfusion reaction: a cytokine shower. Transfusion 1995; 35: 89.Google Scholar
George, JN. Thrombotic thrombocytopenic purpura. N Engl J Med 2006; 354: 1927.Google Scholar
George, JN, El-Harake, M, Raskob, GE. Chronic idiopathic thrombocytopenic purpura. N Engl J Med 1994; 331: 1207.Google Scholar
Handtke, S, Thiele, T. Large and small platelets – (when) do they differ? J Thromb Haemost 20020; 18: 1256.Google Scholar
Haznedaroglu, IC, Goker, H, Turgut, M, et al. Thrombopoietin as a drug: biologic expectations, clinical realities, and future directions. Clin Appl Thromb Hemost 2002; 8: 193.Google Scholar
Hui, P, Cook, DJ, Lim, W, et al. The frequency and clinical significance of thrombocytopenia complicating critical illness. Chest 2011; 139: 271.Google Scholar
Italiano, JE, Shivdasani, A. Megakaryocytes and beyond: the birth of platelets. J Thromb Haemost 2003; 1: 1174.Google Scholar
Marder, VJ, Aird, WC, Bennett, JS, et al., eds. Hemostasis and Thrombosis: Basic Principles and Clinical Practice. 6th edition. Philadelphia: Lippincott Williams & Wilkins. 2012.Google Scholar
Moake, JL. Thrombotic microangiopathies. N Engl J Med 2002; 347: 589.Google Scholar
Mueller-Eckhardt, C. Post-transfusion purpura. Br J Haematol 1986; 64: 419.Google Scholar
Papadopoulos, J, Kane-Gill, S, Cooper, B, eds. Identification and prevention of common adverse drug events in the intensive care unit. Crit Care Med 2010; 36: 6 (suppl.).Google Scholar
Payne, BA, Pierre, RV. Pseudothrombocytopenia: a laboratory artifact with potentially serious consequences. Mayo Clin Proc 1984; 59: 123.Google Scholar
Pene, F, Benoit, DD. Thrombocytopenia in the critically ill: considering pathophysiology rather than looking for a magic threshold. Intens Care Med 2013; 39: 1656.Google Scholar
Rice, TW, Wheeler, AP. Coagulopathy in critically ill patient. Part 1: platelet disorders. Chest 2009; 136: 1622.Google Scholar
Selleng, K, Warkentin, TE, Greinacher, A. Heparin-induced thrombocytopenia in intensive care patients. Crit Care Med 2007; 35: 1165.Google Scholar
Thiolliere, F, Serre-Serpin, AF, Reignier, J, et al. Epidemiology and outcome of thrombocytopenic patients in the intensive care unit: results of a prospective multicenter study. Intens Care Med 2013; 39: 1460.Google Scholar
Various. Drug-induced thrombocytopenia. Chest 2005; 127(2): suppl.Google Scholar
Warkentin, TE, Greinacher, A, eds. Heparin-Induced Thrombocytopenia. 5th edition. London: CRC Press. 2012.Google Scholar
Warkentin, TE, Greinacher, A, Koster, A, et al. Treatment and prevention of heparin-induced thrombocytopenia: ACCP evidence-based clinical practice guidelines Chest 2008; 133 (suppl.): 340S.Google Scholar
Warkentin, TE, Levine, MN, Hirsh, J, et al. Heparin-induced thrombocytopenia in patients treated with low-molecular-weight heparin or unfractionated heparin. N Engl J Med 1995; 332: 1330.Google Scholar
Warkentin, TE, Sheppard, J-AI, Heels-Ansdell, D, et al. Heparin-induced thrombocytopenia in medical surgical critical illness. Chest 2013; 144: 848.Google Scholar
Williamson, DR, Albert, M, Heels-Ansdell, D, et al. Thrombocytopenia in critically ill patients receiving thromboprophylaxis. Chest 2013; 144: 1207.Google Scholar
Yang, Z, Stulz, P, von Segesser, L, et al. Different interactions of platelets with arterial and venous coronary bypass vessels. Lancet 1991; 337: 939.Google Scholar

Bibliography

Anagrelide Study Group. Anagrelide, a therapy for thrombocythemic states. Am J Med 1992; 92: 69.Google Scholar
Bentley, MA, Taylor, KM, Wright, SJ. Essential thrombocythaemia. Med J Aust 1999; 171: 210.Google Scholar
Layzer, RB. Hot feet: erythromelalgia and related disorders. J Child Neurol 2001; 16: 199.Google Scholar
Kurzrock, R, Cohen, PR. Erythromelalgia: review of clinical characteristics and pathophysiology. Am J Med 1991; 91: 416.Google Scholar
Marder, VJ, Aird, WC, Bennett, JS, et al., eds. Hemostasis and Thrombosis: Basic Principles and Clinical Practice. 6th edition. Philadelphia: Lippincott Williams & Wilkins. 2012.Google Scholar
Michiels, JJ, ed. Platelet-dependent vascular complications and bleeding symptoms in essential thrombocythemia and polycythemia vera. Semin Thromb Hemost 1997; 23: 333.Google Scholar
Michiels, JJ, Abels, J, Steketee, J, et al. Erythromelalgia caused by platelet mediated arteriolar inflammation and thrombosis in thrombocythemia. Ann Intern Med 1985; 102: 466.Google Scholar
Sroren, EC, Tefferi, A. Long-term use of anagrelide in young patients with essential thrombocythemia. Blood 2001; 97: 863.Google Scholar
Tefferi, A, Elliott, M, Solberg, L, et al. New drugs in essential thrombocythemia and polycythemia vera. Blood Rev 1997; 11: 1.Google Scholar

Bibliography

Leung, LLK. Thrombotic disorders. In: Scientific American Medicine. Hematology. Hamilton: Dekker Medicine. 2020.Google Scholar
Marder, VJ, Aird, WC, Bennett, JS, et al., eds. Hemostasis and Thrombosis: Basic Principles and Clinical Practice. 6th edition. Philadelphia: Lippincott Williams & Wilkins. 2012.Google Scholar
Winter, M-P, Schernthaner, GH, Lang, IM. Chronic complications of venous thromboembolism. J Thromb Haemost 2017; 15: 1531.Google Scholar

Bibliography

Conway, EM. Thrombomodulin and its role in inflammation. Semin Immunopathol 2012; 34: 107.Google Scholar
Maruyama, I. Recombinant thrombomodulin and activated protein C in the treatment of disseminated intravascular coagulation. Thromb Haemost 1999; 82: 718.Google Scholar
Saito, H, Maruyama, I, Shimazaki, S, 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 2007; 5: 31.Google Scholar
Valeriani, E, Squizzato, A, Gallo, A, et al. Efficacy and safety of recombinant human soluble thrombomodulin in patients with sepsis-associated coagulopathy: a systematic review and meta-analysis. J Thromb Haemost 2020; 18: 1618.Google Scholar
Vincent, JL, Francois, B, Zabolotskikh, I, et al. Effect of recombinant human soluble thrombomodulin on mortality in patients with sepsis-associated coagulopathy: the SCARLET randomized clinical trial. JAMA 2019; 321: 1993.Google Scholar
Yamakawa, K, Fujimi, S, Mohri, T, et al. Treatment effects of recombinant human soluble thrombomodulin in patients with severe sepsis: a historical control study. Crit Care 2011; 15: R123.Google Scholar

Bibliography

Baglin, T. Unraveling the thrombophilia paradox: from hypercoagulability to the prothrombotic state. J Thromb Haemost 2009; 8: 228.Google Scholar
Bick, RL, Kaplan, H. Syndromes of thrombosis and hypercoagulability: congenital and acquired thrombophilias. Clin Appl Thromb Hemost 1998; 4: 25.Google Scholar
Brenner, B, Conard, J, eds. Women’s issues in thrombophilia. Semin Thromb Hemost 2003; 29: 1.Google Scholar
Casini, A, Neerman-Arbez, M, Ariens, RA, et al. Dysfibrinogenemia: from molecular anomalies to clinical manifestations and management. J Thromb Haemost 2015; 13: 909.Google Scholar
Cattaneo, M. Hyperhomocysteinemia, atherosclerosis and thrombosis. Thromb Haemost 1999; 81: 165.Google Scholar
de Moerloose, P, Bounameaux, HR, Mannucci, PM. Screening tests for thrombophilic patients: which tests, for which patient, by whom, when, and why? Semin Thromb Hemost 1998; 24: 321.Google Scholar
den Heijer, M, Rosendaal, FR, Blom, HJ, et al. Hyperhomocystinemia and venous thrombosis: a meta-analysis. Thromb Haemost 1998; 80: 874.Google Scholar
Franchini, M, Mannucci, PM. ABO blood group and thrombotic vascular disease. Thromb Haemost 2014; 112: 1103.Google Scholar
Franchini, M, Martinelli, I, Mannucci, PM. Uncertain thrombophilia markers. Thromb Haemost 2016; 115: 25.Google Scholar
Franco, RF, Reitsma, PH, Lourenco, D, et al. Factor XIII val34leu is a genetic factor involved in the aetiology of venous thrombosis. Thromb Haemost 1999; 81: 676.Google Scholar
Harbin, MM, Lutsey, PL. May-Thurner syndrome: history of understanding and need for defining population prevalence. J Thromb Haemost 2020; 18: 534.Google Scholar
Hirsh, J, Guyatt, G, Albers, GW, et al., eds. Antithrombotic and thrombolytic therapy: ACCP evidence-based clinical practice guidelines (8th edition). Chest 2008; 133: no. 6 (suppl.).Google Scholar
Iba, T, Levy, JH, Levi, M, et al. Coagulopathy of coronavirus disease 2019. Crit Care Med 2020; 48: 1358.Google Scholar
Kearon, C, Crowther, M, Hirsh, J, et al. Management of patients with hereditary hypercoagulable disorders. Annu Rev Med 2000; 51: 169.Google Scholar
Khan, S, Dickerman, JD. Hereditary thrombophilia. Thromb J 2006; 4: 15.Google Scholar
Lane, DA, Mannucci, PM, Bauer, KA, et al. Inherited thrombophilia. Thromb Haemost 1996; 76: 651.Google Scholar
Leung, LLK. Thrombotic disorders. In: Scientific American Medicine. Hematology. Hamilton: Dekker Medicine. 2020.Google Scholar
Mannucci, PM, Franchini, M. Classic thrombophilic gene variants. Thromb Haemost 2015; 114: 885.Google Scholar
Marder, VJ, Aird, WC, Bennett, JS, et al., eds. Hemostasis and Thrombosis: Basic Principles and Clinical Practice. 6th edition. Philadelphia: Lippincott Williams & Wilkins. 2012.Google Scholar
May, R, Thurner, J. The cause of predominantly sinistral occurrence of thrombosis in the pelvic veins. Angiology 1957; 8: 419.Google Scholar
Miletich, JP, Prescott, SM, White, R, et al. Inherited predisposition to thrombosis. Cell 1993; 72: 477.Google Scholar
Oldenburg, J, Schwaab, R. Molecular biology of blood coagulation. Semin Thromb Hemost 2001; 27: 313.Google Scholar
Prins, MH, Hirsh, J. A critical review of the evidence supporting a relationship between impaired fibrinolytic activity and venous thromboembolism. Arch Intern Med 1991; 151: 1721.Google Scholar
Sacher, RA, ed. Thrombophilia: a forum on diagnosis and management in obstetrics, gynecology and surgery. Semin Thromb Hemost 1998; 24: suppl. 1.Google Scholar
Winter, M, Gallimore, M, Jones, DW. Should factor XII assays be included in thrombophilia screening? Lancet 1995; 346: 52.Google Scholar

Bibliography

Fox, LC, Cohney, SJ, Kausman, JY, et al. Consensus opinion on diagnosis and management of thrombotic microangiopathy in Australia and New Zealand. Intern Med J 2018; 48: 624.Google Scholar

Bibliography

Azoulay, E, Bauer, PR, Mariotte, E, et al. Expert statement on the ICU management of patients with thrombotic thrombocytopenic purpura. Intens Care Med 2019; 45: 1518.Google Scholar
Bell, WR, Braine, HG, Ness, PM, et al. Improved survival of thrombotic thrombocytopenic purpura-hemolytic uremic syndrome: clinical experience in 108 patients. N Engl J Med 1991; 325: 398.Google Scholar
Blombery, P, Kivivali, L, Pepperell, D, et al. Diagnosis and management of thrombotic thrombocytopenic purpura (TTP) in Australia. Intern Med J 2016; 46: 71.Google Scholar
Cines, DB, Konkle, BA, Furlan, M. Thrombotic thrombocytopenic purpura: a paradigm shift? Thromb Haemost 2000; 84: 528.Google Scholar
Editorial. TTP – desperation, empiricism, progress. N Engl J Med 1991; 325: 426.Google Scholar
George, JN. Thrombotic thrombocytopenic purpura. N Engl J Med 2006; 354: 1927.Google Scholar
Hovinger, JAK, Heeb, SR, Skowronska, M, et al. Pathophysiology of thrombotic thrombocytopenic purpura and hemolytic uremic syndrome. J Thromb Haemost 2018; 16: 618.Google Scholar
ISTH guidelines for the diagnosis and treatment of thrombotic thrombocytopenic purpura. J Thromb Haemost 2020; 18: 2486 & 2496.Google Scholar
Mannucci, PM. Thrombotic thrombocytopenic purpura: a simpler diagnosis at last? Thromb Haemost 1999; 82: 1380.Google Scholar
Mannucci, PM. Understanding organ dysfunction in thrombotic thrombocytopenic purpura. Intens Care Med 2015; 41: 715.Google Scholar
Mariotte, E, Blet, A, Galicier, L, et al. Unresponsive thrombotic thrombocytopenic purpura in critically ill patients. Intens Care Med 2013; 39: 1272.Google Scholar
Moake, JL, Rudy, CK, Troll, JH, et al. Unusually large plasma factor VIII: von Willebrand factor multimers in chronic relapsing thrombotic thrombocytopenic purpura. N Engl J Med 1982; 307: 1432.Google Scholar
Sadler, JE. Von Willebrand factor, ADAMTS-13, and thrombotic thrombocytopenic purpura. Blood 2008; 112: 11.Google Scholar
Saha, M, McDaniel, JK, Zheng, XL. Thrombotic thrombocytopenic purpura: pathogenesis, diagnosis and potential novel therapeutics. J Thromb Haemost 2017; 15: 1889.Google Scholar
Schleinitz, N, Ebbo, M, Mazodier, K, et al. Rituximab as preventative therapy of a clinical relapse in TTP with ADAMTS 13 inhibitor. Am J Hematol 2007; 82: 417.Google Scholar
Scully, M, Cataland, S, Coppo, P, et al. Consensus on the standardization of terminology in thrombotic thrombocytopenic purpura and related thrombotic microangiopathies. J Thromb Haemost 2017; 15: 312.Google Scholar
Scully, M, Cataland, S, Peyvandi, F, et al. Caplacizumab for acquired thrombotic thrombocytopenic purpura. N Engl J Med 2019; 380: 335.Google Scholar
Tsai, HM, Lian, ECY. Antibodies to von Willebrand factor-cleaving protease in acute thrombotic thrombocytopenic purpura. N Engl J Med 1998; 339: 1585.Google Scholar
VeyradierA, Meyer D. Thrombotic thrombocytopenic purpura and its diagnosis. J Thromb Haemost 2005; 3: 2420.Google Scholar
Zheng, XL. The standard of care for immune thrombotic thrombocytopenic purpura today. J Thromb Haemost 2021; 191: 1864.Google Scholar
Zheng, X, Chung, D, Takayama, TK, et al. Structure of von Willebrand factor-cleaving protease (ADAMTS13), a metalloprotease involved in thrombotic thrombocytopenic purpura. J Biol Chem 2001; 276: 41059.Google Scholar

Bibliography

Ladenson, PW. Hypothyroidism and thyrotoxicosis. In: Scientific American Medicine. Endocrinology & Metabolism. Hamilton: Dekker Medicine. 2020.Google Scholar
Mortimer, R. Thyroid function tests. Aust Prescriber 2011; 34: 12.Google Scholar

Bibliography

Graves, SR, Stenos, J. Tick-borne infectious diseases in Australia. Med J Aust 2017; 206: 320.Google Scholar

Bibliography

Burnham, JP, Kollef, MH. Understanding toxic shock syndrome. Intens Care Med 2015; 41: 1707.Google Scholar
Cone, LA, Woodard, DR, Schlievert, PM, et al. Clinical and bacteriologic observations of a toxic shock-like syndrome due to Streptococcus pyogenes. N Engl J Med 1987; 317: 146.Google Scholar
Davis, JP, Chesney, PJ, Wand, PJ, et al. Toxic-shock syndrome: epidemiologic features, recurrence, risk factors, and prevention. N Engl J Med 1980; 303: 1429.Google Scholar
Fronhoffs, S, Luyken, J, Steuer, K, et al. The effect of C1-esterase inhibitor in definite and suspected streptococcal toxic shock syndrome. Intens Care Med 2000; 26: 1566.Google Scholar
Kain, KC, Schulzer, M, Chow, AW. Clinical spectrum of nonmenstrual toxic shock syndrome (TSS): comparison with menstrual TSS by multivariate discriminant analysis. Clin Infect Dis 1993; 16: 100.Google Scholar
Langmuir, AD, Worthen, TD, Solomon, J, et al. The Thucydides syndrome: a new hypothesis for the cause of the plague of Athens. N Engl J Med 1985; 313: 1027.Google Scholar
Schlievert, PM, MacDonald, KL. Toxic Shock Syndrome. 2nd ed. Philadelphia: WB Saunders.1998.Google Scholar
Seal, DV. Necrotizing fasciitis. Curr Opin Infect Dis 2001; 14: 127.Google Scholar
Stevens, DL. Streptococcal toxic-shock syndrome: spectrum of disease, pathogenesis, and new concepts in treatment. Emerg Infect Dis 1995; 1: 3.Google Scholar
Stevens, DL, Tanner, MH, Winship, J, et al. Severe group A streptococcal infections associated with a toxic shock-like syndrome and scarlet fever toxin. N Engl J Med 1989; 321: 1.Google Scholar

Bibliography

Joiner, KA, Dubremetz, JF. Toxoplasma gondii: a protozoan for the nineties. Infect Immun 1993; 61: 1169.Google Scholar
McCabe, R, Remington, JS. Toxoplasmosis. N Engl J Med 1988; 318: 313.Google Scholar
Wong, S, Remington, JS. Toxoplasmosis in pregnancy. Clin Infect Dis 1994; 18: 853.Google Scholar

Bibliography

Barceloux, DG. Cobalt. J Toxicol Clin Toxicol 1999; 37: 201.Google Scholar
Barceloux, DG. Molybdenum. J Toxicol Clin Toxicol 1999; 37: 231.Google Scholar
Barceloux, DG. Nickel. J Toxicol Clin Toxicol 1999; 37: 239.Google Scholar
Barceloux, DG. Vanadium. J Toxicol Clin Toxicol 1999; 37: 265Google Scholar
Berger, MM, Cavadini, C, Chiolero, R, et al. Influence of large intakes of trace elements on recovery after major burns. Nutrition 1994; 10: 327.Google Scholar
Casaer, MP, Bellomo, R. Micronutrient deficiency in critical illness: an invisible foe? Intens Care Med 2019; 45: 1136.Google Scholar
Chandra, RK. Effect of vitamin and trace-element supplementation on immune responses and infection in elderly patients. Lancet 1992; 340: 1124.Google Scholar
Elia, M. Changing concepts of nutrient requirements in disease: implications for artificial nutritional support. Lancet 1995; 345: 1279.Google Scholar
Fleming, CR. Trace element metabolism in adult patients requiring total parenteral nutrition. Am J Clin Nutr 1989; 49: 573.Google Scholar
Heyland, DK, Dhaliwal, R, Suchner, U, et al. Antioxidant nutrients: a systematic review of trace elements and vitamins in the critically ill patient. Intens Care Med 2005; 31: 327.Google Scholar
Mertz, W. The essential trace elements. Science 1981; 213: 1332.Google Scholar
Prasad, AS, ed. Essential and Toxic Trace Elements in Human Health and Disease. New York: Liss. 1988.Google Scholar
Shenkin, A. Vitamin and essential trace element recommendations during intravenous therapy: theory and practice. Proc Nutr Soc 1986; 45: 383.Google Scholar
Simmer, K, Thompson, RPH. Trace elements. In: Cohen, RD, Lewis, B, Alberti, KGMM, Denman, AM, eds. The Metabolic and Molecular Basis of Acquired Disease. London: Baillere Tindall. 1990, p 670.Google Scholar
Singer, P, Manzanares, W, Berger, MM. What’s new in trace elements? Intens Care Med 2018; 44: 643.Google Scholar
Supplement. The trace elements: their role and function in nutritional support. Nutrition 1995; 2: no.1.Google Scholar

Bibliography

Barton, RN. Trauma and its metabolic products. Br Med Bull 1985; 41: 3.Google Scholar
Blaisdell, FW, Holcroft, JW, eds. Scientific American Surgery Handbook of Trauma. New York: Scientific American. 1999.Google Scholar
Green, DR. Trauma and the immune response. Immunol Today 1988; 9: 253.Google Scholar
Frayn, KN. Hormonal control of metabolism in trauma and sepsis. Clin Endocrinol 1986: 24: 577.Google Scholar
Moore, EE, Cogbill, TH, Malagoni, MA, et al. Scaling systems for organ specific injuries. Curr Opin Crit Care 1996; 2: 450.Google Scholar
Nelson, LD, ed. New advances in the care of critically injured patients. New Horizons: The Science and Practice of Acute Medicine 1999; 7: 1.Google Scholar
Smith, RM, Giannoudis, PV. Trauma and the immune response. J R Soc Med 1998; 91: 417.Google Scholar
Wisner, DH. Current priorities in the management of multiple injury. Curr Opin Crit Care 1996; 2: 463.Google Scholar

Bibliography

Spahn, DR, Bouillon, B, Duranteau, J, et al. The European guideline on management of major bleeding and coagulopathy following trauma: fifth edition. Crit Care 2019; 23: 98.Google Scholar

Bibliography

Fildes, J, Reed, L, Jones, N, et al. Trauma: the leading cause of maternal death. J Trauma 1992; 32: 643.Google Scholar
Knudson, MM. Acute abdominal injuries during pregnancy. Curr Opin Crit Care 1996; 2: 469.Google Scholar
Magriples, U, Chan, DW, Bruzek, D, et al. Thrombomodulin: a new marker for placental abruption. Thromb Haemost 1999; 81: 32.Google Scholar
Pearlman, MD, Tintinalli, JE, Lorenz, RP. Blunt trauma during pregnancy. N Engl J Med 1990; 323: 1609.Google Scholar
Sorensen, VJ, Bivins, BA, Obeid, FN, et al. Management of general surgical emergencies in pregnancy. Am Surg 1990; 56: 245.Google Scholar
Trauma Service. Trauma Guidelines Booklet. Melbourne: Royal Melbourne Hospital. 2011.Google Scholar
Weinberg, L, Steele, RG, Pugh, R, et al. The pregnant trauma patient. Anaesth Intens Care 2005; 33: 167.Google Scholar

Bibliography

Blumberg, HM, Leonard, MK, Jasmer, RM. Update on the treatment of tuberculosis and latent tuberculosis infection. JAMA 2005; 293: 2776.Google Scholar
Catanzaro, A. How to increase the accuracy of the diagnosis of tuberculosis. Pulmonary Perspectives 2004; 21(2): 1.Google Scholar
Darby, J, Black, J, Buising, K. Interferon-gamma release assays and the diagnosis of tuberculosis: have they found their place? Intern Med J 2014; 44: 624.Google Scholar
Davies, PDO. The challenge of tuberculosis. J R Soc Med 2003; 96: 262.Google Scholar
Davies, PDO, De Cock, KM, Leese, J, et al. Tuberculosis 2000. J R Soc Med 1996; 89: 431.Google Scholar
Donoghue, HD, Spigelman, M, Greenblatt, CL, et al. Tuberculosis: from prehistory to Robert Koch, as revealed by ancient DNA. Lancet Infect Dis 2004; 4: 584.Google Scholar
Fordham von Reyn, C. Correcting the record on BCG before we license new vaccines against tuberculosis. J R Soc Med 2017; 110: 428.Google Scholar
Frieden, TR, Sterling, TR, Munsiff, SS, et al. Tuberculosis: a review. Lancet 2003; 362: 887.Google Scholar
Keal, JL, Davies, PDO. Tuberculosis: a forgotten plague? J R Soc Med 2011; 104: 182.Google Scholar
Lancet Conference. The challenge of tuberculosis: statements on global control and prevention. Lancet 1995; 346: 809.Google Scholar
Lawn, SD, Zumla, AI. Tuberculosis. Lancet 2011; 378: 57.Google Scholar
Madkour, MM, ed. Textbook of Tuberculosis. Berlin: Springer-Verlag. 2003.Google Scholar
Milburn, H. Key issues in the diagnosis and management of tuberculosis. J R Soc Med 2007; 100: 134.Google Scholar
Millard, FJC. The rising incidence of tuberculosis. J R Soc Med 1996; 89: 497.Google Scholar
Ormerod, P, Campbell, J, Novelli, V. Chemotherapy and management of tuberculosis in the United Kingdom: recommendations 1998. Thorax 1998; 53: 536.Google Scholar
Parr, JB, Leonard, MK, Blumberg, HM. Tuberculosis. In: Scientific American Medicine. Infectious Diseases. Hamilton: Dekker Medicine. 2020.Google Scholar
Reichman, LB, Tanne, JH. Timebomb: The Global Epidemic of Multi-Drug-Resistant Tuberculosis. New York: McGraw-Hill. 2002.Google Scholar
Snider, DE, La Montagne, JR. The neglected global tuberculosis problem. J Infect Dis 1994; 169: 1189.Google Scholar
Snider, DE, Roper, WL. The new tuberculosis. N Engl J Med 1992; 326: 703.Google Scholar

Bibliography

Crino, PB, Nathanson, KL, Henske, EP. The tuberous sclerosis complex. N Engl J Med 2006; 355: 1345.Google Scholar
Critchley, M, Earle, C. Tubero-sclerosis and allied conditions. Brain 1932; 55: 311.Google Scholar
Curatolo, P, Bombardieri, R, Jozwiak, S. Tuberous sclerosis. Lancet 2008; 372: 657.Google Scholar
Lenoir, S, Grenier, P, Brauner, MW, et al. Pulmonary lymphangiomyomatosis and tuberous sclerosis: comparison of radiographic and thin-section CT findings. Radiology 1990; 175: 329.Google Scholar
Liu, H-J, Krymskaya, VP, Henske, EP. Immunotherapy for lymphangioleiomyomatosis and tuberous sclerosis: progress and future directions. Chest 2019; 156: 1062.Google Scholar
McCormack, FX. Lymphangioleiomyomatosis: a clinical update. Chest 2008; 133: 507.Google Scholar

Bibliography

Abraham, PA, Keane, WF. Glomerular and interstitial disease induced by nonsteroidal anti-inflammatory drugs. Am J Nephrol 1984; 4: 1.Google Scholar
Appel, GB, Bhat, P, Canetta, P. Tubulointerstitial diseases. In: Scientific American Medicine. Nephrology. Hamilton: Dekker Medicine. 2020.Google Scholar
Corwin, HL, Korbet, SM, Schwartz, MM. Clinical correlates of eosinophiluria. Arch Intern Med 1985; 145: 1097.Google Scholar
Fored, CM, Ejerblad, E, Lindblad, P, et al. Acetaminophen, aspirin, and chronic renal failure. N Engl J Med 2001; 345: 1801.Google Scholar
Hoitsma, AJ, Wetzels, JFM, Koene, RAP. Drug-induced nephrotoxicity: aetiology, clinical features and management. Drug Safety 1991; 6: 131.Google Scholar
Kincaid-Smith, P. Analgesic abuse and the kidney. Kidney Int 1980; 17: 250.Google Scholar
Linton, AL, Clark, WF, Driedger, AA, et al. Acute interstitial nephritis due to drugs. Ann Intern Med 1980; 93: 735.Google Scholar
Neilson, EG. Pathogenesis and therapy of interstitial nephritis. Kidney Int 1989; 35: 1257.Google Scholar
Ronco, PM, Flahault, A. Drug-induced end-stage renal disease. N Engl J Med 1994; 331: 1711.Google Scholar
Rossert, J. Drug-induced acute interstitial nephritis. Kidney Int 2001; 60: 804.Google Scholar
Turner, NN, Lameire, N, Goldsmith, DJ, et al. eds. Oxford Textbook of Clinical Nephrology. 4th edition. Oxford: Oxford University Press. 2015.Google Scholar

Bibliography

Arrambide, K, Toto, RD. Tumor lysis syndrome. Semin Nephrol 1993; 13: 273.Google Scholar
Barton, JC. Tumor lysis syndrome in nonhematopoietic neoplasms. Cancer 1989; 64: 738.Google Scholar
Coiffier, B, Mounier, N, Bologna, S, et al. Efficacy and safety of rasburicase (recombinant urate oxidase) for the prevention and treatment of hyperuricemia during induction chemotherapy of aggressive non-Hodgkin’s lymphoma. J Clin Oncol 2003; 21: 4402.Google Scholar
Howard, SC, Jones, DP, Pui, C-H. The tumor lysis syndrome N Engl J Med 2011; 364: 1844.Google Scholar
McCurdy, MT, Shanholtz, CB. Oncologic emergencies. Crit Care Med 2012; 40: 2212.Google Scholar
Zafrani, L, Canet, E, Darmon, M. Understanding tumor lysis syndrome. Intens Care Med 2019; 45: 1608.Google Scholar

Bibliography

Pezaro, C, Woo, HH, Davis, ID. Prostate cancer: measuring PSA. Intern Med J 2014; 44: 433.Google Scholar
Smith, RA, Cokkinides, V, Brooks, D, et al. Cancer screening in the United States, 2010: a review of current American Cancer Society guidelines and issues in cancer screening. CA: A Cancer Journal for Clinicians 2010; 60: 99.Google Scholar
Sturgeon, CM, Lai, LC, Duffy, MJ. Serum tumour markers: how to order and interpret them, BMJ 2010; 339: 852.Google Scholar

Bibliography

Chu, W-M. Tumor necrosis factor. Cancer Lett 2013; 328: 222.Google Scholar
Jani, M, Dixon, WG, Chinoy, H. Drug safety and immunogenicity of tumour necrosis factor inhibitors. Rheumatology 2018; 57: 1896.Google Scholar
Kalliolias, GD, Ivashkiv, LB. TNF biology, pathogenic mechanisms and emerging therapeutic strategies. Nature Rev Rheum 2015; 12: 49.Google Scholar
Lundy, SK, Gizinski, A, Fox, DA. Introduction to clinical immunology: overview of immune response, autoimmune conditions, and immunosuppressive therapeutics for rheumatic diseases. In: Scientific American Medicine. Allergy & Immunology. Hamilton: Dekker Medicine. 2020.Google Scholar
Monaco, C, Nanchahal, J, Taylor, P, et al. Anti-TNF therapy: past, present and future. Int Immunol 2015; 27: 55.Google Scholar
Old, LJ. Tumor necrosis factor. Sci Am 1988; 258: 59.Google Scholar
Udalova, I, Monaco, C, Nanchahal, J, et al. Anti-TNF therapy. Microbiol Spectr 2016; 4: 4.Google Scholar

Bibliography

Hornick, RB, Greisman, SE, Woodward, TE, et al. Typhoid fever. N Engl J Med 1970; 283: 686 & 739.Google Scholar
Parry, CM. Typhoid fever. Curr Infect Dis Rep 2004; 6: 27.Google Scholar
Rabsch, W, Tschape, H, Baumler, AJ. Non-typhoidal salmonellosis: emerging problems. Microbes Infect 2001; 3: 237.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.

  • T
  • J. F. Cade, University of Melbourne
  • Book: Critical Care Compendium
  • Online publication: 05 May 2023
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.

  • T
  • J. F. Cade, University of Melbourne
  • Book: Critical Care Compendium
  • Online publication: 05 May 2023
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.

  • T
  • J. F. Cade, University of Melbourne
  • Book: Critical Care Compendium
  • Online publication: 05 May 2023
Available formats
×