Loading Dose

Whether or not V.S. has a detectable serum concentration of carbamazepine, he

should be given an IV loading dose of either fosphenytoin (20 mg/kg PE IV at 150

mg/minute) or phenytoin (20 mg/kg IV at 50 mg/minute). After administration of

either of these loading doses, serum phenytoin concentrations should remain greater

than 10 mcg/mLfor approximately 24 hours; this will allow time for determination of

V.S.’s serum carbamazepine concentration and estimation of an appropriate

maintenance dose of oral carbamazepine once oral therapy can be restarted. In this

setting, the use of IV phenytoin or fosphenytoin is a temporary measure. V.S.’s

previous positive response to carbamazepine indicates that he should likely continue

to receive this drug as his oral maintenance medication.

IV phenytoin can be administered by direct injection into a running IV line. The

rate of administration should be no faster than 50 mg/minute to minimize the risk of

hypotension and acute cardiac arrhythmias. Cardiovascular status (blood pressure,

electrocardiogram) should be monitored closely during administration. Hypotension

or electrocardiographic abnormalities usually reverse if the administration of

phenytoin is slowed or stopped temporarily. Fosphenytoin can be given by either

direct IV injection or, after dilution in any suitable IV solution, by infusion at up to

150 mg PE/minute.

100 Absence of propylene glycol as a diluent renders fosphenytoin

potentially less likely than phenytoin to cause cardiovascular adverse effects, but this

advantage is not well supported.

220 Electrocardiographic and blood pressure

monitoring is recommended when this drug is given IV. Pruritus and paresthesias,

usually localized to the face and groin, are relatively common side effects during IV

fosphenytoin administration. These sensations are not allergic reactions to the

medication. Their occurrence is related to the administration rate, and they are

reversible with temporary discontinuation or slowing of the injection.

100

p. 1299

p. 1300

Maintenance Therapy

The undetectable serum carbamazepine concentration appears to confirm the role of

nonadherence in this episode of SE. Because V.S. was previously well controlled on

600 mg/day, this would be a reasonable target dose. Because V.S. may not tolerate

carbamazepine if restarted at the prior maintenance dose, gradually escalating up to

this dose should be initiated as soon as V.S. can take oral medication. V.S. should be

counseled regarding the importance of taking his medication according to directions

and any potential barriers to adherence should be addressed.

Alternative Therapies for Refractory Status Epilepticus

Phenobarbital may be useful for treatment of SE if the patient cannot tolerate

phenytoin or when seizures continue after administration of appropriate loading

doses of phenytoin. Patients who receive phenobarbital after being treated with IV

benzodiazepines should be monitored closely for respiratory depression because this

effect may be additive. Equipment and personnel to provide ventilatory assistance

should be available.

208

Status epilepticus that does not respond to lorazepam and a longer-acting agent

(e.g., phenytoin/fosphenytoin, valproate, levetiracetam, phenobarbital, or

lacosamide) is considered refractory status epilepticus. From 20% to 40% of

patients with SE will progress to refractory status epilepticus. In such circumstances,

an alternative longer-acting agent from those mentioned above may be considered;

and, in recent years, the use of such “third-line” agents was a common practice.

Recently, it has become a more common practice to escalate therapy to the use of an

anesthetic agent after failure of second-line therapy.

209 The most commonly used

anesthetic agents for refractory status epilepticus are midazolam and propofol;

pentobarbital is also sometimes used. Significant respiratory depression is expected

with these therapies; patients require intubation and mechanical ventilation. In

addition, vasopressors, such as dopamine or dobutamine, may be required to control

hypotension. Constant EEG monitoring also is required to assess the anticonvulsant

effect of the drug and to gauge the level of anesthesia.

Midazolam is given as a loading dose of 0.2 mg/kg IV and is followed by an IV

infusion of 0.2 to 0.6 mg/kg/hour.

208 Many practitioners adjust the infusion rate to

either control electrographic seizures and/or to produce a burst-suppression EEG

pattern.

221 Most protocols for anesthetic therapy of refractory SE recommend attempts

at gradually reducing the dose of medication after 12 to 24 hours of treatment. If

clinical or EEG seizure activity recurs, the dose is increased again to produce the

desired EEG pattern. Anesthetic agents may need to be continued for several days or

even weeks in some patients.

Continuous IV infusions of propofol or midazolam are also useful for refractory

SE. These therapies appear to be less likely than pentobarbital to cause severe

hypotension that is refractory to vasopressors.

222–225 Because no direct comparative

trials of pentobarbital, propofol, and midazolam have been performed, physician

familiarity and preference often guide the choice between these agents for the

treatment of refractory SE.

KEY REFERENCES AND WEBSITES

A full list of references for this chapter can be found at

http://thepoint.lww.com/AT11e. Below are the key references and websites for this

chapter, with the corresponding reference number in this chapter found in parentheses

after the reference.

Key References

Bialer M et al. Progress report on new antiepileptic drugs: a summary of the Twelfth Eilat Conference (EILAT

XII). Epilepsy Res. 2015;111:85. (86)

Werhahn KJ et al. A randomized, double-blind comparison of antiepileptic drug treatment in the elderly with newonset focal epilepsy. Epilepsia. 2015;56:450. (115)

French JA, Pedley TA. Clinical practice. Initial management of epilepsy. N EnglJ Med. 2008;359:166. (3)

Glauser T et al. Evidence-based guideline: treatment of convulsive status epilepticus in children and adults: report

of the Guideline Committee of the American Epilepsy Society. Epilepsy Curr. 2016;16:48–61. (225)

Glauser TA et al. Ethosuximide, valproic acid, and lamotrigine in childhood absence epilepsy. N Engl J Med.

2010;362:790. (123)

Harden CL et al. Management issues for women with epilepsy-Focus on pregnancy (an evidence-based review): I.

Obstetrical complications and change in seizure frequency, II. Teratogenesis and perinatal outcomes, III.

Vitamin K, folic acid, blood levels, and breast-feeding. Report of the Quality Standards Subcommittee and

Therapeutics and Technology Subcommittee of the American Academy of Neurology and the American

Epilepsy Society. Epilepsia. 2009;50:1229. (187)

Krumholz A et al. Evidence-based guideline: management of an unprovoked first seizure in adults: report of the

Guideline Development Subcommittee of the American Academy of Neurology and the American Epilepsy

Society. Neurology. 2015;84:1705. (4)

Marson AG et al. The SANAD study of effectiveness of valproate, lamotrigine, or topiramate for generalised and

unclassifiable epilepsy: an unblinded randomised controlled trial. Lancet. 2007;369:1016. (124)

Meador KJ et al. Fetal antiepileptic drug exposure and cognitive outcomes at age 6 years (NEAD study): a

prospective observationalstudy. Lancet Neurol. 2013;12:244. (193)

Potschka H. Transporter hypothesis of drug-resistant epilepsy: challenges for pharmacogenetic approaches.

Pharmacogenomics. 2010;11:1427. (87)

Betjemann JP, Lowenstein DH. Status epilepticus in adults. Lancet Neurol. 2015;14:615. (209)

Key Websites

American Epilepsy Society. http://www.aesnet.org.

Epilepsy and Seizure Information for Patients and Health Professionals. http://www.epilepsy.com.

AED Pregnancy Registry. http://www.aedpregnancyregistry.org/.

COMPLETE REFERENCES CHAPTER 60 SEIZURE

DISORDERS

[No authors listed]. Guidelines for epidemiologic studies on epilepsy. Commission on Epidemiology and Prognosis,

International League Against Epilepsy. Epilepsia. 1993;34:592.

Fisher RS et al. Epileptic seizures and epilepsy: definitions proposed by the International League Against Epilepsy

(ILAE) and the International Bureau for Epilepsy (IBE). Epilepsia. 2005;46:470.

French JA, Pedley TA. Clinical practice. Initial management of epilepsy. N EnglJ Med. 2008;359:166.

Krumholz A et al. Evidence-based guideline: management of an unprovoked first seizure in adults: report of the

Guideline Development Subcommittee of the American Academy of Neurology and the American Epilepsy

Society. Neurology. 2015;84:1705–1713.

Luders HO et al. Classification of seizures. In: Wyllie E et al, eds. The Treatment of Epilepsy: Principles and

Practice. 3rd ed. Philadelphia, PA: Lippincott Williams & Wilkins; 2001:287.

Dreifuss FE. Classification of epileptic seizures and the epilepsies. Pediatr Clin North Am. 1989;36:265.

Dreifuss FE. The epilepsies: clinical implications of the international classification. Epilepsia. 1990;31(Suppl 3):S3.

Berg AT et al. Revised terminology and concepts for organization of seizures and epilepsies: Report of the ILAE

Commission on Classification and Terminology, 2005–2009. Epilepsia. 2010;51:676.

Bronen RA et al. Refractory epilepsy: comparison of MR imaging, CT, and histopathologic findings in 117 patients.

Radiology. 1996;201:97.

Berg AT et al. Discontinuing antiepileptic drugs. In: Engel J et al, eds. Epilepsy: A Comprehensive Textbook .

Philadelphia, PA: Lippincott-Raven; 1998:1275.

Callaghan N et al. Withdrawal of anticonvulsant drugs in patients free of seizures for two years: a prospective

study [published correction appears in N EnglJ Med. 1988;319:188]. N EnglJ Med. 1988;318:942.

Matricardi M et al. Outcome after discontinuation of antiepileptic drug therapy in children with epilepsy. Epilepsia.

1989;30:582.

Wiebe S et al. A randomized, controlled trial of surgery for temporal-lobe epilepsy. N EnglJ Med. 2001;345:311.

Bainbridge JL et al. The ketogenic diet. Central Nervous System Practice and Research Network of the

American College of Clinical Pharmacy. Pharmacotherapy. 1999;19:782.

Hassan AM et al. Ketogenic diet in the treatment of refractory epilepsy in childhood. Pediatr Neurol.

1999;21:548.

Tecoma ES, Iragui VJ. Vagus nerve stimulation use and effect in epilepsy: what have we learned? Epilepsy

Behav. 2006;8:127.

Kwan P, Brodie MJ. Early identification of refractory epilepsy. N EnglJ Med. 2000;342:314.

Devinsky O. Patients with refractory seizures. N EnglJ Med. 1999;340:1565.

Perucca E et al. Antiepileptic drugs as a cause of worsening seizures. Epilepsia. 1998;39:5–17.

Karceski S et al. Treatment of epilepsy in adults: expert opinion, 2005. Epilepsy Behav. 2005;7(Suppl 1):S1.

French JA et al. Efficacy and tolerability of the new antiepileptic drugs I: treatment of new onset epilepsy: report

of the Therapeutics and Technology Assessment Subcommittee and Quality Standards Subcommittee of the

American Academy of Neurology and the American Epilepsy Society. Neurology. 2004;62:1252.

French JA et al. Efficacy and tolerability of the new antiepileptic drugs II: treatment of refractory epilepsy: report

of the Therapeutics and Technology Assessment Subcommittee and Quality Standards Subcommittee of the

American Academy of Neurology and the American Epilepsy Society. Neurology. 2004;62:1261.

Garnett WR. Antiepileptic drug treatment: outcomes and adherence. Pharmacotherapy. 2000;20:191S.

Schoenenberger RA et al. Appropriateness of antiepileptic drug level monitoring. JAMA. 1995;274:1622.

Choonara IA, Rane A. Therapeutic drug monitoring of anticonvulsants: state of the art. Clin Pharmacokinet.

1990;18:318.

Hayes G, Kootsikas ME. Reassessing the lower end of the phenytoin therapeutic range: a review of the literature.

Ann Pharmacother. 1993;27:1389.

Tomson T et al. Therapeutic monitoring of antiepileptic drugs for epilepsy. Cochrane Database Syst Rev. 2007;

(1):CD002216.

[No authors listed]. Guidelines for therapeutic monitoring of antiepileptic drugs. Commission on Antiepileptic

Drugs, International League Against Epilepsy. Epilepsia. 1993;34:585.

Tozer TN et al. Phenytoin. In: Evans WE et al, eds. Applied Pharmacokinetics: Principles of Therapeutic Drug

Monitoring. 3rd ed. Vancouver, Canada: Applied Therapeutics; 1992.

Levy RH et al. Carbamazepine, valproic acid, phenobarbital, and ethosuximide. In: Evans WE et al, eds. Applied

Pharmacokinetics: Principles of Therapeutic Drug Monitoring. 3rd ed. Vancouver, Canada: Applied

Therapeutics; 1992.

Woo E et al. If a well-stabilized epileptic patient has a subtherapeutic antiepileptic drug level, should the dose be

increased? A randomized prospective study. Epilepsia. 1988;29:129.

Cloyd JC. Pharmacokinetic pitfalls of present antiepileptic medications. Epilepsia. 1991;32(Suppl 5):S53.

Schmidt D. Reduction of two-drug therapy in intractable epilepsy. Epilepsia. 1983;24:368.

Smith DB et al. Results of a nationwide Veterans Administration Cooperative Study comparing the efficacy and

toxicity of carbamazepine, phenobarbital, phenytoin, and primidone. Epilepsia. 1987;28(Suppl 3):S50.

Thompson PJ, Trimble MR. Anticonvulsant drugs and cognitive functions. Epilepsia. 1982;23:531.

Albright P, BruniJ. Reduction of polypharmacy in epileptic patients. Arch Neurol. 1985;42:797.

Prevey ML et al. Improvement in cognitive functioning and mood state after conversion to valproate

monotherapy. Neurology. 1989;39:1640.

Mirza WU et al. Results of antiepileptic drug reduction in patients with multiple handicaps and epilepsy. Drug

Invest. 1993;5:320.

Pellock JM. Efficacy and adverse effects of antiepileptic drugs. Pediatr Clin North Am. 1989;36:435.

Guberman A. Monotherapy or polytherapy for epilepsy? Can J Neurol Sci. 1998;25:S3.

Chadwick DW et al. A double-blind trial of gabapentin monotherapy for newly diagnosed partial seizures.

International Gabapentin Monotherapy Study Group 945–77. Neurology. 1998;51:1282.

Devinsky O et al. Efficacy of felbamate monotherapy in patients undergoing presurgical evaluation of partial

seizures. Epilepsy Res. 1995;20:241.

Sachdeo RC et al. Topiramate monotherapy for partial onset seizures. Epilepsia. 1997;38:294.

Schacter SC. Tiagabine monotherapy in the treatment of partial epilepsy. Epilepsia. 1995;36(Suppl 6):S2.

Beghi E et al. Withdrawal of antiepileptic drugs: guidelines of the Italian League Against Epilepsy. Epilepsia.

2013;54(Suppl 7):2.

Berg AT, Shinnar S. Relapse following discontinuation of antiepileptic drugs: a meta-analysis. Neurology.

1994;44:601.

Tennison M et al. Discontinuing antiepileptic drugs in children with epilepsy. A comparison of a six-week and a

nine-month taper period. N EnglJ Med. 1994;330:1407.

Malow BA et al. Carbamazepine withdrawal: effects of taper rate on seizure frequency. Neurology.

1993;43:2280.

Camfield P, Camfield C. The frequency of intractable seizures after stopping AEDs in seizure-free children with

epilepsy. Neurology. 2005;64:973.

Mattson RH et al. Comparison of carbamazepine, phenobarbital, phenytoin, and primidone in partial and

secondarily generalized tonic-clonic seizures. N EnglJ Med. 1985;313:145.

Mattson RH et al. A comparison of valproate with carbamazepine for the treatment of complex partial seizures

and secondarily generalized tonic-clonic seizures in adults. The Department of Veterans Affairs Epilepsy

Cooperative Study No. 264 Group. N EnglJ Med. 1992;327:765.

Beydoun A et al. Safety and efficacy of divalproex sodium monotherapy in partial epilepsy: a double-blind,

concentration-response design clinical trial. Depakote Monotherapy for Partial Seizures Study Group.

Neurology. 1997;48:182.

Franceschi M et al. Fatal aplastic anemia in a patient treated with carbamazepine. Epilepsia. 1988;29:582.

Pisciotta AV. Carbamazepine: hematological toxicity. In: Woodbury DM, Penry JK, eds. Antiepileptic Drugs. 2nd

ed. New York, NY: Raven Press; 1982:533.

Pellock JM. Carbamazepine side effects in children and adults. Epilepsia. 1987;28(Suppl 3):S64.

Holmes GL. Carbamazepine: adverse effects. In: Levy RH et al, eds. Antiepileptic Drugs. 5th ed. Philadelphia,

PA: Lippincott Williams & Wilkins; 2002:285.

Camfield C et al. Asymptomatic children with epilepsy: little benefit from screening for anticonvulsant-induced

liver, blood, or renal damage. Neurology. 1986;36:838.

Horowitz S et al. Hepatotoxic reactions associated with carbamazepine therapy. Epilepsia. 1988;29:149.

Hadzic N et al. Acute liver failure induced by carbamazepine. Arch Dis Child. 1990;65:315.

Livingston S et al. Carbamazepine (Tegretol) in epilepsy: nine-year follow-up study with special emphasis on

untoward reactions. Dis Nerv System. 1974;35:103.

Tomson T et al. Relationship of intraindividual dose to plasma concentration of carbamazepine: indication of dosedependent induction of metabolism. Ther Drug Monit. 1989;11:533.

Sanchez A et al. Steady-state carbamazepine concentration-dose ratios in epileptic patients. Clin Pharmacokinet.

1986;11:41.

Oles KS, Gal P. Bioequivalency revisited: Epitol versus Tegretol. Neurology. 1993;43:2435.

Crawford P et al. Are there potential problems with generic substitution of antiepileptic drugs? A review of

issues. Seizure. 2006;15:165.

Ting TY et al. Generic lamotrigine versus brand-name Lamictal bioequivalence in patients with epilepsy: a field

test of the FDA bioequivalence standard. Epilepsia. 2015;56(9):1415–1424.

Privitera MD et al. Generic-to-generic lamotrigine switches in people with epilepsy: the randomized controlled

EQUIGEN trial. Lancet Neurol. 2016;pii:S1474–S4422(16)00014-4. doi:10.1016/S1474-4422(16)00014-4. [Epub

ahead of print]

[No authors listed]. Double-blind crossover comparison of Tegretol-XR and Tegretol in patients with epilepsy.

The Tegretol Oros Osmotic Release Delivery System Study Group. Neurology. 1995;45:1703.

Riss JR et al. Administration of Carbatrol to children with feeding tubes. Pediatr Neurol. 2002;27:193.

Verrotti A et al. Eslicarbazepine acetate: an update on efficacy and safety in epilepsy. Epilepsy Res. 2014;108:1–

10.

McLean MJ et al. Safety and tolerability of gabapentin as adjunctive therapy in a large, multicenter study.

Epilepsia. 1999;40:965.

Leppik IE et al. Safety of tiagabine:summary of 53 trials. Epilepsy Res. 1999;33:235.

Guberman AH et al. Lamotrigine-associated rash: risk/benefit considerations in adults and children. Epilepsia.

1999;40:985.

Sirsi D, Safdieh JE. The safety of levetiracetam. Expert Opin Drug Saf. 2007;6:241.

100.

101.

102.

103.

104.

Martinez W et al. Efficacy, safety, and tolerability of oxcarbazepine monotherapy. Epilepsy Behav. 2006;9:448–

456.

Shneker BF, McAuley JW. Pregabalin: a new neuromodulator with broad therapeutic indications. Ann

Pharmacother. 2005;39:2029.

Jones MW. Topiramate—safety and tolerability. Can J Neurol Sci. 1998;25:S13.

Oommen KJ, Mathews S. Zonisamide: a new antiepileptic drug. Clin Neuropharmacol. 1999;22:192.

Goa KL, Sorkin EM. Gabapentin. A review of its pharmacological properties and clinical potential in epilepsy.

Drugs. 1993;46:409.

Harris JA, Murphy JA. Lacosamide: an adjunctive agent for partial-onset seizures and potential therapy for

neuropathic pain. Ann Pharmacother. 2009;43:1809.

Yuen AW et al. Sodium valproate acutely inhibits lamotrigine metabolism. Br J Clin Pharmacol. 1992;33:511.

Anderson GD et al. Bidirectional interaction of valproate and lamotrigine in healthy subjects. Clin Pharmacol

Ther. 1996;60:145.

Besag FM et al. Carbamazepine toxicity with lamotrigine: pharmacokinetic or pharmacodynamic interaction?

Epilepsia. 1998;39:183.

Welty TE et al. Levetiracetam: a different approach to the pharmacotherapy of epilepsy. Ann Pharmacother.

2002;36:296.

Fattore C et al. Induction of ethinylestradiol and levonorgestrel metabolism by oxcarbazepine in healthy women.

Epilepsia. 1999;40:783.

Falcão A et al. Effect of eslicarbazepine acetate on the pharmacokinetics of a combined

ethinylestradiol/levonorgestrel oral contraceptive in healthy women. Epilepsy Res. 2013;105:368–376.

Bialer M et al. Progress report on new antiepileptic drugs: a summary of the Twelfth Eilat Conference (EILAT

XII). Epilepsy Res. 2015;111:85–141.

Potschka H. Transporter hypothesis of drug-resistant epilepsy: challenges for pharmacogenetic approaches.

Pharmacogenomics. 2010;11:1427.

Piana C et al. Implications of pharmacogenetics for the therapeutic use of antiepileptic drugs. Expert Opin Drug

Metab Toxicol. 2014;10:341–358.

Fitton A, Goa KL. Lamotrigine: an update of its pharmacology and therapeutic use in epilepsy. Drugs.

1995;50:691.

Kilpatrick ES et al. Concentration-effect and concentration-toxicity relations with lamotrigine: a prospective study.

Epilepsia. 1996;37:534.

Caudle KE et al. Clinical pharmacogenetics implementation consortium guidelines for CYP2C9 and HLA-B

genotypes and phenytoin dosing. Clin Pharmacol Ther. 2014;96:542–548.

Tozer TN et al. Phenytoin. In: Evans WE et al, eds. Applied Pharmacokinetics: Principles of Therapeutic Drug

Monitoring. 3rd ed. Vancouver, Canada: Lippincott Williams & Wilkins; 1992:25.

Allen JP et al. Phenytoin cumulation kinetics. Clin Pharmacol Ther. 1979;26:445.

Evens RP et al. Phenytoin toxicity and blood levels after a large oral dose. Am J Hosp Pharm. 1980;37:232.

Ludden TM et al. Rate of phenytoin accumulation in man: a simulation study. J Pharmacokinet Biopharm.

1978;6:399.

Kostenbauder HB et al. Bioavailability and single-dose pharmacokinetics of intramuscular phenytoin. Clin

Pharmacol Ther. 1975;18:449.

Serrano EE et al. Plasma diphenylhydantoin values after oral and intramuscular administration of

diphenylhydantoin. Neurology. 1973;23:311.

Serrano EE, Wilder BJ. Intramuscular administration of diphenylhydantoin. Histologic follow-up studies. Arch

Neurol. 1974;31:276.

Jamerson BD et al. Venous irritation related to intravenous administration of phenytoin versus fosphenytoin.

Pharmacotherapy. 1994;14:47.

Fischer JH et al. Fosphenytoin: clinical pharmacokinetics and comparative advantages in the acute treatment of

seizures. Clin Pharmacokinet. 2003;42:33.

Boucher BA. Fosphenytoin: a novel phenytoin prodrug. Pharmacotherapy. 1996;16:777.

Ramsay RE et al. Intramuscular fosphenytoin (Cerebyx) in patients requiring a loading dose of phenytoin.

Epilepsy Res. 1997;28:181.

Butler RT et al. Drug-induced gingival hyperplasia: phenytoin, cyclosporine, and nifedipine. J Am Dent Assoc .

1987;114:56.

Stinnett E et al. New developments in understanding phenytoin-induced gingival hyperplasia. J Am Dent Assoc .

105.

106.

107.

108.

109.

110.

111.

112.

113.

114.

115.

116.

117.

118.

119.

120.

121.

122.

123.

124.

125.

126.

127.

128.

129.

130.

131.

132.

133.

134.

1987;114:814.

Bruni J. Phenytoin and other hydantoins: adverse effects. In: Levy RH et al, eds. Antiepileptic Drugs. 5th ed.

Philadelphia, PA: Lippincott Williams & Wilkins; 2002:605.

Kuruvilla T, Bharucha NE. Cerebellar atrophy after acute phenytoin intoxication. Epilepsia. 1997;38:500.

Rapport RL, 2nd, Shaw CM. Phenytoin-related cerebellar degeneration without seizures. Ann Neurol.

1977;2:437.

So EL, Penry JK. Adverse effects of phenytoin on peripheral nerves and neuromuscular junction: a review.

Epilepsia. 1981;22:467.

Lovelace RE, Horwitz SJ. Peripheral neuropathy in long-term diphenylhydantoin therapy. Arch Neurol.

1968;18:69.

Fraser LA et al. Enzyme-inducing antiepileptic drugs and fractures in people with epilepsy: a systematic review.

Epilepsy Res. 2015;116:59–66.

Sheth RD. Bone health in pediatric epilepsy. Epilepsy Behav. 2004;5(Suppl 2):S30.

Pack AM et al. Bone mass and turnover in women with epilepsy on antiepileptic drug monotherapy. Ann

Neurol. 2005;57:252.

Brodie MJ et al. Multicentre, double-blind, randomised comparison between lamotrigine and carbamazepine in

elderly patients with newly diagnosed epilepsy. The UK lamotrigine elderly study group. Epilepsy Res.

1999;37:81.

Rowan AJ et al. New onset geriatric epilepsy: a randomized study of gabapentin, lamotrigine, and

carbamazepine. Neurology. 2005;64:1868.

Werhahn KJ et al. A randomized, double-blind comparison of antiepileptic drug treatment in the elderly with

new-onset focal epilepsy. Epilepsia. 2015;56:450–459.

Italiano D, Perucca E. Clinical pharmacokinetics of new-generation antiepileptic drugs at the extremes of age:

an update. Clin Pharmacokinet. 2013;52:627–645.

Martin RC et al. Cognitive functioning in community dwelling older adults with chronic partial epilepsy. Epilepsia.

2005;46:298.

Piazzini A et al. Elderly people and epilepsy: cognitive function. Epilepsia. 2006;47(Suppl 5):82.

Bambara JK et al. Medical decision-making abilities in older adults with chronic partial epilepsy. Epilepsy Behav.

2007;10:63.

Coppola G et al. Lamotrigine versus valproic acid as first-line monotherapy in newly diagnosed typical absence

seizures: an open-label, randomized parallel-group study. Epilepsia. 2004;45:1053.

Frank LM et al. Lamictal (lamotrigine) monotherapy for typical absence seizures in children. Epilepsia.

1999;40:973.

Beran RG et al. Double-blind, placebo-controlled, crossover study of lamotrigine in treatment-resistant

generalised epilepsy. Epilepsia. 1998;39:1329.

Glauser TA et al. Ethosuximide, valproic acid, and lamotrigine in childhood absence epilepsy. N Engl J Med.

2010;362:790.

Marson AG et al. The SANAD study of effectiveness of valproate, lamotrigine, or topiramate for generalised

and unclassifiable epilepsy: an unblinded randomised controlled trial. Lancet. 2007;369:1016.

Mattson RH. Antiepileptic drug monotherapy in adults: selection and use in new-onset epilepsy. In: Levy RH et

al, eds. Antiepileptic Drugs. 5th ed. Philadelphia, PA: Lippincott Williams & Wilkins; 2002:72.

Sato S et al. Benzodiazepines: clonazepam. In: Levy RH et al, eds. Antiepileptic Drugs. 4th ed. New York, NY:

Raven Press; 1995:725.

Glauser TA. Succinimides: adverse effects. In: Levy RH et al, eds. Antiepileptic Drugs. 5th ed. Philadelphia,

PA: Lippincott Williams & Wilkins; 2002:658.

Browne TR et al. Absence (petit mal) seizures. In: Browne TR, Feldman RG, eds. Epilepsy: Diagnosis and

Management. Boston, MA: Little Brown; 1983:61.

Livingston S et al. Petit mal epilepsy: results of a prolonged follow-up study of 117 patients. JAMA.

1965;194:227.

Penry JK, So EL. Refractoriness of absence seizures and phenobarbital. Neurology. 1981;31:158.

Snead OC, 3rd, Hosey LC. Exacerbation of seizures in children by carbamazepine. N EnglJ Med. 1985;313:916.

Shields WD, Saslow E. Myoclonic, atonic and absence seizures following institution of carbamazepine therapy in

children. Neurology. 1983;33:1487.

Zaccara G et al. Clinical pharmacokinetics of valproic acid, 1988. Clin Pharmacokinet. 1988;15:367.

Cloyd JC et al. Comparison of sprinkle versus syrup formulations of valproate for bioavailability, tolerance, and

135.

136.

137.

138.

139.

140.

141.

142.

143.

144.

145.

146.

147.

148.

149.

150.

151.

152.

153.

154.

155.

156.

157.

158.

159.

160.

161.

162.

163.

164.

165.

166.

167.

168.

169.

170.

171.

172.

173.

preference. J Pediatr. 1992;120:634.

Dutta S et al. Comparison of the bioavailability of unequal doses of divalproex sodium extended-release

formulation relative to the delayed-release formulation in healthy volunteers. Epilepsy Res. 2002;49:1.

Fischer JH et al. Effect of food on the serum concentration profile of enteric-coated valproic acid. Neurology.

1988;38:1319.

Bauer LA et al. Valproic acid clearance: unbound fraction and diurnal variation in young and elderly adults. Clin

Pharmacol Ther. 1985;37:697.

Tennison MB et al. Valproate metabolites and hepatotoxicity in an epileptic population. Epilepsia. 1988;29:543.

Eadie MJ et al. Valproate-associated hepatotoxicity and its biochemical mechanisms. Med Toxicol. 1988;3:85.

Dreifuss FE et al. Valproic acid hepatic fatalities: a retrospective review. Neurology. 1987;37:379.

Dreifuss FE et al. Valproic acid hepatic fatalities. II. U.S. experience since 1984. Neurology. 1989;39:201.

Dreifuss FE. Valproic acid hepatic fatalities: revised table. Neurology. 1989;39:1558.

Scheffner E et al. Fatal liver failure in 16 children with valproate therapy. Epilepsia. 1988;29:530.

Koenig SA et al. Valproic acid-induced hepatopathy: nine new fatalities in Germany from 1994 to 2003.

Epilepsia. 2006;47:2027.

Willmore LJ. Clinical manifestations of valproate hepato-toxicity. In: Levy RH et al, eds. Idiosyncratic Reactions

to Valproate: Clinical Risk Patterns and Mechanisms of Toxicity. New York, NY: Raven Press; 1992:3.

Willmore LJ et al. Valproate toxicity: risk-screening strategies. J Child Neurol. 1991;6:3.

Kriel RL et al. Rectal diazepam gel for treatment of acute repetitive seizures. The North American Diastat

Study Group. Pediatr Neurol. 1999;20:282.

Cloyd JC et al. A single-blind, crossover comparison of the pharmacokinetics and cognitive effects of a new

diazepam rectal gel with intravenous diazepam. Epilepsia. 1998;39:520.

Haut SR. Seizure clusters: characteristics and treatment. Curr Opin Neurol. 2015;28:143–150.

Waruiru C, Appleton R. Febrile seizures: an update. Arch Dis Child. 2004;89:751.

Sadleir LG, Scheffer IE. Febrile seizures. Br Med J. 2007;334:307.

McIntyre J et al. Safety and efficacy of buccal midazolam versus rectal diazepam for emergency treatment of

seizures in children: a randomised controlled trial. Lancet. 2005;366:205.

Patterson JL et al. Febrile seizures. Pediatr Ann. 2013;42:249–254.

Delgado-Escueta AV, Janz D. Consensus guidelines: preconception counseling, management, and care of the

pregnant woman with epilepsy. Neurology. 1992;42(Suppl 5):149.

Fischbein CA, Berg IJ. Diazepam to prevent febrile seizures. N EnglJ Med. 1993;329:2033.

Newton RW. Randomised controlled trials of phenobarbitone and valproate in febrile convulsions. Arch Dis

Child. 1988;63:1189.

Farwell JR et al. Phenobarbital for febrile seizures: effects on intelligence and on seizure recurrence [published

correction appears in N EnglJ Med. 1992;326:144]. N EnglJ Med. 1990;322:364.

Sulzbacher S et al. Late cognitive effects of early treatment with phenobarbital. Clin Pediatr. 1999;38:387.

Offringa M, Newton R. Prophylactic drug management for febrile seizures in children. Evid Based Child Health.

2013;8:1376–1485.

Dreifuss FE, Langer DH. Hepatic considerations in the use of antiepileptic drugs. Epilepsia. 1987;28(Suppl

2):S23.

Smythe MA, Umstead GS. Phenytoin hepatotoxicity: a review of the literature. DICP. 1989;23:13.

Howard PA et al. Phenytoin hypersensitivity syndrome: a case report. DICP. 1991;25:929.

Blaszczyk B et al. Antiepileptic drugs and adverse skin reactions: an update. Pharmacol Rep. 2015;67:426.

Shear NH, Spielberg SP. Anticonvulsant hypersensitivity syndrome: in vitro assessment of risk. J Clin Invest.

1988;82:1826.

Pirmohamed M et al. Detection of an autoantibody directed against human liver microsomal protein in a patient

with carbamazepine hypersensitivity. Br J Clin Pharmacol. 1992;33:183.

EngelJN et al. Phenytoin hypersensitivity: a case of severe acute rhabdomyolysis. Am J Med. 1986;81:928.

Reents SB et al. Phenytoin-carbamazepine cross-sensitivity. DICP. 1989;23:235.

Ettinger AB et al. Use of ethotoin in phenytoin-related hypersensitivity reactions. J Epilepsy. 1993;6:29.

Arif H et al. Comparison and predictors of rash associated with 15 antiepileptic drugs. Neurology. 2007;68:1701.

Beran RG. Cross-reactive skin eruption with both carbamazepine and oxcarbazepine. Epilepsia. 1993;34:163.

Asconape JJ. Some common issues in the use of antiepileptic drugs. Semin Neurol. 2002;22:27.

Luef G. Female issues in epilepsy: a critical review. Epilepsy Behav. 2009;15:78–82.

Reimers A. New antiepileptic drugs and women. Seizure. 2014;23:585–591.

174.

175.

176.

177.

178.

179.

180.

181.

182.

183.

184.

185.

186.

187.

188.

189.

190.

191.

192.

193.

194.

195.

196.

197.

198.

199.

200.

McGrath A et al. Pregnancy-related knowledge and information needs of women with epilepsy: a systematic

review. Epilepsy Behav. 2014;31:246–255.

Kluger BM, Meador KJ. Teratogenicity of antiepileptic medications. Semin Neurol. 2008;28:328.

Yerby MS et al. Antiepileptics and the development of congenital anomalies. Neurology. 1992;42(Suppl 5):132.

Mattson RH et al. Use of oral contraceptives by women with epilepsy. JAMA. 1986;256:238.

Back DJ et al. Evaluation of Committee on Safety of Medicines yellow card reports on oral contraceptive-drug

interactions with anticonvulsants and antibiotics. Br J Clin Pharmacol. 1988;25:527.

Johnston CA, Crawford PM. Anti-epileptic drugs and hormonal treatments. Curr Treat Options Neurol.

2014;16:288.

Rosenfeld WE et al. Effect of topiramate on the pharmacokinetics of an oral contraceptive containing

norethindrone and ethinyl estradiol in patients with epilepsy. Epilepsia. 1997;38:317.

Doose DR. Oral contraceptive-AED interactions: no effect of topiramate as monotherapy at clinically effective

dosages of 200 mg or less. Epilepsia. 2002;43(Suppl 7):205.

Krauss GL et al. Antiepileptic medication and oral contraceptive interactions: a national survey of neurologists

and obstetricians. Neurology. 1996;46:1534.

Morrell MJ. Catamenial epilepsy and issues of fertility, sexuality, and reproduction. In: Wyllie E et al, eds. The

Treatment of Epilepsy: Principles and Practice. 3rd ed. Philadelphia, PA: Lippincott Williams & Wilkins;

2001:671.

O’Brien MD, Guillebaud J. Contraception for women with epilepsy. Epilepsia. 2006;47:1419.

Christensen J et al. Oral contraceptives induce lamotrigine metabolism: evidence from a double-blind, placebocontrolled trial. Epilepsia. 2007;48:484.

Vajda FJ et al. The teratogenicity of the newer antiepileptic drugs—an update. Acta Neurol Scand.

2014;130:234–238.

Harden CL et al. Management issues for women with epilepsy—focus on pregnancy (anevidence–

basedreview): I. Obstetrical complications and change in seizure frequency: report of the Quality Standards

Subcommittee and Therapeutics and Technology Assessment Subcommittee of the American Academy of

Neurology and the American Epilepsy Society. Epilepsia. 2009;50:1229.

Harden CL et al. Management issues for women with epilepsy—focus on pregnancy (an evidence-based

review): teratogenesis and perinatal outcomes: report of the Quality Standards Subcommittee and Therapeutics

and Technology Subcommittee of the American Academy of Neurology and the American Epilepsy Society.

Epilepsia. 2009;50:1237.

Harden CL et al. Management issues for women with epilepsy—focus on pregnancy (an evidence-based

review): Vitamin K, folic acid, blood levels, and breast-feeding: report of the Quality Standards Subcommittee

and Therapeutics and Technology Assessment Subcommittee of the American Academy of Neurology and the

American Epilepsy Society. Epilepsia. 2009;50:1247.

Kaneko S et al. Teratogenicity of antiepileptic drugs: analysis of possible risk factors. Epilepsia. 1988;29:459.

Meador KJ et al. NEAD Study Group. In utero antiepileptic drug exposure: fetal death and malformations.

Neurology. 2006;67:407.

Meador KJ, Loring DW. Developmental effects of antiepileptic drugs and the need for improved regulations.

Neurology. 2016;86(3):297–306.

Meador KJ et al. Fetal antiepileptic drug exposure and cognitive outcomes at age 6 years (NEAD study): a

prospective observationalstudy. Lancet Neurol. 2013;12:244–252.

Tomson T et al. Valproate in the treatment of epilepsy in girls and women of childbearing potential. Epilepsia.

2015;56(7):1006–1019.

Chen SS et al. Serum protein binding and free concentration of phenytoin and phenobarbitone in pregnancy. Br J

Clin Pharmacol. 1982;13:547.

Perucca E, Crema A. Plasma protein binding of drugs in pregnancy. Clin Pharmacokinet. 1982;7:336.

Patel IH et al. Valproic acid binding to human serum albumin and determination of free fraction in the presence

of anticonvulsants and free fatty acids. Epilepsia. 1979;20:85.

Pennell PB et al. Lamotrigine in pregnancy: clearance, therapeutic drug monitoring, and seizure frequency.

Neurology. 2008;70(22, pt 2):2130.

Tomson T, Battino D. Pharmacokinetics and therapeutic drug monitoring of newer antiepileptic drugs during

pregnancy and the puerperium. Clin Pharmacokinet. 2007;46:209.

Pack AM. Therapy insight: clinical management of pregnant women with epilepsy. Nat Clin Pract Neurol.

2006;2:190.

201.

202.

203.

204.

205.

206.

207.

208.

209.

210.

211.

212.

213.

214.

215.

216.

217.

218.

219.

220.

221.

222.

223.

224.

225.

[No authors listed]. American Academy of Pediatrics Committee on Drugs: the transfer of drugs and other

chemicals into human milk. Pediatrics. 1994;93:137.

Begg EJ et al. Prospective evaluation of a model for the prediction of milk:plasma drug concentrations from

physiochemical characteristics. Br J Clin Pharmacol. 1992;33:501.

Notarianni LJ et al. An in vitro technique for the rapid determination of drug entry into breast milk. Br J Clin

Pharmacol. 1995;40:333.

Veiby G et al. Epilepsy and recommendations for breastfeeding. Seizure. 2015;28:57–65.

Lowenstein DH, Alldredge BK. Status epilepticus. N EnglJ Med. 1998;338:970.

Wasterlain CG et al. Pathophysiologic mechanisms of brain damage from status epilepticus. Epilepsia.

1993;34(Suppl 1):S37.

Lothman E. The biochemical basis and pathophysiology of status epilepticus. Neurology. 1990;40(Suppl 2):13.

Alldredge BK et al. Treatment of status epilepticus. In: EngelJ et al, eds. Epilepsy: A Comprehensive Textbook.

2nd ed. Philadelphia, PA: Wolters Kluwer; 2008:1357.

Betjemann JP, Lowenstein DH. Status epilepticus in adults. Lancet Neurol. 2015;14:615.

Misra UK et al. Levetiracetam versus lorazepam in status epilepticus: a randomized, open labeled pilot study. J

Neurol. 2012;259:645.

Albers JM et al. Intravenous lacosamide: an effective add-on treatment of refractory status epilepticus. Seizure.

2011;20:428.

Kellinghaus C et al. Intravenous lacosamide as successful treatment for nonconvulsive status epilepticus after

failure of first line therapy. Epilepsy Behav. 2009;14:429.

Treiman DM et al. A comparison of four treatments for generalized convulsive status epilepticus. Veterans

Affairs Status Epilepticus Cooperative Study Group. N EnglJ Med. 1998;339:792.

Leppik IE et al. Double-blind study of lorazepam and diazepam in status epilepticus. JAMA. 1983;249:1452.

Rey E et al. Pharmacokinetic optimization of benzodiazepine therapy for acute seizures. Focus on delivery

routes. Clin Pharmacokinet. 1999;36:409.

Levy RJ, Krall RL. Treatment of status epilepticus with lorazepam. Arch Neurol. 1984;41:605.

Lacey DJ et al. Lorazepam therapy of status epilepticus in children and adolescents. J Pediatr. 1986;108:771.

Silbergleit R et al. Intramuscular versus intravenous therapy for prehospital status epilepticus. N Engl J Med.

2012;366:591.

Glauser T et al. Evidence-based guideline: treatment of convulsive status epilepticus in children and adults: report

of the Guideline Committee of the American Epilepsy Society. Epilepsy Curr. 2016;16:48–61.

Adams B et al. Fosphenytoin may cause hemodynamically unstable bradydysrhythmias. J Emerg Med.

2006;30:75.

Yaffe K, Lowenstein DH. Prognostic factors of pentobarbital therapy for refractory generalized status

epilepticus. Neurology. 1993;43:895.

Koul RL et al. Continuous midazolam infusion as treatment of status epilepticus. Arch Dis Child. 1997;76:445.

Denzel D, Burstein AH. Midazolam in refractory status epilepticus. Ann Pharmacother. 1996;30:1481.

Parent JM, Lowenstein DH. Treatment of refractory generalized status epilepticus with continuous infusion of

midazolam. Neurology. 1994;44:1837.

Claassen J et al. Treatment of refractory status epilepticus with pentobarbital, propofol, or midazolam: a

systematic review. Epilepsia. 2002;43:146.

p. 1300

Ischemic and hemorrhagic stroke are diseases involving the vascular

system of the brain. In the United States, approximately 87% of strokes

are ischemic in nature and 13% are hemorrhagic.

Case 61-1 (Question 1)

Ischemic and hemorrhagic stroke are medical emergencies, requiring

prompt medical attention at the first sign of symptoms. Signs and

symptoms of cerebrovascular disease usually occur acutely and vary

depending on the area of the brain involved. Ischemic and hemorrhagic

events have similar symptoms and must be distinguished before initiating

treatment.

Case 61-2 (Question 1)

Primary prevention is vital to reducing the risk of a stroke. Lifestyle

modifications and control of risk factors are the mainstay of primary

prevention. Important modifiable risk factors include cardiovascular

disease, hypertension, obesity, dyslipidemia, diabetes, smoking and

physical inactivity. The use of antiplatelet agents is recommended for

cardiovascular (including but not specific to stroke) prophylaxis and

reasonable for people whose 10-year risk is >10% as estimated by

cardiovascular risk calculators (see Core Principle 4).

Case 61-1 (Questions 1, 2)

Patients with atrial fibrillation and patent foramen ovale require primary

prevention pharmacotherapy according to their ischemic stroke risk.

Antiplatelet agents or anticoagulants should be used in patients with

these conditions, with selection of an agent dependent on patient

characteristics.

Case 61-1 (Question 2)

Secondary prevention of ischemic stroke and transient ischemic attacks

involves the use of antiplatelet agents. Selection of an agent is

dependent on patient characteristics.

Case 61-2 (Questions 5,6, 7,

8,

9, 10)

Acute treatment of ischemic strokes includes the use of alteplase given

intravenously. Alteplase should be started after confirming an event is

ischemic and not hemorrhagic. The treatment window for use of

alteplase is limited to 4.5 hours after the onset of neurologic symptoms.

Strict criteria for administration of alteplase must be followed to reduce

the risk of intracranial hemorrhage (ICH) and hemorrhagic

complications should be carefully monitored.

Case 61-2 (Questions 2, 3, 4,

5)

The strongest risk factor for hemorrhagic stroke because of non- Case 61-3 (Question 1)

traumatic intracerebral hemorrhage is uncontrolled hypertension.

Intracerebral hemorrhage may also occur as a result of an anatomic

abnormality in the brain or a disease process, such as a brain tumor.

Bleeding disorders, including those induced by anticoagulant drugs, also

predispose patients to intracerebral hemorrhage.

Acute treatment of intracerebral hemorrhage focuses on minimizing

hemorrhage expansion through careful blood pressure control and

reversal of coagulopathies, when appropriate, as well as prevention and

management of elevated intracranial pressure.

Case 61-3 (Questions 2-6)

Modifiable risk factors for hemorrhagic strokes include blood pressure

control, smoking cessation, and avoiding excessive alcohol and cocaine

use.

Case 61-3 (Question 7)

p. 1301

p. 1302

Rehabilitation after a cerebrovascular event is essential to patient

recovery. Common complications encountered in rehabilitation included

spasticity, depression, and neurogenic bowel or bladder.

Pharmacotherapy interventions should be directed at each of these

complications with the goal of improving the patient’s quality of life and

ability to function independently.

Case 61-5 (Question 1)

ISCHEMIC STROKE, HEMORRHAGIC STROKE,

AND TRANSIENT ISCHEMIC ATTACKS

Ischemic stroke, hemorrhagic stroke, and transient ischemic attacks result from either

inadequate blood flow to the brain (i.e., cerebral ischemia) with subsequent

infarction of the involved portion of the central nervous system (CNS) or

hemorrhages into the parenchyma or surrounding structures of the CNS and

subsequent neurologic dysfunction. This group of disorders is the fourth leading

cause of deaths among adults in the United States.

1

Comments

Search This Blog

Archive

Show more

Popular posts from this blog

TRIPASS XR تري باس

CELEPHI 200 MG, Gélule

ZENOXIA 15 MG, Comprimé

VOXCIB 200 MG, Gélule

Kana Brax Laberax

فومي كايند

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

NIFLURIL 700 MG, Suppositoire adulte

Antifongiques مضادات الفطريات

Popular posts from this blog

علاقة البيبي بالفراولة بالالفا فيتو بروتين

التغيرات الخمس التي تحدث للجسم عند المشي

إحصائيات سنة 2020 | تعداد سكَان دول إفريقيا تنازليا :

ما هو الليمونير للأسنان ؟

ACUPAN 20 MG, Solution injectable

CELEPHI 200 MG, Gélule

الام الظهر

VOXCIB 200 MG, Gélule

ميبستان

Popular posts from this blog

TRIPASS XR تري باس

CELEPHI 200 MG, Gélule

Popular posts from this blog

TRIPASS XR تري باس

CELEPHI 200 MG, Gélule

ZENOXIA 15 MG, Comprimé

VOXCIB 200 MG, Gélule

Kana Brax Laberax

فومي كايند

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

NIFLURIL 700 MG, Suppositoire adulte

Antifongiques مضادات الفطريات

Popular posts from this blog

Kana Brax Laberax

TRIPASS XR تري باس

PARANTAL 100 MG, Suppositoire بارانتال 100 مجم تحاميل

الكبد الدهني Fatty Liver

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

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

نمـو الدمـاغ والتطـور العقـلي لـدى الطفـل

CELEPHI 200 MG, Gélule

أخطر أنواع المخدرات فى العالم و الشرق الاوسط

Archive

Show more