Skip to main content

Advertisement

Log in

Neurologic Complications of Commonly Used Drugs in the Hospital Setting

  • Neurology of Systemic Diseases (J Biller, Section Editor)
  • Published:
Current Neurology and Neuroscience Reports Aims and scope Submit manuscript

Abstract

This chapter reviews the neurologic complications of medications administered in the hospital setting, by class, introducing both common and less common side effects. Detail is devoted to the interaction between pain, analgesia, sedation, and their residual consequences. Antimicrobials are given in nearly every hospital setting, and we review their capacity to produce neurologic sequelae with special devotion to cefepime and the antiviral treatment of human immunodeficiency virus. The management of hemorrhagic stroke has become more complex with the introduction of novel oral anticoagulants, and we provide an update on what is known about reversal of the new oral anticoagulants. Both central and peripheral nervous system complications of immunosuppressants and chemotherapies are reviewed. Because diagnosis is generally based on clinical acumen, alone, neurotoxic syndromes resulting from psychotropic medications may be easily overlooked until severe dysautonomia develops. We include a practical approach to the diagnosis of serotonin syndrome and neuroleptic malignant syndrome.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1

Similar content being viewed by others

References

Papers of particular interest, published recently, have been highlighted as: • Of importance

  1. Micromedex. Available from: http://www.micromedexsolutions.com/micromedex2/librarian.

  2. Wijdicks EFM, Rabinstein AA. Neurocritical care (what do i do now?). USA: Oxford University Press; 2011.

    Google Scholar 

  3. Cytochrome P450 3A4 (CYP3A4) inhibitors and inducers 2015. Available from: http://www.uptodate.com/contents/image?imageKey=CARD/76992&topicKey=GAST%2F3568&source=outline_link&search=cytochrome+p450&selectedTitle=2~150&utdPopup=true.

  4. Barr J, Fraser GL, Puntillo K, Ely EW, Gelinas C, Dasta JF, et al. Clinical practice guidelines for the management of pain, agitation, and delirium in adult patients in the intensive care unit. Crit Care Med. 2013;41(1):263–306.

    Article  PubMed  Google Scholar 

  5. Bookwalter T, Gitlin M. Gabapentin-induced neurologic toxicities. Pharmacotherapy. 2005;25(12):1817–9.

    Article  PubMed  Google Scholar 

  6. Sullivan Jr JB, Rumack BH, Peterson RG. Acute carbamazepine toxicity resulting from overdose. Neurology. 1981;31(5):621–4.

    Article  PubMed  Google Scholar 

  7. Yoo L, Matalon D, Hoffman RS, Goldfarb DS. Treatment of pregabalin toxicity by hemodialysis in a patient with kidney failure. Am J Kidney Dis. 2009;54(6):1127–30.

    Article  PubMed  Google Scholar 

  8. Kaufman KR, Parikh A, Chan L, Bridgeman M, Shah M. Myoclonus in renal failure: two cases of gabapentin toxicity. Epilepsy Behav Case Rep. 2014;2:8–10.

    Article  PubMed  PubMed Central  Google Scholar 

  9. Clark SRA, Hocker S. Disabling asterixis due to gabapentin. J Med Cases. 2015;6(7):285–6.

    Article  Google Scholar 

  10. Hansenflaschen JH, Brazinsky S, Basile C, Lanken PN. Use of sedating drugs and neuromuscular blocking-agents in patients requiring mechanical ventilation for respiratory-failure—a national survey. JAMA. 1991;266(20):2870–5.

    Article  CAS  Google Scholar 

  11. Soliman HM, Melot C, Vincent JL. Sedative and analgesic practice in the intensive care unit: the results of a European survey. Br J Anaesth. 2001;87(2):186–92.

    Article  CAS  PubMed  Google Scholar 

  12. Mehta S, McCullagh I, Burry L. Current sedation practices: lessons learned from international surveys. Anesthesiol Clin. 2011;29(4):607–24.

    Article  PubMed  Google Scholar 

  13. Borgeat A, Wilder-Smith OH, Tassonyi E, Suter PM. Propofol and epilepsy: time to clarify! Anesth Analg. 1994;78(1):198–9.

    Article  CAS  PubMed  Google Scholar 

  14. Rosenberg H, Pollock N, Schiemann A, Bulger T, Stowell K. Malignant hyperthermia: a review. Orphanet J Rare Dis. 2015;10:93.

    Article  PubMed  PubMed Central  Google Scholar 

  15. Larach MG, Gronert GA, Allen GC, Brandom BW, Lehman EB. Clinical presentation, treatment, and complications of malignant hyperthermia in North America from 1987 to 2006. Anesth Analg. 2010;110(2):498–507.

    Article  PubMed  Google Scholar 

  16. Mattappalil A, Mergenhagen KA. Neurotoxicity with antimicrobials in the elderly: a review. Clin Ther. 2014;36(11):1489–511. e4.

    Article  CAS  PubMed  Google Scholar 

  17. Morgan JC, Sethi KD. Drug-induced tremors. Lancet Neurol. 2005;4(12):866–76.

    Article  CAS  PubMed  Google Scholar 

  18. Grill MF, Maganti RK. Neurotoxic effects associated with antibiotic use: management considerations. Br J Clin Pharmacol. 2011;72(3):381–93.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Tsiodras S, Zafiropoulou R, Kanta E, Demponeras C, Karandreas N, Manesis EK. Painful peripheral neuropathy associated with voriconazole use. Arch Neurol. 2005;62(1):144–6.

    Article  PubMed  Google Scholar 

  20. Agrawal AK, Sherman LK. Voriconazole-induced musical hallucinations. Infection. 2004;32(5):293–5.

    Article  CAS  PubMed  Google Scholar 

  21. Imhof A, Schaer DJ, Schanz U, Schwarz U. Neurological adverse events to voriconazole: evidence for therapeutic drug monitoring. Swiss Med Wkly. 2006;136(45–46):739–42.

    CAS  PubMed  Google Scholar 

  22. Schliamser SE, Cars O, Norrby SR. Neurotoxicity of beta-lactam antibiotics: predisposing factors and pathogenesis. J Antimicrob Chemother. 1991;27(4):405–25.

    Article  CAS  PubMed  Google Scholar 

  23. Koppel BS, Hauser WA, Politis C, van Duin D, Daras M. Seizures in the critically ill: the role of imipenem. Epilepsia. 2001;42(12):1590–3.

    Article  CAS  PubMed  Google Scholar 

  24. Sugimoto M, Uchida I, Mashimo T, Yamazaki S, Hatano K, Ikeda F, et al. Evidence for the involvement of GABA(A) receptor blockade in convulsions induced by cephalosporins. Neuropharmacology. 2003;45(3):304–14.

    Article  CAS  PubMed  Google Scholar 

  25. Miller AD, Ball AM, Bookstaver PB, Dornblaser EK, Bennett CL. Epileptogenic potential of carbapenem agents: mechanism of action, seizure rates, and clinical considerations. Pharmacotherapy. 2011;31(4):408–23.

    Article  CAS  PubMed  Google Scholar 

  26. Moore TD, Bechtel TP, Ayers LW. Effect of multidose therapy on cerebrospinal fluid penetration of cefazolin. Am J Hosp Pharm. 1981;38(10):1496–9.

    CAS  PubMed  Google Scholar 

  27. Vincken W. Psychotic reaction to cefuroxime. Lancet. 1984;1(8383):965.

    Article  CAS  PubMed  Google Scholar 

  28. Martinez-Rodriguez JE, Barriga FJ, Santamaria J, Iranzo A, Pareja JA, Revilla M, et al. Nonconvulsive status epilepticus associated with cephalosporins in patients with renal failure. Am J Med. 2001;111(2):115–9.

    Article  CAS  PubMed  Google Scholar 

  29. Bragatti JA, Rossato R, Ziomkowski S, Kliemann FA. Cefepime-induced encephalopathy: clinical and electroencephalographic features in seven patients. Arq Neuropsiquiatr. 2005;63(1):87–92.

    Article  PubMed  Google Scholar 

  30. Hillsley RE, Massey EW. Truncal asterixis associated with ceftazidime, a third-generation cephalosporin. Neurology. 1991;41(12):2008.

    Article  CAS  PubMed  Google Scholar 

  31. Chatellier D, Jourdain M, Mangalaboyi J, Ader F, Chopin C, Derambure P, et al. Cefepime-induced neurotoxicity: an underestimated complication of antibiotherapy in patients with acute renal failure. Intensive Care Med. 2002;28(2):214–7.

    Article  CAS  PubMed  Google Scholar 

  32. Fugate JE, Kalimullah EA, Hocker SE, Clark SL, Wijdicks EF, Rabinstein AA. Cefepime neurotoxicity in the intensive care unit: a cause of severe, underappreciated encephalopathy. Crit Care. 2013;17(6):R264.

    Article  PubMed  PubMed Central  Google Scholar 

  33. Hocker S, Rabinstein AA. Cefepime neurotoxicity can mimic postanoxic coma with myoclonic status epilepticus. Neurol Clin Pract. 2011;1(1):73–4.

    Article  Google Scholar 

  34. Lam S, Gomolin IH. Cefepime neurotoxicity: case report, pharmacokinetic considerations, and literature review. Pharmacotherapy. 2006;26(8):1169–74.

    Article  PubMed  Google Scholar 

  35. Grill MF, Maganti R. Cephalosporin-induced neurotoxicity: clinical manifestations, potential pathogenic mechanisms, and the role of electroencephalographic monitoring. Ann Pharmacother. 2008;42(12):1843–50.

    Article  CAS  PubMed  Google Scholar 

  36. Funk MJ, Fusco JS, Cole SR, Thomas JC, Porter K, Kaufman JS, et al. Timing of HAART initiation and clinical outcomes in human immunodeficiency virus type 1 seroconverters. Arch Intern Med. 2011;171(17):1560–9.

    Article  CAS  PubMed Central  Google Scholar 

  37. Kranick SM, Nath A. Neurologic complications of HIV-1 infection and its treatment in the era of antiretroviral therapy. Continuum (Minneap Minn). 2012;18(6 Infectious Disease):1319–37.

    Google Scholar 

  38. McCombe JA, Auer RN, Maingat FG, Houston S, Gill MJ, Power C. Neurologic immune reconstitution inflammatory syndrome in HIV/AIDS: outcome and epidemiology. Neurology. 2009;72(9):835–41.

    Article  CAS  PubMed  Google Scholar 

  39. Antonelli LR, Mahnke Y, Hodge JN, Porter BO, Barber DL, DerSimonian R, et al. Elevated frequencies of highly activated CD4+ T cells in HIV+ patients developing immune reconstitution inflammatory syndrome. Blood. 2010;116(19):3818–27.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Worsley CM, Suchard MS, Stevens WS, Van Rie A, Murdoch DM. Multi-analyte profiling of ten cytokines in South African HIV-infected patients with immune reconstitution inflammatory syndrome (IRIS). AIDS Res Ther. 2010;7:36.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  41. Pfistershammer K, Rieger A, Steinberger P, Kohrgruber N. Identification of PD-1 as a unique marker for discordant immune response in HIV infected patients. Swiss Med Wkly. 2009;139(9–10):28s-s.

    Google Scholar 

  42. Manabe YC, Campbell JD, Sydnor E, Moore RD. Immune reconstitution inflammatory syndrome: risk factors and treatment implications. J Acquir Immune Defic Syndr. 2007;46(4):456–62.

    Article  CAS  PubMed  Google Scholar 

  43. Grant PM, Komarow L, Andersen J, Sereti I, Pahwa S, Lederman MM, et al. Risk factor analyses for immune reconstitution inflammatory syndrome in a randomized study of early vs. deferred ART during an opportunistic infection. PLoS One. 2010;5(7):e11416.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  44. Makadzange AT, Ndhlovu CE, Takarinda K, Reid M, Kurangwa M, Gona P, et al. Early versus delayed initiation of antiretroviral therapy for concurrent HIV infection and cryptococcal meningitis in sub-saharan Africa. Clin Infect Dis. 2010;50(11):1532–8.

    Article  CAS  PubMed  Google Scholar 

  45. Venkataramana A, Pardo CA, McArthur JC, Kerr DA, Irani DN, Griffin JW, et al. Immune reconstitution inflammatory syndrome in the CNS of HIV-infected patients. Neurology. 2006;67(3):383–8.

    Article  CAS  PubMed  Google Scholar 

  46. Falagas ME, Kasiakou SK. Toxicity of polymyxins: a systematic review of the evidence from old and recent studies. Crit Care. 2006;10(1):R27.

    Article  PubMed  PubMed Central  Google Scholar 

  47. Koch-Weser J, Sidel VW, Federman EB, Kanarek P, Finer DC, Eaton AE. Adverse effects of sodium colistimethate. Manifestations and specific reaction rates during 317 courses of therapy. Ann Intern Med. 1970;72(6):857–68.

    Article  CAS  PubMed  Google Scholar 

  48. Fanning WL, Gump DW, Sofferman RA. Side effects of minocycline: a double-blind study. Antimicrob Agents Chemother. 1977;11(4):712–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Jacobson JA, Daniel B. Vestibular reactions associated with minocycline. Antimicrob Agents Chemother. 1975;8(4):453–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Chiu AM, Chuenkongkaew WL, Cornblath WT, Trobe JD, Digre KB, Dotan SA, et al. Minocycline treatment and pseudotumor cerebri syndrome. Am J Ophthalmol. 1998;126(1):116–21.

    Article  CAS  PubMed  Google Scholar 

  51. Tabibian JH, Gutierrez MA. Doxycycline-induced pseudotumor cerebri. South Med J. 2009;102(3):310–1.

    Article  PubMed  Google Scholar 

  52. Becker LD, Miller RD. Clindamycin enhances a nondepolarizing neuromuscular blockade. Anesthesiology. 1976;45(1):84–7.

    Article  CAS  PubMed  Google Scholar 

  53. al Ahdal O, Bevan DR. Clindamycin-induced neuromuscular blockade. Can J Anaesth. 1995;42(7):614–7.

    Article  PubMed  Google Scholar 

  54. Best JA, Marashi AH, Pollan LD. Neuromuscular blockade after clindamycin administration: a case report. J Oral Maxillofac Surg. 1999;57(5):600–3.

    Article  CAS  PubMed  Google Scholar 

  55. Ahmed A, Laes DJ, Bressler EL. Reversible magnetic-resonance-imaging findings in metronidazole-induced encephalopathy. Neurology. 1995;45(3):588–9.

    Article  CAS  PubMed  Google Scholar 

  56. Woodruff BK, Wijdicks EFM, Marshall WF. Reversible metronidazole-induced lesions of the cerebellar dentate nuclei. N Engl J Med. 2002;346(1):68–9.

    Article  PubMed  Google Scholar 

  57. Horlen CK, Seifert CF, Malouf CS. Toxic metronidazole-induced MRI changes. Ann Pharmacother. 2000;34(11):1273–5.

    Article  CAS  PubMed  Google Scholar 

  58. Hemphill JC, Greenberg SM, Anderson CS, Becker K, Bendok BR, Cushman M, et al. Guidelines for the management of spontaneous intracerebral hemorrhage a guideline for healthcare professionals from the American Heart Association/American Stroke Association. Stroke. 2015;46(7):2032–60.

    Article  PubMed  Google Scholar 

  59. Phan TG, Koh M, Wijdicks EFM. Safety of discontinuation of anticoagulation in patients with intracranial hemorrhage at high thromboembolic risk. Arch Neurol. 2000;57(12):1710–3.

    Article  CAS  PubMed  Google Scholar 

  60. Romualdi E, Micieli E, Ageno W, Squizzato A. Oral anticoagulant therapy in patients with mechanical heart valve and intracranial haemorrhage: a systematic review. Thromb Haemost. 2009;101(2):290–7.

    CAS  PubMed  Google Scholar 

  61. Hacke W. The dilemma of reinstituting anticoagulation for patients with cardioembolic sources and intracranial hemorrhage: how wide is the strait between Skylla and Karybdis? Arch Neurol. 2000;57(12):1682–4.

    Article  CAS  PubMed  Google Scholar 

  62. Majeed A, Kim YK, Roberts RS, Holmstrom M, Schulman S. Optimal timing of resumption of warfarin after intracranial hemorrhage. Stroke. 2010;41(12):2860–6.

    Article  CAS  PubMed  Google Scholar 

  63. He J, Whelton PK, Vu B, Klag MJ. Aspirin and risk of hemorrhagic stroke: a meta-analysis of randomized controlled trials. JAMA. 1998;280(22):1930–5.

    Article  CAS  PubMed  Google Scholar 

  64. Thompson BB, Bejot Y, Caso V, Castillo J, Christensen H, Flaherty ML, et al. Prior antiplatelet therapy and outcome following intracerebral hemorrhage: a systematic review. Neurology. 2010;75(15):1333–42.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Weitz JI, Semchuk W, Turpie AG, Fisher WD, Kong C, Ciaccia A, et al. Trends in prescribing oral anticoagulants in Canada, 2008-2014. Clin Ther. 2015;37(11):2506–14 e4.

    Article  PubMed  Google Scholar 

  66. Eckman MH. Decision-making about the use of non-vitamin K oral anticoagulant therapies for patients with atrial fibrillation. J Thromb Thrombolysis. 2015;41(2):234–40.

    Article  CAS  Google Scholar 

  67. Pollack Jr CV, Reilly PA, Eikelboom J, Glund S, Verhamme P, Bernstein RA, et al. Idarucizumab for dabigatran reversal. N Engl J Med. 2015;373(6):511–20. The studies regarding reversal of NOACs will likely have widespread impact on the trend of increasing use of NOACs along with management of acute intracranial hemorrhage associated with concurrent NOAC use. The report associating seizures with use of tranexamic acid, we hope, will prevent morbidity and mortality, as many clinicians are unaware tranexamic acid causes seizures. The report may alter future trends in use of the antifibrinolytic.

    Article  CAS  PubMed  Google Scholar 

  68. Siegal DM, Curnutte JT, Connolly SJ, Lu G, Conley PB, Wiens BL, et al. Andexanet alfa for the reversal of factor Xa inhibitor activity. N Engl J Med. 2015;373:2413–24. The studies regarding reversal of NOACs will likely have widespread impact on the trend of increasing use of NOACs along with management of acute intracranial hemorrhage associated with concurrent NOAC use. The report associating seizures with use of tranexamic acid, we hope, will prevent morbidity and mortality, as many clinicians are unaware tranexamic acid causes seizures. The report may alter future trends in use of the antifibrinolytic.

    Article  CAS  PubMed  Google Scholar 

  69. Sarich TC, Seltzer JH, Berkowitz SD, Costin J, Curnutte JT, Gibson CM, et al. Novel oral anticoagulants and reversal agents: considerations for clinical development. Am Heart J. 2015;169(6):751–7.

    Article  CAS  PubMed  Google Scholar 

  70. Lecker I, Wang DS, Whissell PD, Avramescu S, Mazer CD, Orser BA. Tranexamic acid-associated seizures: causes and treatment. Ann Neurol. 2015;79(1):18–26. The studies regarding reversal of NOACs will likely have widespread impact on the trend of increasing use of NOACs along with management of acute intracranial hemorrhage associated with concurrent NOAC use. The report associating seizures with use of tranexamic acid, we hope, will prevent morbidity and mortality, as many clinicians are unaware tranexamic acid causes seizures. The report may alter future trends in use of the antifibrinolytic.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  71. Apixaban prescribing information: Bristol-Myers Squibb. Available from: http://packageinserts.bms.com/pi/pi_eliquis.pdf.

  72. Dabigatran etexilate mesylate prescribing information]. Available from: http://docs.boehringer-ingelheim.com/Prescribing%20Information/PIs/Pradaxa/Pradaxa.pdf.

  73. Edoxaban prescribing information. Edoxaban prescribing information]. Available from: https://docs.google.com/viewer?url=http%3A%2F%2Fwww.accessdata.fda.gov%2Fdrugsatfda_docs%2Flabel%2F2015%2F206316lbl.pdf.

  74. Rivaroxaban prescribing information. Rivaroxaban prescribing information]. Available from: https://docs.google.com/viewer?url=http%3A%2F%2Fwww.accessdata.fda.gov%2Fdrugsatfda_docs%2Flabel%2F2013%2F022406s004lbl.pdf.

  75. Callegari E, Malhotra B, Bungay PJ, Webster R, Fenner KS, Kempshall S, et al. A comprehensive non-clinical evaluation of the CNS penetration potential of antimuscarinic agents for the treatment of overactive bladder. Br J Clin Pharmacol. 2011;72(2):235–46.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Clegg A, Young JB. Which medications to avoid in people at risk of delirium: a systematic review. Age Ageing. 2011;40(1):23–9.

    Article  PubMed  Google Scholar 

  77. American Psychiatric Association: Diagnostic and Statistical Manual of Mental Disorders, Fifth Edition. Arlington, VA, American Psychiatric Association, 2013.

    Google Scholar 

  78. National Guideline C. Delirium: diagnosis, prevention and management.

  79. Ely EW, Shintani A, Truman B, Speroff T, Gordon SM, Harrell Jr FE, et al. Delirium as a predictor of mortality in mechanically ventilated patients in the intensive care unit. JAMA. 2004;291(14):1753–62.

    Article  CAS  PubMed  Google Scholar 

  80. Hensley ML, Peterson B, Silver RT, Larson RA, Schiffer CA, Szatrowski TP. Risk factors for severe neuropsychiatric toxicity in patients receiving interferon alfa-2b and low-dose cytarabine for chronic myelogenous leukemia: analysis of cancer and leukemia group B 9013. J Clin Oncol. 2000;18(6):1301–8.

    CAS  PubMed  Google Scholar 

  81. Denicoff KD, Rubinow DR, Papa MZ, Simpson C, Seipp CA, Lotze MT, et al. The neuropsychiatric effects of treatment with interleukin-2 and lymphokine-activated killer cells. Ann Intern Med. 1987;107(3):293–300.

    Article  CAS  PubMed  Google Scholar 

  82. FDA-approved prescribing information (2015). Available from: http://www.accessdata.fda.gov/drugsatfda_docs/label/2013/125427lbl.pdf?et_cid=31141095&et_rid=463638624&linkid=http%3a%2f%2fwww.accessdata.fda.gov%2fdrugsatfda_docs%2flabel%2f2013%2f125427lbl.pdf.

  83. Seet RC, Rabinstein AA. Clinical features and outcomes of posterior reversible encephalopathy syndrome following bevacizumab treatment. QJM. 2012;105(1):69–75.

    Article  CAS  PubMed  Google Scholar 

  84. Pro B, Advani R, Brice P, Bartlett NL, Rosenblatt JD, Illidge T, et al. Brentuximab vedotin (SGN-35) in patients with relapsed or refractory systemic anaplastic large-cell lymphoma: results of a phase II study. J Clin Oncol. 2012;30(18):2190–6.

    Article  CAS  PubMed  Google Scholar 

  85. Norden AD, Bartolomeo J, Tanaka S, Drappatz J, Ciampa AS, Doherty LM, et al. Safety of concurrent bevacizumab therapy and anticoagulation in glioma patients. J Neuro-Oncol. 2012;106(1):121–5.

    Article  CAS  Google Scholar 

  86. Carson KR, Evens AM, Richey EA, Habermann TM, Focosi D, Seymour JF, et al. Progressive multifocal leukoencephalopathy after rituximab therapy in HIV-negative patients: a report of 57 cases from the research on adverse drug events and reports project. Blood. 2009;113(20):4834–40.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  87. Major EO, Amemiya K, Tornatore CS, Houff SA, Berger JR. Pathogenesis and molecular biology of progressive multifocal leukoencephalopathy, the JC virus-induced demyelinating disease of the human brain. Clin Microbiol Rev. 1992;5(1):49–73.

    CAS  PubMed  PubMed Central  Google Scholar 

  88. Mavragani CP, Vlachoyiannopoulos PG, Kosmas N, Boletis I, Tzioufas AG, Voulgarelis M. A case of reversible posterior leucoencephalopathy syndrome after rituximab infusion. Rheumatology (Oxford). 2004;43(11):1450–1.

    Article  CAS  Google Scholar 

  89. Argyriou AA, Iconomou G, Kalofonos HP. Bortezomib-induced peripheral neuropathy in multiple myeloma: a comprehensive review of the literature. Blood. 2008;112(5):1593–9.

    Article  CAS  PubMed  Google Scholar 

  90. Fugate JE, Rabinstein AA. Posterior reversible encephalopathy syndrome: clinical and radiological manifestations, pathophysiology, and outstanding questions. Lancet Neurol. 2015;14(9):914–25.

    Article  PubMed  Google Scholar 

  91. Graffeo CS, Dawson ET, Murphy ME, Black DF, Parisi JE, Crum BA, et al. Expanding the spectrum of subacute diencephalic angioencephalopathy. J Clin Neurosci. 2015;23:8–13.

    Article  PubMed  Google Scholar 

  92. Wijdicks EF, Wiesner RH, Krom RA. Neurotoxicity in liver transplant recipients with cyclosporine immunosuppression. Neurology. 1995;45(11):1962–4.

    Article  CAS  PubMed  Google Scholar 

  93. de Groen PC, Aksamit AJ, Rakela J, Forbes GS, Krom RA. Central nervous system toxicity after liver transplantation. The role of cyclosporine and cholesterol. N Engl J Med. 1987;317(14):861–6.

    Article  PubMed  Google Scholar 

  94. Bowyer SL, LaMothe MP, Hollister JR. Steroid myopathy: incidence and detection in a population with asthma. J Allergy Clin Immunol. 1985;76(2 Pt 1):234–42.

    Article  CAS  PubMed  Google Scholar 

  95. Hermans G, Van den Berghe G. Clinical review: intensive care unit acquired weakness. Crit Care. 2015;19:274.

    Article  PubMed  PubMed Central  Google Scholar 

  96. Wolkowitz OM, Burke H, Epel ES, Reus VI. Glucocorticoids. Mood, memory, and mechanisms. Ann N Y Acad Sci. 2009;1179:19–40.

    Article  CAS  PubMed  Google Scholar 

  97. Lee EQW, P. Y Wen. Overview of neurologic complications of non-platinum cancer chemotherapy 2015. Available from: http://www.uptodate.com/contents/overview-of-neurologic-complications-of-non-platinum-cancer-chemotherapy?source=search_result&search=taxanes&selectedTitle=1~150#H24.

  98. Nolan CP, DeAngelis LM. Neurologic complications of chemotherapy and radiation therapy. Continuum (Minneap Minn). 2015;21(2 Neuro-oncology):429–51.

    Google Scholar 

  99. Quasthoff S, Hartung HP. Chemotherapy-induced peripheral neuropathy. J Neurol. 2002;249(1):9–17.

    Article  CAS  PubMed  Google Scholar 

  100. Cavaletti G, Fabbrica D, Minoia C, Frattola L, Tredici G. Carboplatin toxic effects on the peripheral nervous system of the rat. Ann Oncol. 1998;9(4):443–7.

    Article  CAS  PubMed  Google Scholar 

  101. Grunberg SM, Sonka S, Stevenson LL, Muggia FM. Progressive paresthesias after cessation of therapy with very high-dose Cisplatin. Cancer Chemother Pharmacol. 1989;25(1):62–4.

    Article  CAS  PubMed  Google Scholar 

  102. LoMonaco M, Milone M, Batocchi AP, Padua L, Restuccia D, Tonali P. Cisplatin neuropathy: clinical course and neurophysiological findings. J Neurol. 1992;239(4):199–204.

    Article  CAS  PubMed  Google Scholar 

  103. Thompson SW, Davis LE, Kornfeld M, Hilgers RD, Standefer JC. Cisplatin neuropathy. Clinical, electrophysiologic, morphologic, and toxicologic studies. Cancer. 1984;54(7):1269–75.

    Article  CAS  PubMed  Google Scholar 

  104. Extra JM, Marty M, Brienza S, Misset JL. Pharmacokinetics and safety profile of oxaliplatin. Semin Oncol. 1998;25(2 Suppl 5):13–22.

    CAS  PubMed  Google Scholar 

  105. Moes DJ, Guchelaar HJ, de Fijter JW. Sirolimus and everolimus in kidney transplantation. Drug Discov Today. 2015;20(10):1243–9.

    Article  CAS  PubMed  Google Scholar 

  106. FDA approval for everolimus: National Cancer Institute. Available from: http://www.cancer.gov/about-cancer/treatment/drugs/fda-everolimus.

  107. Maramattom BVW, Wijdicks EF. Sirolimus may not cause neurotoxicity in kidney and liver transplant recipients. Neurology. 2004;63(10):1958–9.

    Article  CAS  PubMed  Google Scholar 

  108. Barbas AS, Rege AS, Castleberry AW, Gommer J, Ellis MJ, Brennan TV, et al. Posterior reversible encephalopathy syndrome independently associated with tacrolimus and sirolimus after multivisceral transplantation. Am J Transplant. 2013;13(3):808–10.

    Article  CAS  PubMed  Google Scholar 

  109. Touhami S, Arzouk N, Darugar A, Heron E, Clarencon F, Bodaghi B, et al. Everolimus-induced posterior reversible encephalopathy syndrome and bilateral optic neuropathy after kidney transplantation. Transplantation. 2014;98(12):e102–4.

    Article  PubMed  Google Scholar 

  110. Chaudhry V, Eisenberger MA, Sinibaldi VJ, Sheikh K, Griffin JW, Cornblath DR. A prospective study of suramin-induced peripheral neuropathy. Brain. 1996;119(Pt 6):2039–52.

    Article  PubMed  Google Scholar 

  111. Rosenberg B, Vancamp L, Krigas T. Inhibition of cell division in Escherichia coli by electrolysis products from a platinum electrode. Nature. 1965;205:698–9.

    Article  CAS  PubMed  Google Scholar 

  112. Hilkens PHE, Verweij J, Stoter G, Vecht CJ, Van Putten WLJ, vanden Bent MJ. Peripheral neurotoxicity induced by docetaxel. Neurology. 1996;46(1):104–8.

    Article  CAS  PubMed  Google Scholar 

  113. Mileshkin L, Stark R, Day B, Seymour JF, Zeldis JB, Prince HM. Development of neuropathy in patients with myeloma treated with thalidomide: patterns of occurrence and the role of electrophysiologic monitoring. J Clin Oncol. 2006;24(27):4507–14.

    Article  CAS  PubMed  Google Scholar 

  114. Tanner KD, Levine JD, Topp KS. Microtubule disorientation and axonal swelling in unmyelinated sensory axons during vincristine-induced painful neuropathy in rat. J Comp Neurol. 1998;395(4):481–92.

    Article  CAS  PubMed  Google Scholar 

  115. Bakshi N, Maselli RA, Gospe Jr SM, Ellis WG, McDonald C, Mandler RN. Fulminant demyelinating neuropathy mimicking cerebral death. Muscle Nerve. 1997;20(12):1595–7.

    Article  CAS  PubMed  Google Scholar 

  116. Graf WD, Chance PF, Lensch MW, Eng LJ, Lipe HP, Bird TD. Severe vincristine neuropathy in Charcot-Marie-Tooth disease type 1A. Cancer. 1996;77(7):1356–62.

    Article  CAS  PubMed  Google Scholar 

  117. Chou R, Peterson K, Helfand M. Comparative efficacy and safety of skeletal muscle relaxants for spasticity and musculoskeletal conditions: a systematic review. J Pain Symptom Manage. 2004;28(2):140–75.

    Article  CAS  PubMed  Google Scholar 

  118. Hunter JM. New neuromuscular blocking drugs. N Engl J Med. 1995;332(25):1691–9.

    Article  CAS  PubMed  Google Scholar 

  119. Chollet F, Tardy J, Albucher JF, Thalamas C, Berard E, Lamy C, et al. Fluoxetine for motor recovery after acute ischaemic stroke (FLAME): a randomised placebo-controlled trial. Lancet Neurol. 2011;10(2):123–30.

    Article  CAS  PubMed  Google Scholar 

  120. Waldorf S. AANA journal course. Update for nurse anesthetists. Neuroleptic malignant syndrome. AANA J. 2003;71(5):389–94.

    PubMed  Google Scholar 

  121. Shalev A, Hermesh H, Munitz H. Mortality from neuroleptic malignant syndrome. J Clin Psychiatry. 1989;50(1):18–25.

    CAS  PubMed  Google Scholar 

  122. Mizuno Y, Takubo H, Mizuta E, Kuno S. Malignant syndrome in Parkinson’s disease: concept and review of the literature. Parkinsonism Relat Disord. 2003;9 Suppl 1:S3–9.

    Article  PubMed  Google Scholar 

  123. Gleason KM, McDaniel MR, Feinglass J, Baker DW, Lindquist L, Liss D, et al. Results of the Medications At Transitions and Clinical Handoffs (MATCH) study: an analysis of medication reconciliation errors and risk factors at hospital admission. J Gen Intern Med. 2010;25(5):441–7.

    Article  PubMed  PubMed Central  Google Scholar 

  124. Pierre JM. Extrapyramidal symptoms with atypical antipsychotics: incidence, prevention and management. Drug Saf. 2005;28(3):191–208.

    Article  CAS  PubMed  Google Scholar 

  125. Hawthorne JM, Caley CF. Extrapyramidal reactions associated with serotonergic antidepressants. Ann Pharmacother. 2015;49(10):1136–52.

    Article  PubMed  Google Scholar 

  126. Strange PG. Antipsychotic drugs: importance of dopamine receptors for mechanisms of therapeutic actions and side effects. Pharmacol Rev. 2001;53(1):119–33.

    CAS  PubMed  Google Scholar 

  127. Lima AR, Soares-Weiser K, Bacaltchuk J, Barnes TR. Benzodiazepines for neuroleptic-induced acute akathisia. Cochrane Database Syst Rev. 2002;1:CD001950.

    PubMed  Google Scholar 

  128. Adler L, Duncan E, Angrist B, Hemdal P, Rotrosen J, Slotnick V. Effects of a specific beta 2-receptor blocker in neuroleptic-induced akathisia. Psychiatry Res. 1989;27(1):1–4.

    Article  CAS  PubMed  Google Scholar 

  129. Kramer MS, Gorkin R, DiJohnson C. Treatment of neuroleptic-induced akathisia with propranolol: a controlled replication study. Hillside J Clin Psychiatry. 1989;11(2):107–19.

    CAS  PubMed  Google Scholar 

  130. Adler LA, Peselow E, Rosenthal M, Angrist B. A controlled comparison of the effects of propranolol, benztropine, and placebo on akathisia: an interim analysis. Psychopharmacol Bull. 1993;29(2):283–6.

    CAS  PubMed  Google Scholar 

  131. Albanese A, Bhatia K, Bressman SB, Delong MR, Fahn S, Fung VS, et al. Phenomenology and classification of dystonia: a consensus update. Mov Disord. 2013;28(7):863–73.

    Article  PubMed  PubMed Central  Google Scholar 

  132. Geyer HL, Bressman SB. The diagnosis of dystonia. Lancet Neurol. 2006;5(9):780–90.

    Article  PubMed  Google Scholar 

  133. Gelb DJ, Oliver E, Gilman S. Diagnostic criteria for Parkinson disease. Arch Neurol. 1999;56(1):33–9.

    Article  CAS  PubMed  Google Scholar 

  134. Fann WE, Lake CR. Amantadine versus trihexyphenidyl in the treatment of neuroleptic-induced Parkinsonism. Am J Psychiatry. 1976;133(8):940–3.

    Article  CAS  PubMed  Google Scholar 

  135. DiMascio A, Bernardo DL, Greenblatt DJ, Marder JE. A controlled trial of amantadine in drug-induced extrapyramidal disorders. Arch Gen Psychiatry. 1976;33(5):599–602.

    Article  CAS  PubMed  Google Scholar 

  136. Boyer EW. The serotonin syndrome. N Engl J Med. 2005;352(11):1112–20.

    Article  CAS  PubMed  Google Scholar 

  137. Graudins A, Stearman A, Chan B. Treatment of the serotonin syndrome with cyproheptadine. J Emerg Med. 1998;16(4):615–9.

    Article  CAS  PubMed  Google Scholar 

  138. Mackay FJ, Dunn NR, Mann RD. Antidepressants and the serotonin syndrome in general practice. Br J Gen Pract. 1999;49(448):871–4.

    CAS  PubMed  PubMed Central  Google Scholar 

  139. Dunkley EJ, Isbister GK, Sibbritt D, Dawson AH, Whyte IM. The hunter serotonin toxicity criteria: simple and accurate diagnostic decision rules for serotonin toxicity. QJM. 2003;96(9):635–42.

    Article  CAS  PubMed  Google Scholar 

  140. Pedavally S, Fugate JE, Rabinstein AA. Serotonin syndrome in the intensive care unit: clinical presentations and precipitating medications. Neurocrit Care. 2014;21(1):108–13.

    Article  CAS  PubMed  Google Scholar 

  141. Charness ME, Morady F, Scheinman MM. Frequent neurologic toxicity associated with amiodarone therapy. Neurology. 1984;34(5):669–71.

    Article  CAS  PubMed  Google Scholar 

  142. Bateman DN, Bevan P, Longley BP, Mastaglia F, Wandless I. Cimetidine induced postural and action tremor. J Neurol Neurosurg Psychiatry. 1981;44(1):94.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  143. Chopra A, Kolla BP, Mansukhani MP, Netzel P, Frye MA. Valproate-induced hyperammonemic encephalopathy: an update on risk factors, clinical correlates and management. Gen Hosp Psychiatry. 2012;34(3):290–8.

    Article  PubMed  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding authors

Correspondence to Elliot T. Dawson or Sara E. Hocker.

Ethics declarations

Conflict of Interest

Elliot T. Dawson and Sara E. Hocker declare that they have no conflict of interest.

Human and Animal Rights and Informed Consent

This article does not contain any studies with human or animal subjects performed by any of the authors.

Additional information

This article is part of the Topical Collection on Neurology of Systemic Diseases

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Dawson, E.T., Hocker, S.E. Neurologic Complications of Commonly Used Drugs in the Hospital Setting. Curr Neurol Neurosci Rep 16, 35 (2016). https://doi.org/10.1007/s11910-016-0636-7

Download citation

  • Published:

  • DOI: https://doi.org/10.1007/s11910-016-0636-7

Keywords

Navigation