Benzodiazepines



Benzodiazepines


L. John Greenfield Jr.

Howard C. Rosenberg

Richard W. Homan



Benzodiazepines were developed in 1933 from a class of heterocyclic compounds known since 1891 (1). Clinical interest initially focused on their antianxiety and sedative/hypnotic properties. Soon after the introduction of chlordiazepoxide as an anxiolytic agent in 1960, followed by diazepam (2) and nitrazepam (3), the benzodiazepines became the most widely prescribed drugs in the United States. Most have anticonvulsant properties in animal models, and in 1965 diazepam was first used to treat status epilepticus in humans (4,5). Clonazepam was introduced in the 1970s primarily as an antiepileptic drug (AED) (6), and clobazam was later developed to have a reduced sedative effect (7,8). At the turn of the 21st century, however, only a few benzodiazepines have been approved for acute or long-term use as AEDs in the United States.

The mechanism of action remained obscure until the discovery of high-affinity, saturable benzodiazepine binding to a central nervous system (CNS) receptor (9,10). The benzodiazepines also enhanced inhibitory neurotransmission mediated by γ-aminobutyric acid (GABA), the major inhibitory neurotransmitter of the mammalian brain (11). Subsequent studies confirmed that the brain benzodiazepine receptor was, in fact, a binding site on the GABAA receptor, a ligand-gated chloride channel, where these drugs act as positive allosteric modulators (12).


CHEMISTRY AND MECHANISM OF ACTION

The benzodiazepine structure consists of a benzene ring fused with a 7-member diazepine ring, with nitrogens commonly in the 1 and 4 positions (Fig. 58.1). The clinically active benzodiazepines also have a second benzene ring attached to the 5 position. For this reason, the term benzodiazepine now refers most often to the 5-aryl-1,4 agents. Midazolam and flumazenil have fused R1 and R2 substituents, creating further ring complexity. Most antiepileptic benzodiazepines have the 1,4 structure; however, clobazam is a 1,5-benzodiazepine (7,8,13).


Potency

Benzodiazepine potency correlates with binding affinity at the benzodiazepine sites on neuronal GABAA receptors (Table 58.1) (14,15). An electron-withdrawing group at the 7 position (chloride for diazepam, lorazepam, and clorazepate; a nitro group for clonazepam and nitrazepam) increases receptor binding affinity (16) and especially potency; all useful anticonvulsant benzodiazepines have such a group. Reducing the 7-nitro substituent of clonazepam to produce its less active metabolite (7-amino-clonazepam) greatly decreases receptor-binding affinity and anticonvulsant potency. A methyl group on the nitrogen at position 1 (as in diazepam and clobazam) increases binding affinity and potency; a hydroxyl group at position 3 (as in lorazepam) decreases potency and binding affinity. Several benzodiazepines have a halogen at the 2′ position (chloride for lorazepam and clonazepam) that increases receptor-binding affinity and potency. By itself, however, relative potency is not indicative of a drug’s anticonvulsant selectivity or therapeutic usefulness.


Efficacy

Activity at the GABAA receptor is a function of the drug’s affinity for the benzodiazepine binding site and its intrinsic allosteric effect on that receptor. Individual compounds have widely variable efficacy. Benzodiazepines used as AEDs are thought to be full agonists that maximally enhance GABAA receptor activity. Competitive antagonists bind to the benzodiazepine site but do not affect GABAA receptor function. The antagonist, flumazenil, is used to reverse benzodiazepine-induced sedation in anesthesia (17,18) and to treat benzodiazepine overdose (19). Several “partial agonists” at the benzodiazepine binding site—including abecarnil (20), imidazenil (21), and bretazenil
(22)—although less effective than full agonists like diazepam, are anticonvulsant in animal models and appear less likely to promote tolerance (23,24). Other “inverse agonists” at the benzodiazepine site, including some β-carbolines, inhibit GABA binding or GABA-evoked currents (25). They can induce convulsive seizures or anxiety (25,26) but as yet have no clinical utility.






Figure 58.1 1,4-Benzodiazepine structure. For anticonvulsants, R1 = H or CH3; R3 = H, OH, or COO; R7 = Cl or NO2; and R2′ = H or Cl.


Anticonvulsant Activity

Benzodiazepines are effective against virtually every type of experimental seizure, but individual drugs show large quantitative differences for specific seizure models and other clinical effects (27). Benzodiazepines are particularly effective against seizures induced by the convulsant pentylenetetrazol, often considered a model of generalized absence seizures (28), but are less useful against tonic seizures induced by “maximal electroshock” (29), a model of generalized tonic-clonic convulsions. Relative doses necessary to achieve a particular effect also differ. The diazepam dose to block pentylenetetrazol seizures is 1% of that to abolish the righting response; for clonazepam, it is less than 0.02%, suggesting a larger therapeutic window. However, the dose of diazepam necessary to block maximal electroshock seizures is 11-fold higher than the dose required to suppress pentylenetetrazol seizures but is 2000-fold higher for clonazepam (27); this suggests that diazepam is more useful against generalized tonic-clonic seizures. Diazepam (30) and clorazepate (31) also slow the development of kindling, an animal model in which repeated subconvulsive electrical stimulation produces increasingly severe seizure activity (32).








TABLE 58.1 ANTICONVULSANT ACTIVITY, MOTOR IMPAIRMENT, AND RECEPTOR BINDING OF SOME BENZODIAZEPINES






























Benzodiazepine


ED50 for Clonus Suppression in Kindled Rats (mg/kg)a


ED50 for Ataxia (mg/kg)a


IC50 for Inhibiting [3H]Flunitrazepam-Specific Binding, nMb


Clobazam


2.8


13.2


870


Diazepam


0.4


1.5


78


Clonazepam


0.09


0.9


16


7-Amino-clonazepam


>40



195


a Dose required to inhibit forelimb clonus or to cause ataxia in 50% of amygdala-kindled rats. (Data from Tietz EI, Rosenberg HC, Chiu TH. A comparison of the anticonvulsant effects of 1,4- and 1,5-benzodiazepines in the amygdala-kindled rat and their effects on motor function. Epilepsy Res 1989;3:31-40, except 7-amino-clonazepam data, which is from HC Rosenberg, EI Tietz, and TH Chiu, unpublished data, 1987.)

b Concentration required to displace 50% of 2 nM [3H]flunitrazepam specifically bound to rat cerebral cortical membranes. (Data from EI Tietz, TH Chiu, and HC Rosenberg, unpublished data, 1990.)



GABAA Receptor Complex

The association, or “coupling,” between benzodiazepine binding and GABA binding (33), demonstrated in early binding studies, suggested activity at a specific receptor associated with the bicuculline-sensitive GABAA receptor to modulate its actions on a chloride channel (33, 34, 35). Thus, a “GABAA receptor complex” appeared to incorporate binding sites for GABA, benzodiazepines, and barbiturates with a ligand-gated chloride channel. Electrophysiologic studies demonstrated that benzodiazepines increased the amplitude of GABA-mediated inhibitory postsynaptic potentials (IPSPs) (12) by raising the opening frequency of the GABA-gated chloride channel (36). This mechanism was later confirmed with single-channel studies (37).

In whole-cell patch-clamp recordings of CNS neurons, benzodiazepines shift the concentration-response curve
for GABA leftward (Fig. 58.2), increasing current amplitudes at lower GABA concentrations but not increasing maximal current (38). This shift is caused by an enhanced affinity for GABA at its binding site, with no change in channel-gating kinetics (37). The benzodiazepines thus increase the current produced by low GABA concentrations but have no effect at high GABA concentrations, at which receptor binding is saturated. Studies of GABAergic inhibitory postsynaptic currents (IPSCs) have suggested that GABA is present in the synaptic cleft for 1 to 3 ms at high concentrations (about 1 mM) (39,40). Thus, at individual synapses, benzodiazepines prolong the mIPSC decay phase (41,42) by slowing the dissociation of GABA from the receptor (43,44) without changing maximal mIPSP amplitude. Prolongation of the mIPSC increases the likelihood of temporal and spatial summation of multiple synaptic inputs, which, in turn, raises the amplitude of stimulus-evoked IPSCs. The benzodiazepines thus increase the inhibitory “tone” of GABAergic synapses, which prevents or limits the hypersynchronous firing of neuron populations that underlies seizure activity (45).


GABAA Receptors and Epilepsy

The anticonvulsant properties of benzodiazepines are likely related to the prominent role of GABAA receptors in epilepsy. Substantial evidence links epilepsy with dysfunction of GABAergic inhibition (45). GABAA receptors are the target not only of the benzodiazepines but also of the barbiturates and, indirectly, of tiagabine and vigabatrin, which increase GABA concentration at the synapse (45). Several animal models of epilepsy demonstrate altered numbers or functions of GABAA receptors (46). Moreover, changes in the composition or structure of the transmembrane protein subunits that make up GABAA receptors can result in epilepsy. Expression of GABAA receptor subunits is altered in the hippocampi of experimental animals with recurrent seizures (47) and in patients with temporal lobe epilepsy (48,49). Mice lacking the GABAA receptor β3 subunit have seizures and behavioral features of Angelman syndrome (50), a neurodevelopmental disorder associated with severe mental retardation and epilepsy caused by a mutation affecting the β3 subunit on chromosome 15q11-13 (51). In addition, two mutations in the γ2 subunit that impair GABAA receptor function (52), K289M (53) and R43Q (54), have been linked to a syndrome of childhood absence epilepsy and febrile seizures; a loss-of-function mutation in the α1 subunit was found in a family with autosomal dominant juvenile myoclonic epilepsy (55).


Other Benzodiazepine Actions

With a few exceptions (26), the benzodiazepines derive anticonvulsant properties from specific interaction with GABAA receptors. In one study, their high-affinity interactions with GABAA receptors did not fully explain the anticonvulsant effect, part of which occurred at concentrations much higher than necessary to saturate the GABAA receptor benzodiazepine binding site, was exponential rather than saturable, and was not antagonized by flumazenil (56). Benzodiazepines, but not GABA or muscimol, were anticonvulsant when injected into the substantia nigra pars reticulata (57,58). Pharmacodynamic issues in benzodiazepine metabolism may explain these findings (see p. 834); alternatively, other sites of action may be involved. At doses used to treat status epilepticus, benzodiazepines also inhibit voltage-gated sodium (59) and calcium channels (60) and increase GABA levels in cerebrospinal fluid (61). Benzodiazepines, however, have no interaction with the G-protein-linked GABAB receptor, which can either suppress voltage-gated Ca2+ channels or activate inward-rectifying K+ channels (62).

Benzodiazepines also bind to the “peripheral benzodiazepine receptor” (PBR) (63,64), an 18-kDa protein that functions as part of the mitochondrial permeability transition pore involved in cholesterol transport (64), apoptosis, and regulation of mitochondrial function (65). Although the PBR is widely expressed throughout the body, in the central nervous system (CNS) it is restricted to ependymal cells and glia (63) and is therefore unlikely to have a role in the clinical properties of benzodiazepines.


Excitatory GABAA Currents

Benzodiazepine enhancement of GABAA receptor function may not always be anticonvulsant or even inhibitory. Early in CNS development, neurons do not express the major chloride transporters (e.g., the potassium-chloride cotransporter, KCC2) that lower intracellular chloride concentration and create the negative driving force for chloride ions found in adult neurons. As a result, activation of GABAA-receptor channels can be depolarizing and excitatory and may play a trophic role in neuronal morphogenesis or synaptogenesis, or both (66,67). In fact, endogenous GABA appears to be proconvulsant in early postnatal rat hippocampal slices, as GABAA antagonists blocked epileptiform activity induced by depolarization with high external [K+] (68). However, benzodiazepine efficacy appears to be intact, likely because persistent opening of GABA channels (in the presence of benzodiazepines) may reduce the depolarizing chloride reversal potential, resulting in “shunt” inhibition or subthreshold depolarization that inactivates sodium channels and prevents firing of action potentials (69,70).

The current through GABAA receptor channels can be altered by changes in concentration of intracellular bicarbonate (71,72), which also flows though the channel (73). These changes may underlie a reduction in synaptic GABA current amplitudes during development of benzodiazepine tolerance (74,75). Depolarizing GABAA currents may also be a source of interictal spike activity, as observed in epileptic subiculum neurons in hippocampal brain slices
removed from patients with temporal lobe epilepsy (76). Changes in the GABA current reversal potential might also explain why diazepam can be less effective in children with epileptic encephalopathies (77), and why, rarely, it can cause status epilepticus in patients with Lennox-Gastaut syndrome (78,79).


Molecular Biology of GABAA Receptors

The GABAA receptors have binding sites for agents that modulate receptor function, including the benzodiazepines, barbiturates, neurosteroids, general anesthetics, the novel anticonvulsant loreclezole, and the convulsant toxins picrotoxin and bicuculline. Recent research into the molecular biology of GABAA receptors has clarified the mechanisms of benzodiazepine action.

GABAA receptors belong to the ligand-gated channel superfamily that includes the nicotinic acetylcholine, glycine, and the serotonin 5HT3 receptors (80). GABAA receptors are pentameric (81) transmembrane chloride channels assembled from combinations of protein subunits from several families (Fig. 58.2). Seven subunit families have been identified (12), with 30% to 40% homology between families and about 70% homology within families. In mammals, 16 subunit subtypes have been cloned, including 6 α, 3 β, and 3 γ subtypes, single members of the δ, π (82), and ε (83) families [θ (84) may be the fourth member of the β family (85)], as well as alternatively spliced variants of the β2 and γ2 subtypes. Random pentomeric combinations of all of the subunits would produce tens of thousands of subunit compositions. Because GABAA receptor subtypes are differentially expressed by CNS region and cell type (86) and are developmentally regulated (87,88), the total number of possible isoforms in specific brain regions and individual neurons is reduced. The most common GABA receptor conformation contains the α1, β2, and γ2 subtypes, with presumed stoichiometry of 2α, 2β and a single γ subunit; the δ subunit may in some cases substitute for γ. The subunits are arranged around a central water-filled pore that can open to conduct Cl ions when GABA is bound (Fig. 58.2). Studies of recombinant receptors have shown that individual subunit subtypes confer different sensitivities to GABAA-receptor modulators including benzodiazepines (89,90), loreclezole (91), and zinc ions (92).






Figure 58.2 Model of a GABAA receptor in the plasma membrane. The receptor consists of five closely related subunits, each with four membrane-spanning domains. The receptor is a ligand-gated anion channel, with two binding sites for GABA (between α and β subunits) and one for benzodiazepines (between α and β subunits). In the presence of GABA, the channel becomes permeable to chloride ions, producing the fast inhibitory postsynaptic potential (IPSP). In the presence of a benzodiazepine agonist, GABA binds more efficiently, enhancing the IPSP. Benzodiazepines have no effect in the absence of GABA.



GABAA Receptor Subunits and Benzodiazepine Pharmacology

Benzodiazepine augmentation of GABAA receptor currents requires a γ subunit, and the specificity of benzodiazepine responsiveness is determined by which α subunit is present (12,93). The effect of GABAA receptor subunit composition on benzodiazepine binding has been well characterized by radioligand binding studies (94) and electrophysiology of the recombinant receptors expressed in fibroblasts (85,95). The α1 subunit results in a receptor with high affinity for the imidazopyridine hypnotic zolpidem, defining the “BZ-1” (or Ω-1) receptor type (94,96). The α2 and α3 subunits, combined with β and γ, result in BZ-2 receptors with moderate zolpidem affinity. GABAA receptors with the α5 subunit or the γ3 subunit are sensitive to diazepam but have essentially no affinity for zolpidem and are called BZ-3 receptors. GABAA receptors with the α4 or α6 subunits are insensitive to most benzodiazepines. Given the dependence of benzodiazepine binding and action on α and γ subunits, the benzodiazepine binding site is, not surprisingly, in a cleft between them (97).


GABAA Receptor Subunits Mediate Specific Benzodiazepine Effects

Exploration of the roles of the α subunits has been based on the discovery of a single histidine (H) residue found in all benzodiazepine-sensitive α subunits (H101 in the rat α1 subunit), but not in the benzodiazepine-insensitive α4 or α6 subunits. Mutation of the arginine (R100) to H in α6 dramatically increases benzodiazepine binding in this normally insensitive subunit, while mutation of H101 to R in α1 reduces benzodiazepine sensitivity (98); this H residue was discovered in a strain of “alcohol-nontolerant” rats that had a point mutation in the α6 subunit (R100Q). This spontaneous mutation made their α6-containing GABAA receptors (found mostly in the cerebellum) sensitive to diazepam and likely accounted for their ethanol and benzodiazepine intolerance (99). Subsequent studies have used mice carrying “knock-in mutations” with individual α subunits mutated to be benzodiazepine insensitive. In homozygous α1 (H101R) knock-in mice, the benzodiazepine anxiolytic effect was intact, but the drug did not protect against pentylenetetrazol-induced convulsions and did not produce sedation or amnesia; therefore, binding to the (wild type) α1 subunit may be responsible for sedative, amnestic, and anticonvulsant actions (100,101). Moreover, the sedative-hypnotic zolpidem showed no sedative effect in α1 (H101R) mice (102). Unfortunately, these findings underscore the association between sedative and anticonvulsant efficacy for the benzodiazepines at α1-containing GABAA receptors. Corresponding knock-in mutations of the α2 and α3 subtypes (α2[H101R] and α3[H126R])] suggested that anxiolytic (103) and myorelaxant (104) properties of benzodiazepines derive from α2– and α3-containing GABAA receptors; the α5 subunit is critical for amnestic effects (105).

Further underscoring the role of individual subunits in GABAA receptor function were studies that used antisense oligodeoxynucleotides (ASO) to selectively reduce the expression of specific subunits. Progesterone withdrawal results in anxiety and increased seizure susceptibility associated with an increased expression of the benzodiazepine-insensitive α4 subunit (106). Pretreating rats with an ASO against the α4 subunit prevented the increase in seizure susceptibility (107); this finding may have significance for catamenial epilepsy. Treatment with an ASO for the γ2 subunit increased the convulsive threshold dose for methyl-β-carboline-3-carboxylate, a benzodiazepine inverse agonist, but not for picrotoxin or strychnine (108). ASO γ2 treatment also reduced benzodiazepine binding but not binding to the GABA recognition site (109), indicating a reduction in benzodiazepine binding sites without a change in the number of GABAA-receptor complexes. Similarly, mice lacking the γ2 subunit expressed GABAA receptors with almost no benzodiazepine recognition sites and only a minor reduction in GABA binding sites (110).


Benzodiazepine Tolerance and GABAA Receptor Plasticity

Long-term benzodiazepine treatment gives rise to tolerance, decreases in sedative or anticonvulsant properties, and dependence, the continued need for drug to prevent a withdrawal syndrome (111). Tolerance requires escalation of drug doses and increases the risk of withdrawal seizures. Long-term use of benzodiazepines can also reduce their subsequent effectiveness in acute conditions (112), such as status epilepticus. Withdrawal symptoms typically involve exacerbation of the initial anxiety, insomnia, or seizures and are more common with short-acting than long-acting agents. Rebound symptoms typically return to baseline within 1 to 3 weeks after discontinuation of the drug (113). In animal studies, tolerance develops proportionally to agonist efficacy. Partial agonists produce much less tolerance than full agonists, and the antagonist flumazenil causes no tolerance-related changes in receptor number or function (24). As tolerance to one benzodiazepine may not induce tolerance to a different one, drug-specific interactions at their receptors may be operative (114). The duration of tolerance also varies among benzodiazepines (115). Several studies have noted changes in expression of GABAA receptor subunits (116, 117, 118) as well as altered synaptic physiology (119,120) that depend not only on the drug and dosage but also on the duration and method of administration; all contribute to benzodiazepine receptor occupancy. Measurements of tolerance also vary by seizure model (pentylenetetrazole, bicuculline, pilocarpine, kindling, etc.) and by the behavioral tests used to assess the other benzodiazepine clinical properties (121).



Flumazenil: Uses in Epilepsy

Flumazenil (RO15-1788) binds to the benzodiazepine site without changing GABA site binding or GABA-evoked currents; it thus meets the pharmacologic definition of an antagonist. Used primarily to reverse benzodiazepine-induced sedation (17,18), flumazenil may also reverse hepatic coma (122,123) in patients naive to benzodiazepines; this bolsters arguments for an endogenous benzodiazepine ligand or “endozapine” displaced by flumazenil (124). The role of an endogenous “diazepam binding inhibitor” peptide (125) in inhibitory neurotransmission remains unclear.

Brief exposure to flumazenil can reverse tolerancerelated changes in GABAA receptor function (126,127) and subunit expression (128). The use of intermittent low doses was explored in three patients with daily seizures who had become tolerant to clonazepam (1 mg twice daily) (129). A single intravenous dose of flumazenil (1.5 mg) resulted in mild shivering lasting 30 minutes, followed by seizure freedom for an average of 13 days. Refinement of this approach may extend the use of benzodiazepines as long-term therapy for epilepsy.

Curiously, flumazenil has shown anticonvulsant efficacy in some animal models, possibly as a result of partial agonism at high doses (130,131) or antagonism of an endogenous proconvulsant (129). Epileptiform discharges in hippocampal slices were also reduced (132), and kindling was slowed (133). Doses from 0.75 to 15 mg suppressed focal epileptiform activity in six patients with temporal lobe seizures but did not affect generalized spike-and-wave activity in six patients with generalized seizures (134). Several small studies have suggested possible benefit as an AED in humans (134,135). Orally administered flumazenil reduced seizure frequency by 50% to 75% in 9 of 11 previously untreated individuals with epilepsy and in 9 of 16 patients when used adjunctively (136). Prevention of interictal epileptiform discharges on the electroencephalogram (EEG) was similar to that of diazepam (129,137).

Flumazenil can precipitate seizures in patients with hepatic encephalopathy or benzodiazepine dependence or in patients who have attempted overdose (e.g., with tricyclic antidepressants) (138). Flumazenil has been administered to patients previously treated with benzodiazepines to precipitate partial seizures during inpatient localization of seizure onset (139). [11C]Flumazenil has been used in positron emission tomography studies to demonstrate regions of neuronal loss associated with epilepsy (140,141) and may aid in localizing the seizure focus in patients with dual pathologic conditions (142).


ABSORPTION, DISTRIBUTION, AND METABOLISM

The major anticonvulsant role of the benzodiazepines is as first-line therapy for status epilepticus and seizure clusters. Intravenous administration is preferred (143) but may be difficult or impossible in very young children, necessitating use of a rectal (144, 145, 146, 147, 148, 149), intraosseous (150), buccal (151,152), or nasal (153, 154, 155) route. With intravenous administration, the drug’s effectiveness is determined by how fast it crosses the blood-brain barrier. The benzodiazepines are highly lipophilic and rapidly penetrate the blood-brain barrier (156), although the rate of penetration varies more than 50-fold among agents and depends on their substituent groups (157). Protein binding correlates with lipophilicity and is high for most benzodiazepines (nearly 99% for diazepam). After oral ingestion, the benzodiazepines are fully absorbed except for clorazepate, which is rapidly decarboxylated in the stomach to N-desmethyldiazepam (nordazepam) before absorption.

Despite generally long plasma half-lives, most benzodiazepines are relatively short-acting after administration of a single dose owing to rapid distribution from the brain and vascular compartment to peripheral tissues (158,159). A two-compartment model best describes their pharmacokinetics: high levels occur rapidly in the brain and other wellperfused organs, then decline rapidly with an initial brief half-life as a result of distribution into peripheral tissues and lipid stores; a much slower elimination half-life (t1/2β) related to enzymatic metabolism and excretion follows. For example, the t1/2β of diazepam ranges from 20 to 54 hours (160), but the duration of action after a single intravenous injection is only 1 hour, with peak brain concentrations present for only 20 to 30 minutes (161). Distribution is fastest for the most lipophilic agents. Elimination may be prolonged by enterohepatic circulation, particularly in the elderly. These agents cross the placenta and are secreted into breast milk.

Metabolism occurs in the liver by the cytochrome P (CYP) 450 enzymes CYP3A4 and CYP2C19. Inhibitors of CYP3A4, such as erythromycin, clarithromycin, ritonavir, itraconazole, ketoconazole, nefazodone, and grapefruit juice, can slow benzodiazepine metabolism (162). Relatively little induction of hepatic enzymes occurs. Biologically active metabolites (e.g., nordazepam as a metabolite of diazepam) can significantly prolong the biologic half-lives of these agents. Figure 58.3 illustrates benzodiazepine metabolic pathways. The biotransformation and pharmacokinetics of these drugs have been extensively reviewed (56,163, 164, 165) and are presented in detail below for the individual agents.


DRUG INTERACTIONS

Benzodiazepines interact with other drugs more through pharmacodynamic than pharmacokinetic mechanisms. They are not potent enzyme inducers, nor do they strongly affect plasma protein binding of other drugs. CNS depression is increased when benzodiazepines are given with other CNS-depressant drugs (157,166).
Pharmacokinetic interactions with other anticonvulsants except phenobarbital are infrequent and inconsistent. Diazepam enhances phenobarbital elimination (167), and phenobarbital increases the clearance (168) and lowers plasma levels of clonazepam (169). Valproate reduces diazepam protein binding, increasing free drug levels (170), and enhances diazepam’s CNS effects (167). Other AEDs may augment metabolism and clearance of N-desmethyldiazepam derived from clorazepate (171). Clobazam increases the 10-11 epoxide metabolite of carbamazepine (172).






Figure 58.3 Metabolism of the anticonvulsant benzodiazepines.

Benzodiazepines interact little with other drugs but are affected by cytochrome P450 inhibitors. Cimetidine decreases the clearance of diazepam (173,174) and nitrazepam (175). Rifampin increases the clearance and shortens the half-life of nitrazepam (176). Probenecid markedly increases the half-life of lorazepam (177).


ANTIEPILEPTIC EFFICACY


Status Epilepticus

A cause of significant morbidity and mortality (178,179), status epilepticus requires urgent treatment to avoid neuronal damage and its neurologic consequences (180,181). The benzodiazepines have become initial therapy for status epilepticus owing to their rapid onset and proven efficacy (182), with relatively minor cardiotoxic reactions or respiratory depression compared with barbiturates (183). In the multicenter, double-blind Veterans Affairs Cooperative Status Epilepticus Trial (184), patients were randomized to receive lorazepam (0.1 mg/kg), phenytoin (18 mg/kg), phenobarbital (15 mg/kg), or diazepam (0.15 mg/kg) followed by phenytoin (18 mg/kg). Lorazepam was effective in 64.9% of patients, phenobarbital in 58.2%, diazepam/phenytoin in 55.8%, and phenytoin in 43.6% (p <0.001 lorazepam vs. phenytoin alone). Efficacy appeared to correlate with the rate at which therapeutic drug concentrations were achieved; lorazepam required the shortest time, phenytoin the longest (p <0.001).

Lorazepam and diazepam were compared for treatment of simple partial, complex partial, and secondarily generalized tonic-clonic status epilepticus in a double-blind study of 78 adults with symptomatic localization-related epilepsy (182); idiopathic generalized epilepsy with absence status epilepticus was also represented. After the first injection, lorazepam (4 mg) stopped status epilepticus in 78% of patients and diazepam (10 mg) in 58%; effectiveness was 89% and 76% after the second injection (p = NS between drugs). An open-label, prospective, randomized trial compared lorazepam (0.05 to 0.1 mg/kg) and diazepam (0.3 to 0.4 mg/kg) in children with acute convulsions including convulsive status epilepticus (185). Lorazepam was statistically more effective (p <0.01) after the first dose and apparently safer. Its superior efficacy may be the result of a longer duration of action, based on a longer distribution half-life.

Lorazepam may replace diazepam for prehospital treatment of status epilepticus. A large clinical comparison found that status epilepticus had terminated by the time of arrival at the emergency department in 59.1% of patients treated with lorazepam (2 mg), in 42.6% treated with diazepam (5 mg), and in 21.1% given placebo (186). Rates of circulatory or ventilatory complications were similar (10.6 for lorazepam, 10.3% for diazepam) and lower than those of placebo (22.5%).


Early treatment increases the probability of seizure termination (186), most likely because prolonged seizures alter GABAA receptor susceptibility to benzodiazepines (187). Reduction in benzodiazepine sensitivity of the GABAA receptor can occur within minutes in status epilepticus (188,189) and may be responsible in part for both the persistent epileptic state and its refractoriness. Refractoriness to diazepam may be mediated by N-methyl-D-aspartate (NMDA) receptor mechanisms, as NMDA antagonists improve the response to diazepam in late pilocarpine-induced status epilepticus (190). Moreover, NMDA receptors are upregulated during benzodiazepine tolerance, and NMDA antagonists (e.g., MK801) can block benzodiazepine-withdrawal seizures (191). These findings suggest a possible strategy for treatment of late benzodiazepine-refractory status epilepticus with combinations of a benzodiazepine (e.g., intravenous midazolam) and an NMDA-receptor antagonist such as the dissociative general anesthetic, ketamine. A recent trial of oral ketamine for refractory nonconvulsive status epilepticus showed efficacy in all five children studied (192). Such approaches will require validation in controlled clinical trials.

Both lorazepam and diazepam have been approved by the U.S. Food and Drug Administration (FDA) for treatment of status epilepticus in adults; diazepam has also been approved in children more than 30 days old. Parenteral preparations of midazolam, flunitrazepam, and clonazepam expand the treatment possibilities. Clonazepam for parenteral administration is currently marketed only in Germany and the United Kingdom, and flunitrazepam is not available in the United States. Intramuscular injection and intranasal (153,154), buccal (151), endotracheal (193,194), or rectal (149,195,196) instillation also rapidly produce therapeutic levels and are effective against status epilepticus or seizure clusters.








TABLE 58.2 CLINICAL PHARMACOLOGY OF BENZODIAZEPINES USED FOR ACUTE SEIZURES















































































































Diazepam


Lorazepam


Midazolam


Clonazepam


Characteristic


IV


Rectal


IVa


Buccala


IVa


IMa


IVb


Initial dose (mg)


10-20


0.5-1/kg


4


2-4


0.125-0.15/kg


0.2/kg


0.01-0.09/kg


Infusion rate


8 mg/h





0.15-0.2 mg/kg/h




Minimum effective concentration


500 ng/mL


NA


30 ng/mL


NA


NA


NA


30 ng/mL


Onset of effect (min)


<1


2-6


<2


NA


<2


2-30


<1


Peak effect (min)


3-15


10-120


30


NA


10-50


25 ± 23


NA


Duration of effect


<20 min


NA


>360 min


NA


<50 min


20-120 min


24 h


Protein bound (%)


96-97


96-97


85-93


85-93


95 ± 2


95 ± 2


86 ± 5


Volume of distribution (L/kg)


133


133


12


12


NA


NA


NA


Distribution half-life


0.96-2.2 h


NA


2-3 h


NA


5.7 ± 2.4 min


NA


NA


Elimination half-life (h)


36 ± 4.9


36 ± 4.9


14.1


14.1


1.9 ± 0.6


1.9 ± 0.6


20-80


Abbreviations: IM, intramuscular; IV, intravenous; NA, not available.

a Not approved by the U.S. Food and Drug Administration for seizures.

b Not available in the United States.



Acute Repetitive Seizures

The need for immediate high drug levels to handle serial seizures is less urgent, and ease of administration by family or allied health workers becomes important. Diazepam rectal gel prevents subsequent seizures during seizure clusters (195,196) and can reduce the frequency of emergency department visits (145). Table 58.2 compares the clinical and pharmacologic properties of the benzodiazepines used for acute seizures. Individual agents can be selected for specific clinical situations. Repeated seizures in a patient rapidly weaned from anticonvulsants for inpatient monitoring could be treated with diazepam (rather than lorazepam), as its shorter peak duration of action may be less likely to suppress seizure activity needed later.


Long-Term Chronic Treatment of Epilepsy

Although their long-term use in epilepsy is limited by sedation and tolerance, benzodiazepines may figure in the adjunctive treatment of myoclonic and other generalized seizure types, or seizures with comorbid anxiety disorders. For example, lorazepam improved control of seizures associated with psychological stressors (197). The benzodiazepines can be an integral part of a rational polypharmaceutical regimen, defined as the minimum effective AED combination for seizure control.

Intermittent administration when seizure thresholds are transiently reduced may be the ideal strategy for benzodiazepines. Not only are they suited pharmacokinetically for such an application, but such short-term use also may avoid tolerance. For example, catamenial seizures improved with intermittent use of clobazam (198). Studies
of efficacy for specific indications with the individual agents are discussed below.


ADVERSE EVENTS

With acute treatment of status epilepticus, the primary toxic reaction is respiratory and cardiovascular depression (182,199), which is largely a result of the propylene glycol solvent (200,201). Sedation and amnesia are relatively unimportant in this setting and difficult to distinguish from the effects of status epilepticus itself. Sedation after termination of convulsive status epilepticus, however, often necessitates EEG evaluation to ensure that a conversion to nonconvulsive status epilepticus has not occurred. Administration of benzodiazepines in conjunction with other CNS-active drugs, such as phenobarbital, may enhance respiratory and cardiovascular toxic reactions (166). Rarely, parenteral administration can induce tonic status epilepticus in patients with Lennox-Gastaut syndrome (79,202). Thrombophlebitis may occur (203,204), and intra-arterial injection may produce tissue necrosis (205).

Only gold members can continue reading. Log In or Register to continue

Stay updated, free articles. Join our Telegram channel

Oct 17, 2016 | Posted by in NEUROLOGY | Comments Off on Benzodiazepines

Full access? Get Clinical Tree

Get Clinical Tree app for offline access