+ All Categories
Home > Documents > Epilepsi at Web

Epilepsi at Web

Date post: 05-Apr-2018
Category:
Upload: akylah-azzahra
View: 218 times
Download: 0 times
Share this document with a friend

of 59

Transcript
  • 8/2/2019 Epilepsi at Web

    1/59

    James Fischer, Pharm.D.

    Spring 1998

    Drug Therapy of Epilepsy

    I.Epilepsy - Pathophysiology

    Definitions / Epidemiology

    Functional Anatomy of the Brain

    Pathophysiology

    Etiology

    Diagnosis

    Classification of Epileptic Seizures

    Selected Epileptic Syndromes

    First Aid for Seizures

    I.Drug Therapy

    Goal of Therapy

    General Management of Epilepsy

    Principles of Antiepileptic Drug (AED) Therapy

    Adverse Effects

    Drug Interactions

    Specific Drug Therapy for Epilepsy

    I.Case Study

    GOALS AND OBJECTIVES

    http://www.uic.edu/classes/pmpr/pmpr652/Final/Fischer/epilepsy.html#EPILEPSY%20-%20PATHOPHYSIOLOGYhttp://www.uic.edu/classes/pmpr/pmpr652/Final/Fischer/epilepsy.html#Definitions%20/%20Epidemiologyhttp://www.uic.edu/classes/pmpr/pmpr652/Final/Fischer/epilepsy.html#Functional%20Anatomy%20of%20the%20Brainhttp://www.uic.edu/classes/pmpr/pmpr652/Final/Fischer/epilepsy.html#Pathophysiologyhttp://www.uic.edu/classes/pmpr/pmpr652/Final/Fischer/epilepsy.html#Etiologyhttp://www.uic.edu/classes/pmpr/pmpr652/Final/Fischer/epilepsy.html#Diagnosishttp://www.uic.edu/classes/pmpr/pmpr652/Final/Fischer/epilepsy.html#CLASSIFICATION%20OF%20EPILEPTIC%20SEIZUREShttp://www.uic.edu/classes/pmpr/pmpr652/Final/Fischer/epilepsy.html#Selected%20Epileptic%20Syndromeshttp://www.uic.edu/classes/pmpr/pmpr652/Final/Fischer/epilepsy.html#FIRST%20AID%20FOR%20SEIZUREShttp://www.uic.edu/classes/pmpr/pmpr652/Final/Fischer/epilepsy.html#DRUGhttp://www.uic.edu/classes/pmpr/pmpr652/Final/Fischer/epilepsy.html#GOALhttp://www.uic.edu/classes/pmpr/pmpr652/Final/Fischer/epilepsy.html#MANAGEMENThttp://www.uic.edu/classes/pmpr/pmpr652/Final/Fischer/epilepsy.html#PRINCIPLEShttp://www.uic.edu/classes/pmpr/pmpr652/Final/Fischer/epilepsy.html#ADVERSEhttp://www.uic.edu/classes/pmpr/pmpr652/Final/Fischer/epilepsy.html#INTERACTIONShttp://www.uic.edu/classes/pmpr/pmpr652/Final/Fischer/epilepsy.html#SPECIFIChttp://www.uic.edu/classes/pmpr/pmpr652/Final/Fischer/epilepsy.html#CASEhttp://www.uic.edu/classes/pmpr/pmpr652/Final/Fischer/epilepsy.html#Definitions%20/%20Epidemiologyhttp://www.uic.edu/classes/pmpr/pmpr652/Final/Fischer/epilepsy.html#Functional%20Anatomy%20of%20the%20Brainhttp://www.uic.edu/classes/pmpr/pmpr652/Final/Fischer/epilepsy.html#Pathophysiologyhttp://www.uic.edu/classes/pmpr/pmpr652/Final/Fischer/epilepsy.html#Etiologyhttp://www.uic.edu/classes/pmpr/pmpr652/Final/Fischer/epilepsy.html#Diagnosishttp://www.uic.edu/classes/pmpr/pmpr652/Final/Fischer/epilepsy.html#CLASSIFICATION%20OF%20EPILEPTIC%20SEIZUREShttp://www.uic.edu/classes/pmpr/pmpr652/Final/Fischer/epilepsy.html#Selected%20Epileptic%20Syndromeshttp://www.uic.edu/classes/pmpr/pmpr652/Final/Fischer/epilepsy.html#FIRST%20AID%20FOR%20SEIZUREShttp://www.uic.edu/classes/pmpr/pmpr652/Final/Fischer/epilepsy.html#DRUGhttp://www.uic.edu/classes/pmpr/pmpr652/Final/Fischer/epilepsy.html#GOALhttp://www.uic.edu/classes/pmpr/pmpr652/Final/Fischer/epilepsy.html#MANAGEMENThttp://www.uic.edu/classes/pmpr/pmpr652/Final/Fischer/epilepsy.html#PRINCIPLEShttp://www.uic.edu/classes/pmpr/pmpr652/Final/Fischer/epilepsy.html#ADVERSEhttp://www.uic.edu/classes/pmpr/pmpr652/Final/Fischer/epilepsy.html#INTERACTIONShttp://www.uic.edu/classes/pmpr/pmpr652/Final/Fischer/epilepsy.html#SPECIFIChttp://www.uic.edu/classes/pmpr/pmpr652/Final/Fischer/epilepsy.html#CASEhttp://www.uic.edu/classes/pmpr/pmpr652/Final/Fischer/epilepsy.html#EPILEPSY%20-%20PATHOPHYSIOLOGY
  • 8/2/2019 Epilepsi at Web

    2/59

    1. Review the pathophysiology of epilepsy including etiology, diagnostic criteria,

    factors to consider in differentiating epilepsy from other disorders, classification of

    epileptic seizures and first aid measures for the patient with seizures.

    2. Identify the clinical manifestations and EEG findings associated with the different

    types of epileptic seizures. At the conclusion of these lectures, the student should be

    able to classify a given patient's seizure type based on data provided concerning the

    clinical description and EEG results.

    3. Provide an understanding of the basic principles involved in the drug treatment of

    epilepsy, including factors to consider in the initiation and assessment of antiepileptic

    drug (AED) therapy.

    4. The student should be able to recommend an initial AED regimen or alteration in

    regimen (including dose schedule, monitoring parameters) for an epileptic patient

    based on information concerning patient's seizure type, medical history, previous

    drug therapy and pertinent laboratory data.

    5. Explain the role of plasma concentration monitoring in AED therapy and be aware

    of the therapeutic serum concentration range for the major AED.

    6. Review the adverse effects seen during therapy with the major AED. The student

    should be aware of the causative factors, clinical importance, prevention and/or

    management of these adverse effects.

    7. The student should be aware of the potential drug interactions that may occur

    among the AED, and the mechanisms and clinical implications of these interactions.

    REQUIRED READINGS

    1. Fischer JH (ed). The Epilepsy Counseling Guide. MPE Communications inc., Fair

    Lawn, New Jersey, 1994.

    2.Garnett WR. Epilepsy, inPharmacotherapy: A Pathophysiologic Approach, 3rd ed.

    Dipiro JT, Talbert RL, Yee GC, Matzke GR, Wells BG, Posey LM, eds. Appleton &

    Lange, Stamford, CT. 1997; 1179-1209.

    EPILEPSY - PATHOPHYSIOLOGY

    I. Definitions/ EpidemiologyA. Seizures are discrete, time-limited alterations in brain function - including changes

    in motor activity, autonomic function, consciousness, or sensation -that result from an

    abnormal and excessive electrical discharge of a group of neurons within the brain.

    The clinical manifestations of a seizure reflect the area of the brain from which the

    seizure begins (i.e., seizure focus) and the spread of the electrical discharge. Clinical

    manifestations accompanying a seizure are numerous and varied, including

    indescribable bodily sensations, "pins and needles" sensations, smells or sounds, fear

  • 8/2/2019 Epilepsi at Web

    3/59

    or depression, hallucinations, momentary jerks or head nods, staring with loss of

    awareness, and convulsive movements (i.e., involuntary muscle contractions) lasting

    seconds to minutes. Convulsions are specific type of seizures where the attack is

    primarily manifested by involuntary muscular contractions.

    B. Seizures are symptoms of a disturbance in brain function. Up to 10% of the

    population will experience a seizure during their lifetime. However, in the majority of

    these people, seizures occur either as an isolated incident or secondary to an acute

    reversible medical condition, such as fever, trauma, metabolic disorder, or alcohol or

    drug intoxication. Since seizures do not usually recur in these patients once the

    underlying cause has been corrected, these patients are not considered to have

    epilepsy and do not require long-term antiepileptic drug therapy.

    C. Epilepsy is defined as a condition characterized by recurrent (two or more)

    seizures unprovoked by any immediately identifiable cause.

    D. Epilepsy is the third most common neurologic disorder, following stroke and

    Alzheimer=s disease. Approximately 2 million people (0.5% - 1.5% of population) inthe United States have active epilepsy. Each year 50 per 100,000 individuals in the

    United States will be diagnosed with epilepsy (125,000 new cases/year), with the

    highest frequency of newly identified cases occurring among children < 5 years (50-

    100 cases/100,000) and adults >65 years of age (70-150 cases/100,000).

    II. Functional Anatomy of the Brain

    The material in sectionII. is informational only (i.e., it will not be included on test)

    and is included to provide a brief overview of the neuroanatomy pertaining to

    seizures and epilepsy. The upper and largest portion of the brain is referred to as the

    cerebrum. The cerebrum is comprised of grey and white matter. The outer layer of

    grey matter forms the cerebral cortex and consists largely of nerve cells (neurons) andsupportive glial cells. White matter, comprised of myelinated nerve fibers, lies

    beneath the cerebral cortex. These myelinated fibers connect the cerebrum with other

    parts of the brain (projection fibers), the front of the brain to the back portion,

    different areas on the same side of the cerebrum (association fibers), and one side of

    the brain to the other (commissural fibers). As shown in the figure above, the

    cerebrum is incompletely divided into the left and right hemispheres by the medial

    longitudinal fissure. The left and right cerebral hemispheres are interconnected by a

    large fiber bundle located beneath the medial fissure, the corpus callosum. The right

    hemisphere controls the left side of the body and "sees" the left half of space; the left

    hemisphere controls the right side of the body and "sees" the right half of space. In

    most right-handed individuals, the left hemisphere controls language functions suchas the ability to speak (frontal lobe) and understand language (temporal lobe). As

    shown below, each cerebral hemisphere is divided into 5 lobes: frontal, parietal,

    temporal, occipital, and the insula (located on the underside of brain). Specific areas

    in each lobe are associated with different functions. Injury or abnormal functioning of

    these cortical areas can cause partial seizures, with the initial symptoms of the seizure

    often reflecting the function of that area.

  • 8/2/2019 Epilepsi at Web

    4/59

    The neocortex (cortical area covering surface of brain), hippocampus, and other

    mesial temporal frontal areas are frequent sites of seizure onset. Subcortical areas,

    such as the thalamus, substantia nigra, and corpus striatum, are thought to play key

    roles in the spread of seizure activity and generation of generalized seizures. In the

    "normal" brain, inhibitory stimuli from these subcortical areas modulate excitatory

    neurotransmission between the cortex and other brain areas and limit the spread ofabnormal electrical signals. Depression of the inhibitory activity of these areas in the

    brains of patients with epilepsy may facilitate the spread of seizure activity following

    an initial partial seizure or the generation of primary generalized seizures.

    III. Pathophysiology

    The onset of a seizures appears to occur when a small group of abnormal neurons

    undergo prolonged depolarizations associated with the rapid firing of repeated action

    potentials. These abnormally discharging epileptic neurons recruit adjacent neurons

    or neurons with which they are connected into the process. A clinical seizure occurs

    when the electrical discharges of a large number of cells become abnormally linked

    together, creating a storm of electrical activity in the brain. Seizures may then spread

    to involve adjacent areas of the brain or through established anatomic pathways to

    other distant areas.

    On a fundamental level, seizures can be viewed as resulting from an imbalance

    between excitatory and inhibitory processes in the brain. Proposed mechanisms for

    the generation and spread of seizure activity within the brain include abnormalities in

    the membrane properties of neurons, changes in the ionic micro environment

    surrounding the neuron, decreased inhibitory neurotransmission which is primarily

    by gamma-amino butyric acid (GABA), or enhanced excitatory neurotransmissionwhich is primarily mediated by the acidic amino acid, glutamate. The different

    antiepileptic drugs (AEDs) act by affecting one or more of these processes. Specific

    mechanisms of action of the AEDs include: modulation of voltage dependent ion

    channels (carbamazepine, phenytoin, valproic acid), enhancement of activity of the

    major inhibitory neurotransmitter in the brain, GABA (phenobarbital,

    benzodiazepines, tiagabine), and suppression of excitatory neurotransmission

    (lamotrigine, felbamate).

    IV. Etiology

    A. In 60-70% of patients, no specific cause for their seizures can be identified.Epilepsy in these patients is referred to as being idiopathic (i.e., no definite cause).

    B. Infants/children: congenital malformations, perinatal injuries or hypoxia,

    developmental neurologic disorders, metabolic defects, injury, and infection are

    common causes of seizures.

    C. Young Adults: head trauma, brain tumors, infection, and arteriovenous

    malformations are common causes of seizures.

  • 8/2/2019 Epilepsi at Web

    5/59

    D. Elderly: cerebrovascular disease, CNS degenerative diseases, and brain tumors are

    common causes.

    E. Genetic - risk increased 2-3 times in individuals with first degree relative with

    epilepsy.

    V. Diagnosis

    A. Steps in Diagnosis of Epilepsy

    1. Confirm patient has epilepsy

    a. Other conditions to consider in differential: pseudoseizures, syncope, breath

    holding spells, narcolepsy, hemiplegic migraines, drug toxicity, transient ischemia

    attack.

    b. Following factors delineate epilepsy from above: abrupt onset, genuine loss of

    consciousness (if not simple partial), brief duration, rapid recovery, stereotypic

    episodes.

    c. Pseudoseizures (seizures occurring on a psychogenic basis, "hysterical seizures")

    present a common and difficult diagnostic problem, especially since many patients

    may have both pseudoseizures and epilepsy. Factors differentiating pseudoseizures

    from epilepsy include:

    1. Precipitating factors with a strong emotional orpsychological component

    2. "Non-physiological" seizure patterns - violent thrashing orflailing of all 4 limbs, particularly when movements

    asynchronous; preservation of consciousness despite motor

    activity of arms and legs; rage and directed violence as ictal

    events; gradual build up and prolonged resolution of

    seizure; lack of tongue biting, incontinence and postictal

    confusion.

    3. Personal and family history of psychiatric disease

    4. Repeatedly normal interictal EEG and lack of any responseto therapeutic levels of antiepileptic drugs

    5. Definitive differentiation requires use of simultaneous videoand EEG recording

    2. Correct classification of seizure type and, if possible, epileptic syndrome

    3. Identification of any underlying cause

    B. Diagnostic Evaluation

  • 8/2/2019 Epilepsi at Web

    6/59

    1. History -

    a. Medical history of patient and family history

    b. Description of seizure - important to obtain exact details of episode from patient

    and/or observer. Description should include details on:

    1. Events preceding seizure: What was happening before theseizure ? What time was it ? Does patient recognize onset of

    seizure by a smell, visual disturbance, sound or odd feeling ?

    2. Events during the seizure: What are the initial events ? Isconsciousness lost or altered ? What kind of body

    movements occurred ? How long did the seizure last ? Did

    the person urinate or bite his/her tongue ?

    3. Events after the seizure (i.e., postictal period): Is the patient

    alert, drowsy, or confused ? Was there any numbness orweakness ?

    4. An effort should also be made to identify any precipitating

    factors. Factors known to precipitate epilepsy in susceptible

    patients include: sleep deprivation, fever, emotional stress,

    lack of food, alcohol/drug withdrawal, pregnancy, menses,

    and various sensory stimuli (i.e., photosensitivity, television,

    reading, eating, music). Identification and avoidance, where

    possible, of these factors may assist in reducing the

    frequency of seizures.

    2. Physical and Neurological Examination

    3. Clinical Laboratory data

    4. Electroencephalography (EEG)

    a. Measurement of fluctuations in electrical activity within brain recorded from

    electrodes on scalp. Role: confirm presence of epilepsy, diagnosis of seizure type, long

    term prognosis.

    b. EEG findings alone do not confirm or deny diagnosis of epilepsy. Important to

    correlate EEG findings to clinical events.

    1. Approximately 5% of patients without epilepsy haveepileptiform discharges on their EEG.

    2. Of patients with epilepsy, only about 50% have epileptiformactivity on their first EEG.

    c. Detection of abnormal EEG enhanced by use of multiple recordings and specific

    activating techniques.

  • 8/2/2019 Epilepsi at Web

    7/59

    1. photic stimulation, hyperventilation, sleep deprivation (these

    are usually performed with standard EEG exam);

    2. Video-EEG monitoring: provides correlation betweenclinical seizure and electrical activity in brain

    3. Nasopharyngeal and sphenoidal electrodes: allows fordetection of abnormal electrical activity on underside of

    cortex

    d. EEG patterns having clinical correlations

    1. 3 Hz spike and wave complex: Absence seizures

    2. Slow spike and wave complex: minor motor seizures (i.e.,atypical absence, tonic, atonic)

    3. Hypsarrhythmia: infantile spasms4. Polyspike and wave complex: myoclonic seizures

    5. Neuroimaging Studies: Magnetic resonance imaging (MRI) or computed

    tomography are useful in identifying structural lesions in brain. MRI appears more

    sensitive in detecting lesions in patients with epilepsy. Consider in patients > age 18, in

    children with partial seizures, and in presence of abnormal neurologic findings, or

    focal slow-wave abnormalities on EEG.

    VI. CLASSIFICATION OF EPILEPTIC SEIZURES

    Seizures are classified according to their clinical features and patterns seen on theEEG. Epileptic seizures are divided into two broad categories based on the symptoms

    and EEG findings observed at the outset of the seizure. If the initial onset indicates

    involvement of both sides of the brain, the seizures are referred to as generalized

    seizures. If the initial onset indicates involvement of only a localized area of the brain,

    they are referred to as partial seizures.

    A. Partial Seizures - those seizures where initial onset arises from a localized area of

    brain. Partial seizures are caused by localized injury to brain and diagnostic

    evaluation for the presence of a focal lesion (i.e., tumor, vascular malformation,

    stroke, trauma, neurodegenerative disease) is required. However, in majority of

    patients, cause will remain unknown (idiopathic). Partial seizures are further

    subdivided based on whether consciousness is maintained (i.e., simple partial) or

    impaired (i.e., complex partial) during the seizure. Partial seizures are most common

    type experienced by adults.

    1. Simple partial seizures

    a. No loss of consciousness; During a simple partial seizure, the patient is alert and

    able to respond to questions or commands and afterwards remembers what happened

  • 8/2/2019 Epilepsi at Web

    8/59

    during the seizure. Simple partial seizures may precede complex partial or

    secondarily generalized seizures, in which case they are referred to as auras.

    b. Clinical manifestations of simple partial seizure usually relate to the particular

    area of brain involved, and thus a wide variety of symptoms are possible, including

    motor, sensory, autonomic, and psychic manifestations. For any given patient,

    symptoms will be same with each seizure.

    c.Motor seizures generally reflect involvement of the motor or supplementary motor

    cortex and cause a change in muscle activity. Tonic (neck stiffening, sustained

    deviation of eyes to one side) or clonic (jerking) movements are most common.

    Abnormal movements may be restricted to one body part or spread to other muscles

    on same side or both sides (secondary generalization) of the body.

    d.Sensory seizures are often manifested by hallucinations or illusions involving one of

    the senses - touch (paresthesia or numbness in one or more body parts), smell (patient

    may smell a disagreeable odor), taste (abnormal or disagreeable taste), vision

    (unformed or formed visual hallucinations), and hearing (buzzing sound, ringing inears, music, voices).

    e.Autonomic seizures can cause changes in heart or breathing rate, sweating,

    goosebumps, or strange or unpleasant sensation in abdomen, chest or head.

    f.Psychic seizures, arising from the limbic system and neocortical areas of the frontal

    and temporal lobes, affect how the patient thinks, feels, and experiences things.

    Manifestations of psychic seizures include feelings of fear, anxiety, depression, deja

    vu, jamais vu, and dissociative phenomena such as autoscopy (out of body

    experience).

    g. Duration 30 seconds or less; No postictal symptoms, although patient=s with partialmotor seizures may experience a temporary numbness or weakness in the affected

    body part (Todd's paralysis)

    h. EEG findings: local contralateral discharge starting over the cortical area

    corresponding to symptoms

    j. Prognosis: good seizure control obtained in 30-50%

    2. Complex partial seizures (temporal lobe, psychomotor epilepsy)

    a. Impairment of consciousness observed: In this context, consciousness refers to

    patients' ability to normally interact and respond to their environment. Thus,although patients may appear to be conscious, closer examination shows that they are

    unaware of their environment; fail to respond or respond inappropriately to

    questions; and afterward, are unable to remember the episode. Complex partial

    seizures involve portions of brain concerned with maintenance of consciousness and

    memory, and generally imply bilateral involvement of temporal or frontal lobes and

    limbic system.

  • 8/2/2019 Epilepsi at Web

    9/59

    b. Associated with initial aura (i.e., simple partial seizure) in >50% of patients; the

    aura is a simple partial seizure which may then progress to a complex partial (and/or

    generalized tonic-clonic) seizure. Most common forms of aura: fear, rising epigastric

    sensation, unilateral "funny feeling" or "numbness", or visual disturbances; focal

    twitching of face or fingers.

    c. Simple to complex automatisms (repetitive motor activity that is purposeless,

    undirected, and inappropriate) are frequently observed during complex partial

    seizures. Examples include repetitive chewing or swallowing, lip smacking, fumbling

    movements of fingers or hands, picking at clothing, mumbling, moving about

    aimlessly, purposeless behavior, and clumsy perseverance of a preceding motor act.

    d. Average duration 1 to 3 minutes

    e. Postictal phase - confusion, lethargy, altered behavior, amnesic for event

    f. EEG findings: unilateral or, frequently, bilateral discharge in temporal or

    frontotemporal region.

    g. Prognosis: good seizure control in 40-60%

    h. Most common seizure type seen in adult, account for up to 60% of adult epilepsies

    3. Partial Seizures Secondarily Generalized - partial seizure may progress through

    several stages reflecting spread of discharge to different brain areas. For example,

    seizure may begin as simple partial (i.e., aura), progress to complex partial and

    subsequently become secondarily generalized (tonic-clonic). The initial clinical events

    of a seizure, described by patient or observer, will usually provide a reliable

    indication of whether seizure is partial or generalized. Sometimes, however, the focal

    onset may not be apparent from clinical data, due to either rapid spread of dischargeor location of focus in brain area not associated with an obvious behavioral function,

    thus EEG findings are needed for accurate classification.

    B. Generalized Seizures - those seizures where first clinical changes indicate initial

    involvement of both hemispheres. The initial clinical event is a loss of consciousness.

    Various types of generalized seizures differentiated by absence or presence of specific

    motor activity.

    1. Generalized Tonic-Clonic (Grand Mal)

    a. Loss of consciousness is quickly followed by a sudden fall to ground. In the tonic

    phase, muscles become rigid and the simultaneous contractions of diaphragm andchest muscles may produce the characteristic "epileptic cry". The patient's eyes roll

    up or turn to the side and the tongue may be bitten. The rigidity is replaced shortly by

    series of synchronous clonic movements of head, face, legs and arms. Autonomic

    changes also observed included: increased blood pressure, heart rate, and bladder

    pressure; pupillary mydriasis; hypersecretion of skin and salivary glands; cyanosis of

    skin.

  • 8/2/2019 Epilepsi at Web

    10/59

    b. Although onset may occur at any age, most commonly occurs during second decade

    of life.

    c. Average duration 2 to 5 minutes.

    d. Postictally, patients lethargic/sleepy lasting several minutes to hours.

    e. Incontinence seen in early postictal phase in approx. 35% of patients.

    f. In patients with primary generalized tonic-clonic seizures, seizures seen most

    commonly on awakening and to a lesser extent in evening when relaxing.

    g. Prognosis: good seizure control in 70-85%.

    2. Absence (Petit Mal)

    a. Onset between 4 and 14 years and often resolve by age 18.

    b. Clinical description - Brief episodes of staring with impairment of awareness and

    responsive that begin without warning and end suddenly, leaving patient alert and

    attentive. In simple absence seizures, patient only stares. In the more common

    complex absence seizures, staring is accompanied by simple automatic movements

    such as blinking of eyes, drooping of head, or chewing.

    c. Duration - short (10-45 secs), patients usually unaware of occurrence.

    d. Abrupt recovery without after effects

    e. Characteristic EEG pattern of 3 per second spike and waves; may be precipitated

    in large percent of patients by hyperventilation.

    f. 25 to 50% of patients go on to develop generalized tonic-clonic (GTC) seizures.

    g. Development and intelligence are usually normal and long term prognosis is good,

    particularly in patients who do not develop GTC.

    h. Important in children to differentiate from complex partial seizures, since

    treatment and prognosis vary. In contrast to absence, complex partial seizures usually

    have a longer duration (minutes vs. seconds), are often preceded by aura, and

    typically have a brief period of postictal confusion. Also, the EEG pattern is markedly

    different between the two seizure types.

    3 Atypical Absence

    a. Onset between 1 to 7 years of age

    b. Clinical description - similar to typical absence except that loss of responsiveness

    during seizure is often less complete and more gradual in onset and cessation; Also

    clonic, tonic and atonic components (i.e., increase or decreases in muscle tone) are

    more pronounced than in typical absence. Commonly seen in patients with Lennox-

    Gaustaut syndrome in conjunction with myoclonic, atonic and tonic seizures.

  • 8/2/2019 Epilepsi at Web

    11/59

    c. EEG findings: slow spike and wave (< 2.5 Hz) discharge and/or incompletely

    generalized spike-waves

    d. Not precipitated by hyperventilation

    e. Often associated with mental retardation or structural CNS damage

    f. Prognosis: response to therapy and long term prognosis dependent on presence of

    underlying neurologic deficit and/or mental retardation. Good response seen only 20-

    30% of patients.

    4. Atonic seizures

    a. Onset usually between age of 2 to 5 years

    b. Clinical description- sudden and total loss of muscle tone and posture control that

    causes eyelids to drop, head to nod and patient to fall to ground - "drop attack"; not

    necessarily associated with loss of consciousness. Must wear helmet to protect from

    head injury. May or may not have postictal symptoms.

    c. Average duration 10 to 60 seconds; brief, if any, postictal symptoms

    d. Other seizure types common in patients with atonic seizures. May be observed in

    conjunction with myoclonic seizures and atypical absence (Lennox-Gaustaut

    Syndrome)

    e. Prognosis to therapy dependent on presence of underlying neurological deficit

    and/or mental retardation

    5. Myoclonic Seizure

    a. Sudden, brief shock-like jerk of a muscle or group of muscles, often occurs in

    healthy people as they fall asleep. Epileptic myoclonus usually causes synchronous

    and bilateral jerks of the neck, shoulders, upper arms, body, and upper legs.

    b. Myoclonic seizures occur in a variety of epilepsy syndromes.

    6. Tonic seizures

    a. Characterized by sudden bilateral stiffening of the body, arms, or legs. Tonic

    seizures usually last less than 20 seconds and are more common during sleep.

    b. Primarily seen in younger children; commonly associated with metabolic disorder

    or underlying neurological deficit

    c. Frequently occur with other seizure types and in various epilepsy syndromes

    d. Duration 10-60 seconds; brief, if any, postictal symptoms

    Note material on handout after this point will not be covered in class and you are not

    responsible for this information on the test.

  • 8/2/2019 Epilepsi at Web

    12/59

    VII. Selected Epileptic Syndromes

    Epilepsy syndromes are defined by a cluster of characteristics, including seizure type,

    EEG, neurologic status, age at seizure onset, family history, and prognosis. While

    seizure type is most important determinant of drug selection, classifying the epilepsy

    syndrome provides information on the responsive to drug therapy, expected duration

    of drug therapy, and long term prognosis.

    A. Infantile Spasms

    1. Consist of sudden flexion of the head with abduction and extension of arms,

    accompanied by flexion of knees and often a little grunt or cry. Spasms may also be

    extension rather than flexion. Spasms commonly occur in series of 2 or more.

    2. Onset most commonly between 4 to 7 months of age

    3. Definite etiology can be established in 60% of patients

    4. Mortality rate 11-23%; developmental retardation 80-90%

    5. Characterized by spasms, developmental retardation, hypsarrhythmia pattern on

    EEG.

    6. Spasms may be flexor (jackknife), extensor or mixed flexor-extensor.

    7. Unique among seizure types in responsiveness to ACTH/corticosteroids.

    8. Poor treatment prognosis; spasms usually disappear by age 3 or 4, but child left

    profoundly handicapped, retarded, and often with Lennox-Gaustaut syndrome (see

    under atypical absence seizures). Patients who are normal prior to onset and who

    respond to therapy have slightly better prognosis.

    B. Febrile Seizures

    1. Convulsions that occur with fever (> 38oC) in children between 6 months and 6

    years of age, not secondary to an infection of brain or meninges.

    2. Prevalence: 2 to 5% of all children will have a febrile seizure before 6 y/o; Peak

    incidence at 2 years of age.

    3. Etiology: strong genetic predisposition

    4. Primarily occur as generalized tonic-clonic seizures, but partial seizures occur in

    10-15% of patients.

    5. Prognosis: febrile convulsions usually have benign course; although 2 to 4% will go

    on to develop afebrile epilepsy. Intellectual dysfunction and neurologic sequelae may

    occur following febrile status epilepticus.

    6. Factors associated with increased risk of developing afebrile epilepsy: Preexisting

    neurologic abnormality; family history of afebrile seizures; and a complicated initial

  • 8/2/2019 Epilepsi at Web

    13/59

    seizure (> 20 minutes duration 2 or more seizures in same illness, and/or focal febrile

    seizure).

    7. Treatment

    1. Symptomatic - control fever and seizures

    2. Prophylactic Antiepileptic Drug Therapy - controversial;present recommendations to consider prophylactic

    treatment only in patients with two or more of risk factors

    above; or in patients with recurrent (3 or more) febrile

    seizures who are under 3 years of age. Current

    recommendation for prophylactic therapy is the intermittent

    administration of rectal or oral diazepam at time of febrile

    episode.

    C. Lennox-Gastaut Syndrome: This syndrome is characterized by the triad of

    intractable seizures, mental and developmental retardation, and slow spike and wavepattern on the EEG. Seizures (tonic, atonic, atypical absence, myoclonic, and tonic-

    clonic) usually begin between ages 1 and 6 years and respond poorly to antiepileptic

    drugs. Behavioral problems are common and probably result from the underlying

    neurologic injury, effects of frequent seizures and head injuries, and high-dose

    combinations of antiepileptic drugs.

    D. Benign Rolandic epilepsy. This syndrome frequently begins in children with a

    family history of epilepsy. The most characteristic sign is a partial motor or

    somatosensory seizure involving the face. Tonic-clonic seizures may also occur,

    especially during sleep. The seizures are infrequent (some patients require no

    medications), are easily controlled with antiepileptic drug therapy, and stop

    spontaneously by age 15. Mental development is unaffected.

    E. Juvenile myoclonic epilepsy: These myoclonic seizures, with or without tonic-clonic

    or absence seizures, usually begin shortly before or after puberty but may first occur

    in early adulthood. Myoclonic and tonic-clonic seizures most often occur in the early

    morning, shortly after the patient awakens. Mental developemnt is normal. In most

    patients seizures are well controlled by valproic acid alone, but the disorder requires

    life long therapy.

    VIII. FIRST AID FOR SEIZURES

    A. Generalized Tonic-Clonic Seizures

    1. Prevent person from hurting himself or herself. Place something soft under the

    head, loosen tight clothing, and clear area of sharp or hard objects.

    2. Do not force any objects into patient's mouth.

    3. Do not restrain patient's movements.

    4. Turn patient on his or her side to allow saliva to drain from mouth

  • 8/2/2019 Epilepsi at Web

    14/59

    5. Stay with patient until seizure ends naturally.

    6. Do not pour liquids into patient's mouth or offer any food, drink or medication

    until fully awake.

    7. Give artificial respiration if patient does not resume breathing after seizure.

    8. Provide area for patient to rest until fully awakened, accompanied by responsible

    adult.

    9. Be reassuring and supportive when consciousness returns.

    10. While a convulsive seizure is not usually a medical emergency, presence of any of

    the following signs indicate the need for immediate medical attention:

    a. Seizure lasting longer than 10 minutes or occurrence ofsecond seizure.

    b. Difficulty in rousing at 20-minute intervals.

    c. Complaints of difficulty with vision

    d. Vomiting

    e. Persistent headache after a rest period

    f. Unconsciousness with failure to respond

    g. Unequal size pupils or excessively dilated

    B. Nonconvulsive Seizures (Absence and Complex Partial)

    1. Do not restrain patient.

    2. Remove harmful objects from patient's path.

    3. Calmly try to encourage patient to sit down or encourage him or her away from

    dangerous situations. If person does not respond to these measures, force should not

    be used.

    4. Observe but do not approach patient who appears angry or combative.

    5. Remain with patient until fully alert.

    DRUG THERAPY

    I. GOAL OF THERAPY

    A. Primary goal of drug therapy is the complete suppression of seizures in the absence

  • 8/2/2019 Epilepsi at Web

    15/59

    of disabling side-effects. Prognosis of epilepsy has improved in last decade, and at

    present about 60-70% of newly diagnosed patients can be expected to achieve

    complete seizure control following institution of effective monotherapy (one drug).

    B. When epilepsy cannot be controlled completely, the aim of treatment is to attain

    the best compromise between the desire to maximize seizure control and the need to

    keep side-effects within acceptable limits for the individual patient.

    C. Therapy should maintain or restore the patient=s lifestyle and ability to lead an

    active life.

    II. GENERAL MANAGEMENT OF EPILEPSY

    A. Appropriate diagnostic evaluation

    B. Identify and correct underlying cause

    C. Treatment of Seizures

    1. Assess necessity of drug therapy

    a. Drug therapy not indicated for seizures due to acutereversible medical problem

    b. Therapy not necessary for certain benign epilepsies (febrileseizures, rolandic epilepsy)

    c. Following first unprovoked seizure- while some benefit may

    occur by initiating therapy in high risk patients, present

    consensus is to delay therapy until patient experiences a

    second unprovoked seizure.

    2. Institution of appropriate antiepileptic drug therapy

    3. Identify and avoid if possible any precipitating factors (i.e., alcohol, lack of sleep,

    emotional stress, fever, lack of food, exposure to flickering light, menstruation)

    4. Evaluation for surgery or implantation of vagal nerve stimulator in patients

    refractory to drug therapy.

    D. Prevention of complications of seizures

    1. Early control/termination of seizures

    2. Avoidance of intolerable drug-induced adverse effects

    3. Attention to and treatment of psychosocial complications

    III. PRINCIPLES OF ANTIEPILEPTIC DRUG (AED) THERAPY

    A. Select most appropriate drug based on:

  • 8/2/2019 Epilepsi at Web

    16/59

    1. Seizure Type (see Table 1) - Seizure type is the primary criteria for antiepileptic

    drug selection. Although not necessary for drug selection, knowledge of the epilepsy

    syndrome is also useful since it provides additional information including the

    expected responsiveness of the patient=s seizure disorder to drug therapy, expected

    duration of drug therapy, and long term prognosis.

    2. Final selection among drugs having equal efficacy for a given seizure type should be

    individualized for each patient based on other factors such as potential adverse

    effects, convenience of administration, cost, and patient=s lifestyle. (see Individual

    drug monographs, Section VI).

    B. Optimization of therapy requires individualization of dosage. Once an agent has

    been selected, therapy should initiated by starting at the low end of the drug=s

    recommended dosage range and slowly increasing the dose until seizures are

    controlled or intolerable adverse effects develop. Following each dose increase, time

    should be allowed for the drug to come to steady-state before evaluating the patient=s

    clinical response at that dosage level and deciding whether further adjustments are

    needed.

    1. Initial Dosage: AED therapy should be gradually introduced during the first month

    of therapy to minimize the gastrointestinal and neurologic side effects that commonly

    occur with initiation of AED treatment. The frequency of these side effects tends to

    decrease over the first few months as tolerance develops. Although seen to some

    extent with all the AEDs, these initiation related side effects may be especially

    troublesome with carbamazepine, ethosuximide, felbamate, lamotrigine, primidone,

    tiagabine, topiramate and valproic acid. In addition to the GI and CNS effects listed

    above, the occurrence of rash with lamotrigine is related to the rate of dose initiation.

    To minimize these initiation related side effects, these agents are usually started at

    subtherapeutic dosages and the dose gradually increased over several weeks to therecommended dose range. Specific initiation schedules for these drugs are provided in

    their monographs (see section VI). If intolerable adverse effects develop during the

    titration process, the dose should be reduced to the previous level that the patient

    tolerated and, after symptoms subside, the titration process restarted using smaller

    dosage increments. Since initiation related adverse effects are less problematic with

    gabapentin, phenytoin, and phenobarbital, therapy with these agents is generally

    started at dosages within the recommended dose range.

    2. Before considering therapy with a given AED as a failure and switching to another

    drug, the following factors should be reevaluated:

    a. Diagnosis of epilepsy

    b. Classification of seizure type and/or epilepsy syndrome

    c. Presence of an active lesion

    d. Adequate dosage and/or duration of therapy (i.e., was dosepushed to maximal tolerated level, was adequate time

  • 8/2/2019 Epilepsi at Web

    17/59

    allowed for steady-state to be achieved following each dose

    adjustment).

    e. Compliance with medication regimen (noncompliance ismost common cause of drug failure and breakthrough

    seizures)

    3. If seizures continue, despite a maximally tolerated dose of the first AED, a second

    AED should be selected. Therapy with the second drug should be initiated as

    described above. Once the dose of the second drug has been titrated into its

    recommended dose range, the initial drug should then be gradually withdrawn over

    1-3 weeks (more rapid if being performed as inpatient). After the first drug has been

    withdrawn, the dose of the second drug, taken alone (i.e., monotherapy), should then

    be increased until seizures are controlled or intolerable side effects develop. This

    process should be continued until monotherapy with two or three of the primary

    drugs has failed. Only after this should combination or polytherapy be considered.

    Factors to consider when combining drugs include the patient=s previous clinical

    response (i.e., seizure control, side effects) to each drug alone, mechanism of action(theoretically combining drugs having differing mechanisms of action would appear

    preferable), adverse effect profiles, and pharmacokinetic properties.

    4. Epilepsy surgery should also be considered in patients who have failed

    monotherapy with the primary drugs and an initial attempt with polytherapy.

    C. Monotherapy

    Monotherapy with the AEDs is preferred for most patients. Advantages of

    monotherapy as compared to polytherapy (multiple drug therapy) include: equal or

    superior efficacy to combination drug therapy; reduced frequency of adverse effects;

    absence of drug interactions; lower cost; enhanced ability to correlate response,adverse effects, and abnormal lab values to specific drug; reduced risk of birth

    defects; and improved compliance due to simpler and less intrusive regimens

    D. Monitoring Therapy

    1. Seizures - patients should be encouraged to keep a seizure diary and accurately

    record the type, duration, and time of occurrence of any seizures. In assessing

    therapy, the following seizure related parameters should be considered:

    a. Change in seizure frequency - important to assess not onlychange in absolute number of seizures, but also length of

    seizure-free interval

    b. Change in seizure pattern or type

    c. Altered time of occurrence

    2. Adverse Effects: patients should be educated to the type of adverse effects that may

    occur with the AEDs and to record in their seizure diary any adverse effects and the

  • 8/2/2019 Epilepsi at Web

    18/59

    time of day at which they occur. Specific adverse effects that should be monitored for

    include:

    a. Dose-Dependent Neurological Effects - (see section V, VI) thefollowing should be assessed at each visit: mental status;

    cerebellar signs (i.e., nystagmus, balance, coordination,

    tremor); visual problems (blurring, double vision); cognitive

    function

    b. Idiosyncratic/Chronic Adverse Effects: (see section V, VI)

    3. Laboratory Tests

    a. Baseline (i.e. prior to starting therapy) lab tests should

    include liver function tests (SGOT, SGPT, alkaline

    phosphatase), serum albumin, complete blood cell count

    with differential, urinalysis, and serum electrolytes.

    b. In otherwise healthy and asymptomatic patients, routine

    laboratory monitoring after starting therapy is unnecessary

    with clinical laboratory tests only being repeated if indicated

    by the patient's clinical condition. For patients with

    abnormal baseline laboratory tests, further work up is

    required to evaluate their cause and follow-up monitoring

    performed as indicated. (i.e., for a patient with a low WBC

    started on carbamazepine, a CBC should be obtained every

    month for first 1-3 months, then quarterly for next year, and

    then every 6-12 months thereafter).

    4. AED Plasma Concentrations: When used appropriately, AED plasmaconcentrations provide a useful adjunct tool to clinical monitoring to assist in

    optimizing therapy (particularly for carbamazepine, phenytoin, primidone, valproic

    acid). Because of infrequent occurrence of seizures in most patients and wide

    interpatient variability in AED pharmacokinetics, availability of target plasma

    concentrations is helpful in guiding dose adjustments.[Please note however that the

    final assessment of therapy is the patient=s clinical response (i.e., seizure control and

    side effects) not what the plasma concentration is.] AED plasma concentration

    monitoring also is useful in assessing medication compliance and evaluating cause of

    an unexpected loss of seizure control or occurrence of drug toxicity. Role of plasma

    concentration monitoring with newer AEDs is not established and is not currently

    recommended.

    E. Appropriate Use of AED Plasma Concentrations

    1. General Guidelines for Use of AED Plasma Concentrations

    a. A major problem with the use of AED plasmaconcentrations is tendency of many clinicians to treat the

    published therapeutic ranges as a fixed range that is optimal

  • 8/2/2019 Epilepsi at Web

    19/59

    for every patient. It is important to remember that these

    ranges are derived from uncontrolled studies and at best

    should be viewed as a target range to initially aim for as one

    is attempting to optimize therapy in a given patient.

    b. Considerable interpatient variability in therapeutic plasmaconcentrations exists for these drugs, with many patients

    being controlled at levels below or above the published

    ranges. An important point to remember in monitoring AED

    plasma concentrations is that the therapeutic concentration

    for a specific patient is that which controls seizures without

    producing significant adverse effects, regardless of whether

    or not it is within the published therapeutic range for that

    drug.

    c. AED plasma concentrations should be interpreted in termsof what is occurring clinically with patient.

    d. Monitoring of AED plasma concentrations should berestricted to answering questions concerning specific clinical

    problems. Routine monitoring of plasma concentrations in

    otherwise stable patients serves no purpose except to tell you

    what you already know and unnecessarily increases the cost

    of therapy.

    2. Appropriate Indications for AED Plasma Concentration Monitoring

    a. Guiding dosage adjustments

    b. Identifying patient=s individual therapeutic range

    c. Determing cause of unexpected loss of seizure control oroccurrence of drug toxicity

    d. Evaluating clinical consequences of addition/removal ofpotential interacting drug

    e. Assessment of patient compliance

    3. Factors to Consider Before Obtaining AED Plasma Concentration

    a. Determine reason for sampling, since this will guide whenand at what time to obtain sample

    b. Should the plasma sample be reflective of steady-state ?While not all plasma concentrations need to be at steady-

    state, obtaining a plasma sample prior to achievement of

    steady-state will not provide an accurate assessment of

  • 8/2/2019 Epilepsi at Web

    20/59

    patient=s clinical response to the current drug regimen and

    may result in inappropriate dosage adjustments. To ensure a

    new steady-state has been reached, plasma samples should

    not be obtained for at least 4-5 half lives afetr most recent

    dosage adjustment or change in concurrent therapy.

    c. Sampling time. In most instances, sampling time should beconsistent with times of previous samples to allow an

    accurate assessment between changes in patient=s clinical

    response and plasma concentrations. Obviously exceptions

    to this rule exist. For example, if the indication for the

    plasma level is dose related toxicity, obtaining sample at time

    of peak concentration or occurrence of symptoms may be

    helpful.

    d. Determine need for obtaining free (unbound) plasma drugconcentration or plasma concentration of metabolite.

    4. Evaluation of AED Plasma Concentrations

    a. When was last dose given ?

    b. Is this concentration reflective of steady-state ? (When wasdrug started, when did last dosage change occur, when were

    regimens of any concurrent drugs last changed)

    c. Is patient compliant ?

    d. What is patient=s clinical response to this concentration ?

    e. Is there an intercurrent illness or concomitant drug thatmight alter plasma concentrations or clinical response to the

    AED ?

    f. What are therapeutic goals for this patient ?

    5. Factors Associated with Individual Variation in Therapeutic Plasma Concentration

    a. Seizure Type- patients with partial seizures have been shown

    to require higher plasma concentrations than patients with

    primary generalized seizures (Lambie et al, 1976; Schmidt &

    Haenel, 1984).

    b. Severity of Epilepsy- higher plasma concentrations shown tobe required in patients with higher pretreatment seizure

    frequency and patients in status epilepticus.(Lund, 1974;

    Schmidt & Haenel, 1984)

    c. Altered plasma protein binding- relationship between free

  • 8/2/2019 Epilepsi at Web

    21/59

    and total AED plasma concentration altered. In patients

    with decreased protein binding (increased free fraction),

    clinical response would be expected at a lower total plasma

    concentration than if patient's plasma protein binding were

    normal.

    d. Active Metabolites

    e. Concurrent Drug Therapy: Pharmacodynamic Interactions

    1). Enhancement of Concentration-Related Toxicity: Concurrent use of other AEDs

    (polytherapy), alcohol, antidepressant, antihistamines, antipsychotics,

    benzodiazepines, narcotic analgesics, and sedative hypnotics may result in the

    occurrence of concentration-related neurotoxicity at lower than expected plasma

    concentrations.

    2). Drugs Antagonizing Anti-Seizure Effect of AEDs: bupropion, clozapine,

    imipenem-cilastin, isoniazid, reserpine, tricyclic antidepressant, theophylline andcocaine/amphetamines may lower the seizure threshhold.

    F. Evaluation of Patient with Chronic Active Epilepsy

    1. Review diagnosis/etiology

    2. Review compliance

    a. Evaluation of compliance

    1. Direct questioning of patient using open ended question (e.g.

    "How do you take your medicine?" "Which dose is moredifficult to remember?"

    2. Review of refill patterns

    3. Pill counts

    4. AED plasma concentrations

    a. Approaches for Improving Compliance

    1. Patient education

    2. Simplification of dosage regimens

    3. Flexible, patient-specific administration schedules

    4. Medication calendar

    5. Use of pill cups, boxes, or watch with alarm

  • 8/2/2019 Epilepsi at Web

    22/59

    3. Review Drug History

    AED history should include drugs, doses used, and clinical response in terms of any

    beneficial effects or adverse effects.

    4. Form Treatment Plan

    5. Reduction of Polytherapy

    a. Assess present therapy

    b. Withdraw any nonessential drugs, and drugs present atsubtherapeutic plasma concentrations

    c. If not present, initiate therapy with desired AED(s) andtitrate dose to therapeutic serum concentration range

    d. Concurrent with above, begin reducing drugs that are

    producing adverse effects or potentially toxic.

    e. Once therapeutic plasma concentrations of desired drug isachieved, remove any other AED(s) as appropriate.

    f. Plan should be reassessed based on clinical response andplasma concentration monitoring following each change in

    drug and/or dose.

    g. Anticipate changes that may occur in pharmacokinetics ofremaining drugs as potentially interacting drugs are

    removed.h. Tapering Schedule for withdrawing AED therapy: To

    minimize any exacerbation in seizure control or the

    occurrence of withdrawal seizures, ensure that plasma

    concentrations of desired drug(s) are in the usual

    therapeutic range and then slowly withdrawing the

    unwanted drug over several days to weeks. While the exact

    time schedule for tapering the dose of the drug to be

    withdrawn will depend on the patient=s clinical condition

    and setting, a general recommendation would be to decrease

    the dose by 25% every 1 to 2 weeks. If possible the tapering

    process with carbamazepine, phenobarbital and

    benzodiazepines should be even slower than this, i.e., 25%

    every 2 to 4 weeks. If an exacerbation of seizures occurs

    during drug withdrawal, the dose should be increased to the

    previous level and then, after seizure control has been

    restored, the tapering process restarted using a more

    gradual schedule (i.e., decreasing the dose by a smaller

    amount each time and allowing a longer time between dose

  • 8/2/2019 Epilepsi at Web

    23/59

    decreases).

    G. Termination of Antiepileptic Drug Therapy

    1. Need for continued AED therapy can be reevaluated when patient has been seizure

    free for 2 to 5 years. Once seizure free for 2 to 5 years, studies have shown that 60-

    75% of children and 40-60% of adults may be successfully withdrawn frommedication.

    2. Factors favoring a low risk for recurrence of seizures after medication withdrawal

    include (Annegers et al, Epilepsia 1979; Emerson et al, NEJM 1981; Thurston et al,

    NEJM 1982; Callaghan et al, NEJM 1988; MRC Trial Group, Lancet 1991 ):

    a. Minimum 2 year seizure free period

    b. Normal electroencephalogram

    c. Short duration of epilepsy prior to seizures being controlled

    d. Few seizures after starting AED therapy

    e. Controlled achieved with monotherapy

    f. Age of less than 16 years at onset of seizures

    g. Presence of absence seizures only

    3. Decision should be made on individual basis considering consequences of seizure

    recurrence on patient=s employment, education, lifestyle, and driving priveleges.

    Although little data is available to indicate the optimal withdrawal rate of AED

    therapy or to support a relationship between withdrawal rate and seizure relapse,

    most investigators recommend that treatment be withdrawn gradually over a period

    of at least three months (Callaghan et al, NEJM 1988; Chadwick et al, Br Med J

    1985).

    H. Patient Education

    The main aim of education in the patient with epilepsy is to provide the patient and/or

    their care givers with knowledge needed to allow their active participation in

    management of their epilepsy. Goals of education in these patients include providing:

    1). An understanding of their disease state;

    2). The goals and limitations of drug therapy;

    3). How to appropriately manage their disease (i.e., observing and recording seizures

    and any changes in seizure activity in diary; types of adverse effects to watch for,

    information to record about adverse effects in diary; how and when to report any

    problems with adverse effects or seizure control; what to do if miss dose)

  • 8/2/2019 Epilepsi at Web

    24/59

    4). An understanding of the concept of a therapeutic plasma concentration range,

    steady-state, and relation to regular drug intake (i.e., what occurs when miss dose and

    how long will it take once therapy is restarted for plasma concentrations to return to

    previous level);

    5). An understanding of the importance of compliance with their medication regimen

    and what measures or aides can be used to improve compliance. Also since sooner or

    later every patient will miss a dose, they should be instructed on how to handle missed

    doses. In general, if a single dose is missed, the patient should be instructed to take the

    missed dose as soon as possible. If two sequential doses are missed, one should be

    taken immediately and the other with the next regular dose. If more than two doses

    are missed, patients should contact their physician as the strategy in this situation will

    depend on patient=s clinical condition and on the AED.

    6). Understanding and recognition of drug related adverse-effects and potential drug

    interactions (Who to contact if have questions concerning a potential drug interaction

    or adverse effect, What adverse effects to be aware, When to contact their health care

    provider);

    7). For women of childbearing potential, birth control options and the

    risks/complications associated with pregnancy in a woman with epilepsy should be

    discussed (see pages 24-26 of The Epilepsy Counseling Guide).

    IV. ADVERSE EFFECTS

    A. Dose (Plasma Concentration) Related Adverse Effects

    1. These adverse effects primarily represent the toxic effects of the AEDs on the

    central nervous system, and include somnolence, fatigue, dizziness, vision changes

    (double or blurry vision), nystagmus, ataxia (incoordination), tremor, gastrointestinaldisturbances, difficulty thinking, and behavioral disorders. These adverse effects are

    dose-related and more prominent at higher AED plasma concentrations. Although

    their severity and frequency may vary among agents, the concentration-related

    adverse effects are qualitatively similar among the different antiepileptic drugs

    (AEDs) and are seen in all patients if large enough doses are given. The occurrence of

    these adverse effects end up being the therapy limiting endpoint for most patients.

    2. Due to additive neurologic effects of the AEDs, these adverse effects are more

    frequent and occur at lower plasma concentrations in patients receiving AED

    polytherapy (i.e. more than one AED).

    3. The occurrence of these adverse effects, unrelated to dose, is particularly

    prominent during initiation of therapy (especially with carbamazepine, ethosuximide,

    primidone, felbamate, lamotrigine, tiagabine, topiramate, and valproic acid), but

    disappear as tolerance develops. For this reason, therapy with these drugs should be

    started with low doses and the dose slowly titrated up to the recommended

    maintenance over several weeks.

    4. Transient neurotoxicity during the first hours following drug ingestion may relate

  • 8/2/2019 Epilepsi at Web

    25/59

    to an excessive fluctuation in plasma concentrations between doses and high peak

    plasma concentrations. These peak plasma concentration-related neurologic effects

    commonly result from use of inappropriate dosage intervals relative to drug=s half-

    life or administration of rapidly absorbed dosage formulations.

    5. The occurrence and severity of these neurologic side effects can be minimized by:

    a. Initiating therapy at a low dose and slowly increasing thedose

    b. Avoiding large dosage changes

    c. Restricting therapy to one drug when ever possible

    d. Adjusting the administration schedule (e.g., administrationof the largest dose at bedtime, dividing the daily dose into

    smaller doses given more frequently)

    e. If the occurrence of these adverse effects is primarilyassociated with the time of peak serum concentrations, their

    occurrence may be reduced by administering smaller doses

    more frequently or switching to a more slowly absorbed

    formulation.

    f. Reduction in total daily dose.

    6. Impairment of Cognitive Function/Behavioral Disorders

    a. Impairment of cognitive function (i.e., difficulty thinking)

    and behavioral disorders are the adverse effects that mostsignificantly impact on the patient=s quality of life. Factors

    contributing to these problems in patients with epilepsy

    include the disease state, psychosocial stresses, and AED

    therapy.

    b. Adverse effects on cognitive function and behavior (i.e.,

    hyperactivity, irritability, sleep disturbances, altered mood,

    depression) have been reported with all of the AEDs. The

    severity of these effects is increased with higher plasma

    concentrations and polytherapy.

    c. Differential effects among the various AEDs have not beenapparent in controlled studies, with the exception of

    phenobarbital and the benzodiazepines. Phenobarbital

    appears to decreases cognitive performance to greater extent

    than other AEDs in both adults and children. Barbiturates

    have greatest effect on behavior, including hyperactivity

    which may be seen in 10-50% of children receiving

  • 8/2/2019 Epilepsi at Web

    26/59

    phenobarbital. Newer AEDs, such as gabapentin and

    lamotrigine, appear to have minimal effects on either

    cognitive function or behavior.

    d. Management: recognize that these adverse effects may occur

    with any of AEDs, restrict therapy to lowest effective dose

    and avoid polytherapy, particularly with barbiturates

    B. Idiosyncratic (Hypersensitivity) Adverse Effects

    1. All of the current AEDs, with the exception of some of the newer AEDs

    (gabapentin, topiramate, tiagabine), have been associated with the occurrence of rare,

    but serious idiosyncratic reactions, including aplastic anemia, skin rash,

    hepatotoxicity, pancreatitis, lupus-like reaction, and exfoliative dermatitis/Stevens-

    Johnson syndrome. These reactions are generally rare (about 1 in 20,000 to 50,000

    newly treated), unpredictable, non-dose related and usually occur during the first few

    months of therapy.

    2. Previous recommendations for routine monitoring of blood and urine analyses

    during AED therapy were primarily designed to detect these reactions. However,

    because of their rare occurrence, the frequent occurrences of transient and clinically

    insignificant changes in lab indices during chronic AED therapy, and the fact that

    clinical symptoms are usually present before lab testing reveals any abnormalities;

    the more recent recommendations given in section III. D.3. have been adopted.

    3. Most important approach to monitoring for these serious idiosyncratic effects is

    close clinical monitoring of the patient and education of the patient and family about

    the symptoms that may indicate serious adverse effects and the importance of

    reporting these immediately to their physician.

    a. Severe dermatologic or hepatic reaction withcarbamazepine, lamotrigine, phenytoin, phenobarbital,

    primidone: rash and fever

    b. Valproic acid induced hepatotoxicity or pancreatitis: nausea,vomiting, lethargy, loss of seizure control, jaundice, coma

    c. Aplastic anemia/granulocytopenia: abnormal bruising orbleeding, persistent infection

    4. Proposed Mechanism For Idiosyncratic Adverse Effects With Heterocyclic AED

    Recent studies suggest that the idiosyncratic (hepatotoxicity, rash, pseudolymphoma,

    aplastic anemia) and teratogenic adverse effects occurring with the heterocyclic AEDs

    (i.e., carbamazepine, lamotrigine, phenytoin, phenobarbital, primidone ) may result

    from accumulation of toxic arene oxide metabolites of these drugs. The accumulation

    of arene oxide metabolites results from genetic defect of drug metabolism and is

    enhanced by polytherapy. These AEDs are metabolized by hepatic cytochrome P-450

    monoxygenase to a chemically reactive intermediate metabolite (arene oxide) which is

  • 8/2/2019 Epilepsi at Web

    27/59

    then rapidly detoxified by epoxide hydrolase to an inactive hydroxylated metabolite.

    The predisposition of certain patients to develop idiosyncratic toxicity with these

    drugs appears to be due to a genetic or drug-induced (i.e., valproic acid) reduction in

    epoxide hydrolase activity allowing accumulation of the toxic arene-oxide metabolites.

    Additionally enzyme induction of monoxygenases by other concurrent AED (such as

    carbamazepine, phenobarbital, phenytoin, primidone) serves to further increase thelevels of the reactive arene oxides.

    5. Skin Rash

    Skin rash is the most common hypersensitivity reaction with the AEDs and is seen in

    5-15% of patients receiving carbamazepine, ethosuxmide, lamotrigine, phenytoin, and

    phenobarbital. Onset of rash is usually within first 1-3 weeks of therapy and is

    reversible on discontinuing the causative agent. Although a mild morbilliform rash is

    most common, skin reactions may rarely progress to more severe forms, such as

    exfoliative dermatitis, Stevens Johnson syndrome, and toxic epidermal necrolysis. The

    presence of fever, eosinophilia, desquamation, mucus membrane ulceration, and

    painful dermatitis may serve to differentiate between the benign and more seriousskin reactions. Of the current AEDs, skin rash is only rarely reported with valproic

    acid, gabapentin, felbamate, topiramate, and tiagabine.

    The occurrence of skin rash with lamotrigine therapy is increased by concurrent

    valproic acid therapy, a high starting dose, and a rapid dose escalation schedule when

    initiating therapy. To minimize the occurrence of rash with lamotrigine, therapy

    should be started at a very low dose, particularly if already receiving valproic acid,

    and slowly increased into the effective dose range over 1-3 months.

    6. Hepatotoxicity

    a. Remember: it is not unusual (2 to 40% of patients) to seetransient elevated serum transaminases and alkaline

    phosphatase levels without any clinical symptoms in patients

    on AED (particularly children). These observations will in

    the vast majority of cases resolve on continued therapy and

    require no therapeutic intervention. It is important that

    these cases be differentiated from the rare but more severe

    hepatotoxicity observed with AED treatment: delayed

    hypersensitivity (seen with PHT, Pb, CBZ, Pr) and

    irreversible hepatic coma (VPA).

    b. Delayed Hypersensitivity Reaction

    1. Reported with all of the heterocyclic AEDs (i.e.,carbamazepine, lamotrigine, phenytoin, phenobarbital,

    primidone). Mechanism thought to be an idiosyncratic

    metabolic abnormality in susceptible patients.

    2. Onset in majority of patients within first 6 weeks of therapy.

  • 8/2/2019 Epilepsi at Web

    28/59

    3. Elevated liver enzymes accompanied by signs and symptoms

    of hypersensitivity: rash (86%), fever (84%),

    lymphadenopathy (68%), jaundice (54.5%), hepatomegaly

    (52%), eosinophilia (77%). (Oxley et al; 1983).

    4. 38% of cases may develop fatal hepatic necrosis, multiorganfailure, disseminated intravascular coagulation.

    5. Requires discontinuation of therapy.

    a. Valproate Hepatotoxicity

    1. Usually occurs during first 6 months of therapy.

    2. Proposed mechanism: biochemical defect in fatty-acid

    metabolism, which is accentuated by an enhanced

    production of toxic valproic acid metabolites, which may be

    increased by concurrent therapy with enzyme-inducingAEDs, and concurrent disease states.

    3. Risk factors for development of valproate hepatotoxicity

    include young age and use of polytherapy. Presence of other

    neurological disorders, biochemical diseases, or preexisting

    liver disease further increase risk. Highest risk of hepatic

    fatality is in patients < 2 years of age and on polytherapy.

    Risks for development of fatal hepatic failure range from

    1/500 in children < 2 y/o on polytherapy to 1/37,000 for

    monotherapy patients.

    4. Prodromal symptoms: sudden loss of seizure control,malaise, lethargy, drowsiness, weakness, vomiting, anorexia

    and/or jaundice. In most cases, routine monitoring of liver

    function tests will be of little value in detecting onset of acute

    toxicity.

    5. Prevention

    a. Avoid administering VPA as part of polytherapy regimen tochildren < 3; unless either monotherapy has failed or

    potential benefits of polytherapy merit risk.

    b. Avoid administering VPA to patients with preexisting liverdisease and/or a family history of childhood hepatic disease.

    c. Administer VPA in as low a dose as possible for seizurecontrol

    d. Avoid concomitant administration of valproate and

  • 8/2/2019 Epilepsi at Web

    29/59

    salicylate,

    e. Monitor clinically for such symptoms as vomiting, headache,edema, jaundice, or seizure breakthrough, especially after

    febrile illness. If such symptoms develop, VPA therapy

    should be discontinued until a definitive diagnosis is made.

    Patient should be made aware of these symptoms and what

    to do if occur, when initiated on VPA. Greater physician and

    patient awareness of the primary risk factors have resulted

    in a decrease incidence of fatal VPA hepatotoxicity from

    0.93/10,000 in 1978-84 to 0.20/10,000 in 1985-86.

    a. Felbamate : Acute hepatic failure with felbamate, 16 casesreported after introduction between 1/94 to 12/94.

    Mechanism unknown. Has resulted in severe restrictions

    being placed on use of drug.

    7. Aplastic Anemia/Agranulocytosis

    Although commonly associated with carbamazepine, serious hematological reactions

    (i.e., aplastic anemia and agranulocytosis) have been reported with all of the AEDs,

    except gabapentin, lamotrigine, topiramate, and tiagabine. The frequency of aplastic

    anemia with carbamazepine has been estimated at 1 in 200,000 patients. In contrast,

    to the extremely rare occurrence with carbamazepine and the other AEDs, thirty

    cases of aplastic anemia were reported with felbamate from 1/94 to 12/94, which

    represents an approximate frequency of 1 in 2000 patients.

    Routine monitoring of CBCs is usually of little value in detecting the onset of aplastic

    anemia. The most important approach is to clinical monitor the patient for the

    occurrence of any signs or symptoms indicative of a serious hematologic reaction,

    including bruising, bleeding, and persistent infection.

    C. Chronic (Systemic) Adverse Effects

    1. Long-term AED therapy may lead to a variety of chronic adverse effects, including

    connective tissue, endocrine, GI, hematologic, and neurologic disorders. Chronic AED

    toxicity tends to be drug specific and is not directly related to AED serum

    concentration. While not usually life threatening, these chronic adverse effects may

    have a significant impact on the patient's quality of life. Many of these adverse effects

    can be avoided or minimized by appropriate preventative measures. Factors that may

    predispose a patient to chronic AED toxicity include a long duration of therapy,polytherapy, extended administration of high dosages, repeated or prolonged episodes

    of acute toxicity, poor diet or hygiene, and institutionalization.

    2. Neurological

    a. Chronic Cerebellar Degeneration (Encephalopathy)

    1). Primarily seen in patients receiving prolonged (> 5-10y) phenytoin treatment with

  • 8/2/2019 Epilepsi at Web

    30/59

    high serum concentrations and/or dose. Similar syndrome also reported to rarely

    occur with primidone and phenobarbital.

    2). Characterized by confusion, delirium, muscular hypotonia, choreoathetoid

    movements, orofacial dyskinesias.

    3). May improve in some patients following reduction in dose and/or discontinuationof phenytoin.

    4). Related to chronic degeneration of purkinje cells in cerebellum; actual role of

    phenytoin in this disorder controversial since most patients also have long history of

    uncontrolled generalized tonic-clonic seizures.

    a. Peripheral Neuropathy- reported in approximately 8.5-18%of patients receiving long term phenytoin therapy at high

    doses. Primarily manifested as sensory loss. May or may not

    improve on decreasing dose. May respond to folate

    supplementation. Also has been reported withcarbamazepine and barbiturates.

    3. Gastrointestinal

    a. Increased weight gain - Reported for valproic acid (VPA),

    primarily in children. Usually reverses with continued

    therapy. If not, consider reduction in caloric intake, dose

    reduction or discontinuation of therapy. Mechanism

    unknown. In the recently completed VA Cooperative study

    in adult epileptics, incidence of a large weight gain (>12 lbs/

    5.5 kg) with VPA was 20 % overall and 13 % after 12

    months of therapy.

    b. Anorexia and weight loss (>10 kg) is reported in 5-15% ofpatients receiving felbamate. Mechanism unknown. Usually

    reversible with continued therapy. If persists, the options

    include attempting to increase patient caloric intake,

    reducing the felbamate dose, or discontinuing therapy.

    4. Hematological

    a. Leukopenia, thrombocytopenia and various anemias havebeen reported in isolated cases with the all the AED, except

    gabapentin and lamotrigine. A transient and/or dose-

    dependent leukopenia is seen in 5-10% of patient receiving

    carbamazepine. In most cases, this leukopenia is clinicaaly

    insignificant and requires no action. Presence of persistent

    leukopenia (WBC

  • 8/2/2019 Epilepsi at Web

    31/59

    calls for discontinuation of carbamazepine.

    b. Neonatal Coagulation Defects

    1). Seen in infants born to mothers on phenobarbital, primidone, carbamazepine

    and/or phenytoin. Onset of this hemorrhagic disorder is usually within the first 24

    hours after birth and consists of hemorrhage from atypical sites such as pleural andabdominal cavity. This is in contrast to hemorrhagic disease of the newborn which

    does not develop until 2 to 5 days postpartum and where bleeding is more superficial.

    2). Mechanism thought to be competitive inhibition of Vitamin K metabolism in fetal

    liver preventing production of vitamin K dependent clotting factors.

    3). Management:

    a). Mothers receiving antiepileptic drugs should avoid other drugs with adverse

    effects on hemostatic system during last trimester (i.e., aspirin, indomethacin,

    thiazides, promethazine).

    b). Consider cesarean section if difficult or traumatic delivery is expected.

    c). Mother receiving AED should be given oral Vitamin K1 20 mg/day the last 2 to 4

    weeks of pregnancy

    d). Cord blood should be submitted for immediate clotting studies and fresh frozen

    plasma given if diminished vitamin K dependent factors are found.

    e). Infant should be given 1 mg IV phytonadione immediately after birth.

    f). The neonate should then be monitored carefully and should receive an exchange

    transfusion at the first sign of development of hemorrhage.

    a. Megaloblastic Anemia

    1). Occurs in 0.15-0.75% of epileptic patients on AED, primarily with phenytoin but

    also reported with Pb, Pr and CBZ.

    2). Mechanism appears to be due phenytoin-induced malabsorption of dietary folate

    and/or induction of metabolism.

    3). Readily responds to treatment with exogenous folic acid 1 to 5 mg/day.

    4). Not uncommon to see subclinical macrocytosis and/or low serum folate levelswithout accompanying signs of megaloblastic anemia (1 to 30% of patients on

    phenytoin); present recommendations are not to treat these patients with folate.

    a. A dose-dependent thrombocytopenia and/or plateletdysfunction (due to inhibition of platelet aggregation) has

    been reported infrequently in patients on valproic acid. This

    side effect is probably rare or at least has little clinical

  • 8/2/2019 Epilepsi at Web

    32/59

    significance. Although regular monitoring is probably not

    warranted, a baseline platelet count should be obtained in

    patients starting on VPA. As a precaution, patients on VPA

    prior to surgery should have a platelet count and bleeding

    time performed.

    5. Endocrine

    a. Osteomalacia

    1). Primarily a biochemical disorder with decreased concentrations of Vitamin D and

    calcium, rarely associated with the development of clinical disease.

    2). Attributed to increased hepatic metabolism of vitamin D and/or inhibition of

    calcium absorption. Has been associated with phenytoin, phenobarbital, primidone

    and carbamazepine.

    3). Risk factors associated with the development of clinical disease include: poor

    dietary intake of vitamin D, low exposure to sunlight, drug dose and duration,

    multiple anticonvulsants and male sex.

    4). Treatment: 4000 IU vitamin D3/mm3 BSA per day for 4 months, then maintenance

    therapy of 1000 IU/day.

    a. SIADH

    1). Reported primarily in elderly patients on carbamazepine. Water retention with

    hyponatremia; clinically manifested as headache, mental confusion, dyspnea, and

    acute loss of seizure control after prolonged carbamazepine treatment.

    2). Mechanism unknown; in patients in whom it occurs, it appears to be related to

    serum concentration and may improve with decrease in dose. Baseline urinalysis and

    serum sodium should be obtained in patients prior to starting carbamazepine with

    follow up tests done in patients with occurrence of above clinical symptoms. Often

    mistaken for acute carbamazepine toxicity, should be considered in elderly patients

    complaining of above symptoms.

    a. Hyperglycemia: an uncommon adverse effect reported for

    phenytoin; related to phenytoin impairment of normal

    insulin response to glucose. For most epileptic patients of no

    clinical significance, however should be kept in mind when

    phenytoin being used in diabetic patients. To preventcomplications in these patients, phenytoin should be used in

    lowest effective dose.

    6. Cardiac

    a. Carbamazepine: Cardiac conduction disturbances (2nd or3rd degree A-V block) have been rarely reported with

  • 8/2/2019 Epilepsi at Web

    33/59

    carbamazepine. Reversible on discontinuation. Most

    important risk factor appears to be advanced age. Elderly

    patient starting on CBZ should have EKG performed before

    and after initiation of therapy.

    b. Intravenous Phenytoin: Ventricular arrhythmias,

    conduction disturbances, and hypotension may occur

    following IV phenytoin administration. Solvent, propylene

    gylcol, in parenteral solution appears to be contributing

    factor. Occurrence related both to rate of administration

    and patient age (i.e., more common in elderly patients).

    7. Connective Tissue Disorders

    a. Gingival Hyperplasia

    1). Observed in up to 50% of patients on chronic phenytoin therapy.

    2). Proposed mechanism: alteration of connective tissue repair process and decrease

    in salivary levels of IgA. Initial tissue damage may be related to accumulation of

    arene oxide metabolites as severity worse in patients on polytherapy.

    3). Severity may be reduced by careful oral hygiene.

    a. Hirsutism, acne, hyperpigmentation, and coarsening offacial features- not uncommonly observed in children and

    young adults on chronic phenytoin therapy. Incidence of

    facial hirsutism reported up to 30% in young females.

    b. Alopecia- Reported in 2-12% of patients on valproic acid;characterized by temporary thinning of hair with curly orwavy regrowth. Has not been shown to be dose related

    (although some studies have reported that it occurs more

    commonly in patients on high doses) and usually resolves in

    2-6 weeks without any adjustments in VPA therapy

    required. Reversible on discontinuation of drug.

    c. Dupuytren's Contractures: characterized by palmar

    nodules, frozen shoulder, generalized joint pain. Although

    first reported with AEDs in 1925, the 1985 VA monotherapy

    trial was first study to show direct association with

    phenobarbital and primidone therapy. Seen after at least 6

    months of therapy with Pb or Pr and reversible on D/C.

    Incidence 5-10% with an increasing incidence longer

    patients on drug.

    8. Teratogenicity

    a. Expected incidence of congenital malformations (Browne,

  • 8/2/2019 Epilepsi at Web

    34/59

    1983):

    o 2-3% general population

    o 4-5% children born to mothers with epilepsy not taking

    AED

    6-11% children born to mothers with epilepsy taking AED; an incidence similar to

    that in untreated mothers has also been reported in offspring of epileptic fathers.

    a. Present data does not support a specific teratogenic effectfrom AED, but indicate that congenital abnormalities that

    occur normally in epileptic patients occur more frequently

    with use of AED. Present data supporting an association

    between AED exposure and development of malformations

    include:

    1). higher malformation rates in children of treated mothers as compared to

    untreated mothers with epilepsy

    2). higher AED serum concentrations found in serum of mothers with malformed

    children than mothers of healthy children (Dansky et al, Neurology 3:15, 1980)

    3). higher malformation rates seen in infants exposed to polytherapy than those

    exposed to monotherapy during gestation (Lindhout et al, Epilpesia 25:77, 1984;

    Kaneko et al, Epilepsia 29:459,1988). Both the Lindhout and Kaneko studies indicate

    that the most teratogenic AED combinations are those with VPA plus one or more

    enzyme inducing AEDs (PHT, Pb, Pr, CBZ).

    4). higher malformation rates are not seen in infants exposed to maternal seizuresduring gestation than in infants whose mothers had no seizures

    a. With the exceptions of trimethadione and valproic acid,recent studies do not indicate a difference among different

    AEDs in either the incidence or type of malformations

    observed.

    b. Primary abnormalities seen: cardiovascular malformations(2.0%) and cleft lip/palate (1.8%), Skeletal abnormalities

    (1.0%), hypospadias (0.5%), dysmorphia with

    developmental retardation (0.5-1%). Incidence of neural

    tube defect (spina bifida) has been reported as 1-2% forinfants exposed to valproic acid in utero and 0.5-1% for

    infants exposed to carbamazepine.

    c. "Fetal AED syndrome" is manifested by cranial facialanomalies, dysmorphic nasal features, epicanthal folds, wide

    mouth, prominent lips, digit hypoplasia and mental

    retardation. Although at one time primarily associated with

  • 8/2/2019 Epilepsi at Web

    35/59

    phenytoin use, more recent studies show that incidence of

    this syndrome with phenytoin is no different than with the

    other AED.

    d. Mechanisms of AED Teratogenicity

    1). AED-induced folate deficiency

    2). Binding of certain AED due to chemical properties as weak acids (i.e.,VPA) to fetal

    tissue

    3). Increased accumulation of arene oxide metabolites of AED and/or decreased

    activity of epoxide hydrolase activity

    a. Management of Epilepsy in Pregnancy

    Refer to Prepregnancy Counseling Guidelines for Women with Epilepsy on page 25 of

    the Epilepsy Counseling Guide.

    V. DRUG INTERACTIONS

    A. Pharmacodynamic Interactions

    1. Enhance AED Neurotoxicty:Concurrent use of other AEDs (polytherapy), alcohol,

    antidepressant, antihistamines, antipsychotics, benzodiazepines, narcotic analgesics,

    and sedative hypnotics may result in the occurrence of concentration-related

    neutotoxicity at lower than expected plasma concentrations.

    2. Drugs Antagonizing Anti-Seizure Effect of AEDs: bupropion, clozapine, imipenem-

    cilastin, isoniazid, reserpine, tricyclic antidepressant, theophylline and

    cocaine/amphetamines may lower the seizure threshhold

    B. Pharmacokinetic Drug Interactions (see Tables 3A and 3B)

    1. Interference with absorption

    2. Reduction in plasma protein binding

    3. Enhancement/Inhibition of hepatic metabolismVI. SPECIFIC DRUG THERAPY FOR EPILEPSY

    Note: Information listed below is based on instructor=s experience and review of

    literature. Indications and dosages may therefore differ from those found in drug=s

    FDA approved product information.

    A. Phenytoin (DilantinR) (PHT)

  • 8/2/2019 Epilepsi at Web

    36/59

    Indications: simple or complex partial seizures, primary or secondarily generalized

    tonic-clonic seizures, convulsive status epilepticus

    Mechanism: blockade or inactivation of neuronal sodium channels, possible action on

    calcium conductance

    Target Plasma Concentration Range: 10-20 mg/mL

    Half Life: 7-42 hours (due to non-linear kinetics dependent on plasma concentration)

    Time to Steady-State: 4 - 21 days

    Adverse Effects

    Concentration Dependent: nystagmus, double-vision, blurred vision, incoordination,

    drowsiness, dizziness, headache

    Idiosyncratic: aplastic anemia, granulocytopenia, hepatotoxicity, rash, exfoliative

    dermatitis/Stevens-Johnson, Lupus-like reaction

    Chronic: gum hypertrophy, acne, hirsutism, peripheral neuropathy, chronic

    cerebellar damage, megaloblastic anemia, osteoporosis, fetal vitamin K depletion.

    Dosage: Maintenance Dose-4-6 mg/kg/day (300-500 mg/day) in adults, 4-10 mg/kg/day

    in children. Initiation of therapy: Therapy with phenytoin can usually be initiated at

    the recommended maintenance dose. Generally therapy is started at a dose of 4-5

    mg/kg/day (or 300 mg/day) in adults and 6-8 mg/kg/day in children < 12 years.

    Following initiation of therapy, steady-state is not achieved for 1-3 weeks because of

    wide variation in pharmacokinetics among individuals and non-linear kinetics of

    drug. Due to saturable hepatic metabolism, subsequent dose adjustments should be

    limited to 30-100mg/day increments once serum concentrations are above 7.5 ug/ml.Administer in 1-2 daily doses.

    Available Dosage Forms:

    a. 30mg and 100 mg capsules as phenytoin sodium

    b. 50 mg chewable tablet, 30 mg/5ml and 125 mg/5 ml oralsuspension with amount expressed as phenytoin acid

    c. 50 mg/ml phenytoin sodium injectable solution for IV useonly

    d. 50 mg phenytoin sodium equivalents/ml fosphenytoin

    sodium injectable solution for IV and IM use

    Advantages: first line agent for partial seizures, inexpensive, once or twice daily

    administration, availa


Recommended