Treatment with Antipsychotics



Treatment with Antipsychotics





As in other areas of clinical science, serendipity as well as careful investigation contributed to our knowledge about antipsychotics, including their



  • Discovery


  • Potential mechanisms of action


  • Range of complications

The efficacy of chlorpromazine (CPZ) was discovered primarily by chance in exploratory clinical trials after it had been initially synthesized as an antihistamine. Its discovery, however, was not entirely fortuitous, as it was chosen for human investigation because it was mildly sedating. The concept of an antipsychotic, however, was unknown. The sedative properties of CPZ then led the French anesthesiologist and surgeon Henri Laborit to use it in a lytic cocktail to reduce autonomic response with surgical stress (1). He also persuaded many clinicians to try it for the treatment of a wide variety of other disorders. In this context, he encouraged Delay and Deniker (2) who then administered CPZ to psychotic patients. The rest is history.

Even though CPZ did not produce a permanent cure for schizophrenia (or for any other psychotic disorder), it had a dramatic impact, benefiting many as no other treatment had before (3). News of its effectiveness spread rapidly, and within 2 years CPZ was used worldwide, altering the lives of millions of psychotic patients in a positive manner.

The relocation of treatment from chronic hospital settings to outpatient community mental health centers is, in great part, due to the efficacy of antipsychotics. Naturalistic studies before the era of psychotropics revealed that two of three psychotic patients (primarily schizophrenia patients) spent most of their lives in state asylums. Before the mid-1950s, there had been a steady increase in state hospital populations, which paralleled the general population growth, but after the introduction of antipsychotics, there was a marked reduction in those hospitalized for various psychoses. Presently, more than 95% of these patients live outside of the hospital, even though many continue to relapse or demonstrate residual symptoms. Thus, although the antipsychotics are not a panacea, they make community-based care a reality for many who would otherwise remain chronically institutionalized.

Reserpine, used as a folk medicine in India, was found to have antipsychotic properties at about the same time as CPZ. Both agents affected the dopaminergic system, albeit in different ways, but the functional results were similar [i.e., lowering dopamine (DA) activity]. This phenomenon continues to be an important factor in hypotheses about the mechanism of action of these drugs and for biological theories about the pathophysiology of psychotic disorders.

The revolutionary idea that drugs could exert a specific antipsychotic effect, coupled with a growing recognition of their significant drawbacks, led to the search for other substances with similar beneficial properties and fewer adverse effects. Thus, pharmacological animal screens, followed by systematic clinical investigations with chemically or structurally related compounds, led to a variety of effective, better tolerated drugs for schizophrenia and other major psychotic disorders.


The therapeutic efficacy of clozapine, the first truly different antipsychotic, spawned the development of a succession of newer agents (e.g., risperidone [RISP], olanzapine [OLZ], quetiapine [QTP], ziprasidone [ZPD], aripiprazole [ARIP], paliperidone [PALI], iloperidone [ILP], and asenapine [ASEN]). Other new compounds (e.g., bifeprunox, lurasidone, sertindole) may also receive US Food and Drug Administration (FDA) marketing approval in the near future (3a, 3b). Antipsychotics can be classified by their chemical class, potency, activities at receptors, or liability for causing neurological adverse effects. This text refers to these drugs as first-generation antipsychotics (FGAs) and second-generation antipsychotics (SGAs). FGAs include all of the agents marketed before clozapine. Currently marketed SGAs worldwide include clozapine, sulpiride, amisulpride, zotepine, RISP, OLZ, QTP, ZPD, ARIP, PALI, ILP, and ASEN. Although these agents have previously been referred to as atypical or novel, these terms seem dated, as SGAs are the most commonly prescribed antipsychotic agents in the United States, Canada, and most other countries.

Table 5-1 presents the classes, trade and generic names, and typical dose ranges for the antipsychotics available in the United States. Figures 5-1 and 5-2 present the basic chemical structure of antipsychotic agents.


Pharmacodynamics

All antipsychotics work in part through the central DA-2 (D2) receptor (most cause blockade, but some [e.g., ARIP and bifeprunox] are partial agonists at the D2 receptor). Extrapyramidal reactions, particularly parkinsonian symptoms, can be a major adverse effect of many of these drugs as well as an important clue to their mechanism of action. True Parkinson’s disease is caused by a DA deficiency in the nigrostriatal system. Further, crystallographic data have demonstrated that the molecular configuration of CPZ is similar to that of DA, which could explain its ability to block the receptors of this neurotransmitter. Drugs with similar structures that do not block DA receptors (e.g., promethazine, imipramine) do not have antipsychotic activity. For example, the isomer of flupenthixol that blocks DA receptors is an effective antipsychotic, whereas the isomer that does not is ineffective (4). Other DA receptors, D1, D3, D4 and D5, may also be implicated in psychosis.








TABLE 5-1 ANTIPSYCHOTIC AGENTS































































































































































































Class/Trade Name


Generic Name


Dosage (Range, Orally, Per Day)


Phenothiazines



Aliphatics




Thorazine


Chlorpromazine


100-1,000 mg




Sparine


Promazine


25-1,000 mg




Vesprin


Triflupromazine


20-150 mg



Piperidines






Mellaril


Thioridazine


30-800 mg



Serentil


Mesoridazine


20-200 mg




Quide


Piperacetazine


20-160 mg



Piperazines




Stelazine


Trifluoperazine


2-60 mg




Prolixin


Fluphenazine


5-40 mg




Trilafon


Perphenazine


2-60 mg




Tindal


Acetophenazine


40-80 mg




Compazine


Prochlorperazine


15-125 mg


Thioxanthenes



Navane


Thiothixene


6-60 mg



Taractan


Chlorprothixene


10-600 mg


Dibenzoxazepines



Loxitane


Loxapine


20-250 mg


Butyrophenones



Haldol


Haloperidol


3-20 mg



Inapsin


Droperidol


2.5-10 mga


Dihydroindolones



Moban


Molindone


15-225 mgb


Dibenzodiazepines



Clozari


Clozapin


100-900 mg


Benzisoxazole



Risperdal


Risperidone


2-8 mg



Invega


Paliperidone


3-12 mg



Fanapt


Iloperidone


12-24 mg


Thienobenzodiazepines



Zyprexa


Olanzapine


5-20 mg


Dibenzothiazepines



Seroquel


Quetiapine


75-800 mg


Dibenzo-oxepino pyrroles



Saphris


Asenapine


10-20 mg


Benzisothiazolyls



Geodon


Ziprasidone


40-160 mg


Quinolinones



Abilify


Aripiprazole


5-30 mg


Diphenylbutylpiperidines



Orap


Pimozide


1-10 mg


a Administered intramuscularly

b “Recently taken off the market.”


Interestingly, some SGAs are nearly devoid of extrapyramidal adverse effects in their usual clinical dosing range. This is an important observation because it means that these two
effects can be dissociated. Initially, the term neuroleptic was applied to all drugs that produce both extrapyramidal and antipsychotic effects, but agents such as clozapine, RISP, OLZ, QTP, ZPD, ARIP, and others can be antipsychotics without being neuroleptics, making the interchangeable use of the terms neuroleptic and antipsychotic no longer appropriate.






Figure 5-1 Basic requirements for antipsychotic activity with first-generation antipsychotics (FGAs); distance between N10 of radical and the base N atom must equa at least three carbons (n = 3), with a suitable substituent R2 mainly determining the qualitative properties of the group; the A2 substituent R1 mainly determines the quantitative properties of the individual compounds.

The evidence that antidopaminergic agents ameliorate psychosis and that full DA agonists can produce psychosis or worsen preexisting disorders supports a central role for DA in any theory of pathophysiology. Further, there is a high correlation among directly measured D2-type receptor binding, the clinical potency of these agents, animal behavioral models, and data about DA blockade.






Figure 5-2 Chemical structures of second-generation antipsychotics (SGAs).

Other drug effects that decrease DA activity also support this position. Thus, when DA synthesis is blocked by α-methyl-p-tyrosine (AMPT), the dose necessary for an antipsychotic effect is reduced (i.e., the dose-response curve is shifted to the left by the interaction between DA and AMPT). A drug such as reserpine that can deplete DA stores has relatively mild antipsychotic properties. Also, partial DA agonists (e.g., ARIP, bifeprunox) bind to DA receptors with higher affinity than endogenous DA but result in lower receptor activity. As a result, under conditions of high activity, they functionally decrease the effects of DA (5).


DOPAMINERGIC PATHWAYS OF THE CENTRAL NERVOUS SYSTEM

In 2000, Arvid Carlsson, Paul Greengard, and Eric Kandel received the Nobel Prize in Physiology or Medicine for their study of nerve cell communication (6). Their work included studies of the neurotransmitter DA and its role in Parkinson disease and schizophrenia. Because the existing evidence strongly implicates the DA system in
psychosis, it is pertinent to discuss the central pathways of this neurotransmitter. There are five tracts:



  • The striatal system (A-9)


  • The mesolimbic system (A-10)


  • The mesocortical system (A-10)


  • The retinal system


  • The neurohypophyseal system

The last system is of interest in regard to elevation in prolactin, an effect most closely associated with FGAs, RISP, and PALI.

Postmortem studies of patients with idiopathic Parkinson’s disease demonstrate cell loss in the striatal system (A-9), directly implicating this tract vis-a-vis the drug-induced pseudoparkinsonian adverse effects. The assumption that psychosis is related to the A-10 system is made by exclusion. Evidence also indicates that clozapine may differentially block DA pathways. Specifically, it seems to act on the mesolimbic dopaminergic system (A-10), while being relatively inactive in the striatal system (A-9); however, this remains controversial. Chronic administration of clozapine decreases the firing rate of A-10 mesocortical tract DA neurons while sparing A-9 neurons of the nigrostriatal pathway. By contrast, FGAs reduce the firing rate of both A-9 and A-10 neurons.

Because clozapine may block specific DA receptors, its antipsychotic activity could be consistent with an antidopaminergic mechanism of action. Conversely, clozapine does not typically induce extrapyramidal symptoms, which are presumably subserved by the A-9 system. Thus, although clozapine is known to block striatal DA receptors, in positron emission tomography (PET) studies, resolution is not sufficient to clarify effects on other tracts. Furthermore, low doses of metoclopramide, which significantly decrease the number of DA neurons spontaneously active in A-9, do not have antipsychotic effects (although high doses do) but can induce tardive dyskinesia (TD) as well as acute extrapyramidal side effects (EPSs).


NEUROPHYSIOLOGICAL STUDIES

Another way to measure striatal (A-9) or mesolimbic (A-10) activity is through neurophysiological studies involving the firing rate of these neurons. The acute administration of antipsychotics will cause DA neurons to initiate a burst pattern of firing, with the opposite occurring when a DA agonist is administered. Eventually, some degree of tolerance develops, and the enhanced DA synthesis decreases with time on drug, as demonstrated in both human cerebrospinal fluid (CSF) studies and rat brain slices. On a more chronic basis, repeated drug administration results in a dramatic decrease in the proportion of spontaneously active dopaminergic neurons (i.e., excitation-induced depolarization blockade of the DA neuron spike-generating region). Both the depolarization blockade and the antipsychotic effects of these drugs develop slowly. FGAs induce nigrostriatal and mesolimbic DA neurons into depolarization blockade, whereas clozapine induces depolarization only in the mesolimbic-mesocortical DA neurons.

The working assumption that the striatal system is involved only with extrapyramidal function (e.g., parkinsonian adverse effects, dystonias, TD) and the mesolimbic or mesocortical system is involved only with psychosis may be an oversimplification. Many of the neuroanatomical studies on the identified dopaminergic tracts are done with rats. In the monkey, by contrast, there are many more DA tracts that are either absent in the rat or at least markedly different. Human systems could be different from those of the rat or the monkey. Understanding the neuropharmacology of the antipsychotics is further complicated by the fact that neither the mesolimbic-mesocortical nor the striatal system is homogeneous but may include various subsystems.


DOPAMINE AUTORECEPTORS

There are also dopaminergic presynaptic receptors (or autoreceptors) that generally play a negative feedback role. These autoreceptors sense the level of neurotransmitter and produce a negative feedback influence on DA synthesis and release. Thus, high DA release results in high intrasynaptic concentrations, which stimulate the autoreceptors, ultimately inducing a slowing of DA synthesis and release. Conversely, blockade of the postsynaptic receptors may lead to positive feedback loops, which can



  • Activate tyrosine hydroxylase in the presynaptic dopaminergic neuron


  • Increase DA synthesis


  • Increase the firing rate


  • Increase the levels of DA metabolites such as homovanillic acid (HVA)


Further complicating the picture is the induction of autoreceptor supersensitivity with chronic antipsychotic administration, as well as the fact that at least one tract—the mesocortical, which projects to the prefrontal cortex—may lack such autoreceptors. Schaffer, et al conducted preliminary, single-dose studies with apomorphine at a dose that stimulated presynaptic DA autoreceptors reducing synthesis and neuroleptic threshold (NT) release. This produced a measurable acute antipsychotic effect (7).


DECREASING DOPAMINERGIC ACTIVITY

Given the typical time course of adaptation to the biochemical and electrophysiological effects induced by antipsychotics in most dopaminergic systems, it is important to consider the time course for their efficacy. Apparent clinical benefit can be seen within a few hours after treatment is initiated, improves at a roughly linear rate for the first 14 days, and then gradually levels off.

Time-dependent changes in plasma HVA (a major metabolite of DA) during antipsychotic treatment are consistent with the hypothesis that the mechanism of these agents involves a slowly developing decrease in presynaptic DA synthesis and release. Thus, there is an initial increase in HVA followed by a more sustained decrease. Although decreases in plasma HVA appear to parallel the time course of antipsychotic efficacy, peripheral DA systems may also contribute to plasma or urine HVA levels. Further, because different brain areas may or may not develop tolerance, it is quite possible that CSF HVA levels reflect those areas that produce most of the HVA but are not relevant to the psychotic process. With these caveats in mind, human CSF and plasma data are generally consistent with animal studies, both of which contribute considerable evidence implicating DA blockade and its aftermath. The relevance of these findings to the biological mechanisms subserving psychosis, however, is yet to be determined.


TOLERANCE

It is important to examine whether tolerance develops to the antipsychotic effect of these agents. For example, Sharma et al. (8) found that nontolerant psychotic patients had a significantly inferior clinical response to antipsychotics, in contrast to their tolerant counterparts. Further, they found an earlier age of illness onset and a more refractory course in the nontolerant group. Although biochemical and electrophysiological tolerances (i.e., return of HVA to more normal levels and depolarization inactivation, respectively) develop in most DA systems, they do not occur in all. Prefrontal and cingulate cortices, for example, may be spared, perhaps because of the absence of autoreceptors in these tracts. Evidence also indicates that many areas in nonhuman primate brains develop biochemical tolerance to antipsychotic-induced HVA rise but other areas do not. Thus, with chronic treatment, certain areas (cingulate, dorsofrontal, and orbitofrontal cortex) maintain their HVA increases. Patients who were examined postmortem after chronic antipsychotic therapy were found to have increased HVA levels in certain areas, such as the cingulate and perifalciform cortex. It is also interesting that stress and benzodiazepine (BZD) receptor agonists can selectively stimulate DA neurons in the mesoprefrontal area but not in other dopaminergic tracts.

By using D2-blocking isotopes in a PET scanner, it is possible to image the striatum in psychotic patients. Evidence from an early PET scan study indicated that some drug-free schizophrenia patients manifested an increase in D2 DA receptors, but another study failed to find such an alteration (9,10). These studies used different receptor ligands, different assumptions, and both had a small sample size, so definitive conclusions are not possible. Finally, when PET scans are done on patients receiving clinically effective doses of FGAs or SGAs, all of these agents (including haloperidol [HPDL], clozapine, RISP, OLZ, and ZPD) produced varying levels of D2 receptor occupancy in the striatum (11,12 and 13). Furthermore, they do so at the typical doses used to treat schizophrenia. In addition, Kapur et al. (13) found that only transiently high D2 receptor occupancy occurred with QTP, suggesting that this was sufficient for efficacy, while minimizing the possibility of EPS or elevated prolactin levels.


INCREASING DOPAMINERGIC ACTIVITY

If decreasing dopaminergic activity benefits psychosis, what is the effect of increasing dopaminergic activity? Potentiation of DA by a variety of mechanisms is a common denominator for
inducing paranoia, hallucinations, and other manifestations of psychosis. For example,



  • L-Dopa (3,4-dihydroxyphenylalanine), which is converted to DA in the body, can produce psychosis as a side effect


  • Amantadine, another dopaminergic drug, can also induce psychotic symptoms


  • Stimulants, such as amphetamine and methylphenidate, are potent releasers of DA, whereas cocaine also interferes with its reuptake, increasing DA concentrations at synaptic sites; all can produce paranoid reactions in some abusers


  • Bromocriptine, apomorphine, lisuride, and other direct-acting DA agonists benefit Parkinson’s disease and can also cause psychotic reactions at high doses

As noted, large doses of amphetamine, cocaine, and other sympathomimetics can cause acute paranoid reactions, either spontaneously in abusers or experimentally in normal volunteers. An injection of a large amphetamine dose in normal control subjects, for example, often produces a paranoid psychosis resembling schizophrenia within hours. Frequent smaller doses over several days can also produce a paranoid psychotic reaction. The duration of an episode usually parallels the length of time the drug remains in the body.

Additional evidence comes from studies of increasing dopaminergic activity in patients with active psychosis. Small intravenous doses of methylphenidate (e.g., 0.5 mg/kg) can result in a marked exacerbation of an acute schizophrenic episode (8). By contrast, such doses usually do not produce psychotic symptoms in normal control subjects or in remitted patients. Methylphenidate is more potent in this regard than dextroamphetamine, consistent with the hypothesis that it intensifies psychotic symptoms by releasing central intraneuronal stores of norepinephrine and DA from the reserpine-sensitive pool (14).

Finally, sensitivity to an amphetamine test dose in recovered patients may predict an impending relapse (15).


OTHER PATHWAYS

Interestingly, when physostigmine, a drug that increases brain acetylcholine (ACh), is administered before methylphenidate, it prevents the exacerbation, indicating that the worsening may be mediated by a dopaminergic-cholinergic imbalance (16). Physostigmine itself, however, does not reduce psychosis, suggesting that the underlying process is not amenable simply to altering cholinergic tone. Subsequently, nicotinic cholinergic receptors were reported to play an important role in the inhibition of the perception of extraneous environmental stimuli. This observation has implications for the deficits in sensory gating reported in schizophrenia (17).

Other systems (e.g., neurotransmitter, neuropeptide, neurohormonal) may also play important central or modulating roles in terms of the psychotic process or adverse effects. In addition to DA and ACh, the following have been implicated:



  • Serotonin (5-HT)


  • Norepinephrine (NE)


  • γ-Aminobutyric acid (GABA)


  • Glutamate (GLU)


  • Neurohormones


  • Neuropeptides

Table 5-2 provides a qualitative summary of the relative affinities (or binding profiles) of SGAs for various relevant neuroreceptors.

One leading hypothesis as to why newer agents produce fewer EPS and perhaps provide greater benefit for negative symptoms is that they have a more potent effect on the serotonin 5-HT2A receptor subtype. Thus, the ratio of 5-HT2A to D2 blockade has become an important measure of the potential “atypicality” of an agent. In addition, these novel antipsychotics have a neuroreceptor profile that differs substantially from that of the FGAs. For example, clozapine is a dibenzothiazepine derivative with affinity for a variety of neurotransmitter receptors, including several serotonin receptors (5-HT1C, 5-HT3, 5-HT6, and 5-HT7) that may modulate DA activity and contribute to the atypical action of this agent. It is likely that clinically effective doses of clozapine also antagonize 5-HT2A and 5-HT2C receptors (18). The resulting increased 5-HT2A/D2 ratio may be one basis for the unique antipsychotic efficacy of clozapine (19). At clinical doses, clozapine also has a lower affinity for D2 receptors, which may account for its decreased propensity to evoke EPS, as well as its negligible neuroendocrine effects (20,21). Other agents (e.g., ZPD,
ARIP) are partial agonists at the 5-HT1A receptor, which may also modulate DA activity.








TABLE 5-2 NEURORECEPTOR BINDING PROFILE OF SECOND-GENERATION ANTIPSYCHOTICSa
























































































































































































































Receptor


Clozapine


Risperidone


Olanzapine


Quetiapine


Ziprasidone


Aripiprazole


Paliperidone


lloperidone


Asenapine


D1


High


Low


High


Low/mod


Low


Low


Low


Low


High


D2


Low


High


High


Low/mod


High


Very high


High


High


High


D3



High


High



High


High


High


High


High


D4


High


High


High


None


Mod


Low


High


Mod


High


D6



Mod






Mod




D7



High






High




5-HT1A





Low/mod


High


High



Low


High*


5-HT2A


High


High


High


Low/mod


High


High


High


High


High


5-HT2A/D2 ratio


High


High


High


High


Very high


Low


High


High


High


5-HT2C


High





High


High




High


5-HT3


High



High



Very low






5-HT10






High






5-HT6


High



High



High




Mod


High


5-HT7



High




High



High


Mod


High


Alpha1


Mod/high


High


Mod/high


Mod/high


Mod


Mod


High


Mod


High


Alpha2


Mod


High


Low


Mod/high


Very low



High



High


Histamine1


High


None


High


High


Mod


Low



Low


High


Muscarinic1


High


Very low


High


None


Very low


Very low


Very low


Very low


Very low


aBased on K1 data


* Also High affinity for 5-HT1B; 5-HT2B; 5-HT5.


Mod, moderate.





Pharmacokinetics

Clinically relevant pharmacokinetic factors involving antipsychotics include



  • Good absorption from the gastrointestinal tract


  • An extensive “first-pass” hepatic effect


  • Subsequent high systemic clearance due to a large hepatic extraction ratio each time the plasma recirculates through the liver


  • Extensive distribution (VD) due to highly lipophilic character


  • Plasma half-life (t1/2) of typically about 20 hours


  • Primary route of elimination through hepatic metabolism (except PALI)


  • The presence of metabolites with varying pharmacological profiles (e.g., some may be more effective than their parent compound [e.g., mesoridazine], some may not reach the brain [e.g., sulfoxides], and some may have greater toxicity than the parent compound).

Table 5-3 lists the relevant pharmacokinetic properties of several SGAs.


THERAPEUTIC DRUG MONITORING

The use of drug plasma levels to effect optimal clinical response and to minimize adverse or toxic effects is standard practice in general medicine (e.g., phenytoin, digoxin) as well as in psychiatry (e.g., lithium, tricyclic antidepressants [TCAs], valproate [VPA]; see Chapter 2). The theoretical basis for plasma level monitoring rests on several factors, including



  • The existence of a long interval between drug administration and clinical response


  • Large interindividual differences in response to the same dose for the same diagnosis, in part reflecting differences in plasma levels achieved on a given dose


  • Possible usefulness in helping to establish the minimally effective dose


  • Possible usefulness in helping to estimate the average dose required to achieve a certain concentration when a positive correlation exists between a given steady state concentration (Css) and the dose required.

Unfortunately, for a number of methodological and clinical reasons, the success achieved with this strategy for some classes of drugs (e.g., certain mood stabilizers) has not been achieved with the monitoring of antipsychotic steady state plasma concentrations. Difficulties in study design have contributed to this uncertainty, including



  • Insufficient sample sizes, especially at the low and the high ranges


  • Inclusion of refractory, nonhomogeneous, nonadherent patients


  • Nonrandom adjustment of the dose based on response, adverse effects, or initial presentation


  • Concurrent treatments


  • Too brief an observation period for clinical effects to occur


  • Variable time of blood sampling


  • Inadequate evaluation of clinical response as well as the indefinite nature of the response


  • Presence of numerous active metabolites


  • Inadequate assay methods, such as radioreceptor assays


EXTRAPOLATION FROM PLASMA TO BRAIN LEVELS

It would be desirable to measure central nervous system (CNS) antipsychotic levels in human beings. Although this is possible with emerging imaging techniques, expense and technological barriers currently prohibit their routine clinical use. Direct measurements can also be made on postmortem tissue of patients who had recently discontinued the drug or were on antipsychotics at the time of death (22). HPDL is concentrated
in the brain at levels about 10 to 30 times higher than the plasma level. Animal studies find a brain-to-plasma ratio of about 20:1. Limited information from animal studies after drug withdrawal also shows a more rapid disappearance from plasma than from the brain. Although the half-life of HPDL is about 24 hours in plasma, even several days after cessation of oral dosing, significant CNS D2 receptor activity is still evident. Indeed, PET studies indicate that after withdrawal of HPDL or fluphenazine decanoate, D2 blockade can persist for several months. Thus, it is likely that human brain levels are approximately 10 to 20 times higher than plasma levels and that these agents stay in the brain longer than in plasma.








TABLE 5-3 PHARMACOKINETICS OF SECOND-GENERATION ANTIPSYCHOTICS






























































































Clozapine


Risperidone


Olanzapine


Quetiapine


Ziprasidone


Aripiprazole


Paliperidone


lloperidone


Asenapine


Absorption


Well absorbed


Rapid and complete


Well absorbed


Rapid


Rapid with food


Not affected by food


Well absorbed


Well absorbed


Well absorbed


Time to peak concentration


1-6 hours


1-3 hours


5 hours


About 1.5 hours


3.8-5.2 hours


3 hours


24 hours


2-4 hours


0.5-1.5 hours


Serum elimination half-life


12 hours


About


20 hours


20-70 hours


3-7 hours


5-10 hours


75 hours


23 hours


18 hours


24 hours


Time to Css


3-4 days


5-7 days


7 days


2-3 days


1-3 days


14 days


4-5 days


3-4 days


3 days


Protein binding


97%


89%; 77% for metabolite


93%


80%


>99%


98%


74%


95%


95%


Metabolism


CYP 1A2, 3A4


CYP 2D6


CYP 1A2, 2D6


Primarily by CYP 3A4


CYP 3A4


CYP 2D6, 3A4


Not clinically relevant


CYP 2D6, 3A4


UGT 1A4


CYP 1A2


Active metabolite


Norclozapine


9-OH-RISP (paliperidone)


None


None


None


Dehydro-ARIP


None


P 88


Activity related to parent drug


CYP, cytochrome P450; RISP, risperidone; ARIP, aripiprazole.




PLASMA LEVEL STUDY DESIGNS

Although large interindividual variability in steady state plasma concentrations among patients treated with similar doses of a given antipsychotic is well established, the existence of a critical range of plasma concentrations for therapeutic response or significant adverse effects remains controversial. A body of data from a number of fixed-dose studies, however, indicates a possible threshold for response or a linear or curvilinear relationship between plasma levels and clinical response for agents such as


Data comparing plasma concentrations with clinical response for ZPD and more recent SGAs are not currently available (45).

In addition, prospective studies in large numbers of acutely-ill patients targeting certain plasma levels to test a putative therapeutic threshold or range have been conducted with HPDL (46).


Chlorpromazine

Curry et al. (47,48) found a wide range of effective plasma drug levels in schizophrenia patients treated with comparable doses of CPZ, establishing that an upward or downward shift of 50% in dose usually produced adverse effects or a psychotic exacerbation, respectively. May et al. (49) found no relationship between plasma levels and response in 48 patients on fixed doses of CPZ (6.6 mg/kg/day). Effective plasma drug levels in this agent are particularly difficult to evaluate, however, because of its many potentially confounding metabolites, which are typically not measured.


Fluphenazine

Several studies investigated fluphenazine plasma levels with either the oral or the parenteral long-acting (LA) form. In one, clinical response as a function of the mean Css of oral fluphenazine suggested a therapeutic upper end based on three nonresponding patients who had a mean Css above 2.8 ng/mL (24). Further, a low end was suggested by two nonresponders and one partial responder, whose levels were below 0.2 ng/mL. Van Putten et al. (26) found that higher fluphenazine plasma levels (up to 4.23 ng/mL) were significantly associated with a higher rate of improvement; however, 90% (65 of 72 patients) experienced disabling adverse effects with levels greater than 2.7 ng/mL. In a 2-year, double-blind comparison of 5 or 25 mg of fluphenazine decanoate, Marder et al. (50) found a significant relationship between fluphenazine plasma levels and psychotic exacerbations after 6 to 9 months of maintenance therapy. Thus, those with levels less than 0.5 ng/mL did much worse than those with levels more than 1.0 ng/mL. These data suggest that the ideal plasma level range for many patients may be between 1.0 and 2.8 ng/mL. However, in a later study, Marder et al. (51) attempted to randomize patients into different plasma concentration ranges of fluphenazine. The results failed to support this strategy.


Trifluoperazine

We reported on a potential therapeutic window with the commonly used phenothiazine trifluoperazine (27). Specifically, there was evidence for a lower therapeutic threshold, around 1 ng/mL, and a suggestion of an upper end around 2.3 ng/mL (Fig. 5-3). Based on a review of dose-response studies, we concluded
that 9 to 15 mg of trifluoperazine would be comparable to 300 mg of CPZ and should fall near the lower part of the linear portion of the dose-response curve.






Figure 5-3 Brief Psychiatric Rating Scale (BPRS) change scores in relationship to trifluoperazine plasma levels. (From Janicak PG, Javaid JI, Sharma RP, et al. Trifluoperazine plasma levels and clinical response. J Clin Psychopharmacol. 1989;9:340-346, with permission.)


Thiothixene

Yesavage et al. (52) treated 48 acute schizophrenia patients with thiothixene (80 mg/day), measuring serum and red blood cell (RBC) concentrations 2 hours after the morning dose. Serum levels ranged from 3 to 45 ng/mL, with a linear relationship between clinical response during the first week of treatment and serum (r = 0.5) and RBC levels (r = 0.64). By contrast, Mavroidis et al. (28) found a curvilinear relationship between thiothixene plasma levels and clinical response. Thus, levels ranging from 2.0 to 15 ng/mL, measured 10 to 12 hours after the last dose, were associated with clinical improvement.


Haloperidol

HPDL, the most commonly prescribed FGA, has only one pharmacologically active metabolite (i.e., reduced HPDL). We reviewed the literature on HPDL plasma levels and summarized the outcome (53). Although there are several studies examining the relationship between HPDL steady state plasma levels and clinical response, they have used varying methodologies in terms of patient selection, symptom profile, diagnostic criteria, and assay techniques as well as variable or fixed-dose schedules. Hence, the results of these studies are difficult to interpret collectively. As with other agents, the results with the fixed doses of HPDL are conflicting, although at least six early studies demonstrated a curvilinear relationship (i.e., therapeutic window) between HPDL plasma levels and clinical response. Although optimal levels differed slightly among these studies, the mean low end was 4.2 ng/mL and the mean high end was 16.8 ng/mL (Table 5-4). By contrast, some studies did not find a correlation between plasma HPDL levels and clinical response (54).

Bleeker et al. (55) and Shostak et al. (56) also attempted to study the low end of a putative HPDL plasma level therapeutic window and found that none of three and two of five patients with low plasma levels, respectively, were responders. This contrasted with their outcome in the middle range, where 5 of 23 and 5 of 6, respectively, responded. Conversely, five other studies deliberately probed the upper end of the therapeutic window. Doddi et al. (57), Kirch et al. (58), and Bigelow et al. (59), using medium or high HPDL doses, failed to find any evidence for an upper end of the therapeutic window in newly admitted patients. Rimon et al. (60) used extremely high HPDL doses (120 mg/day) in very chronic patients but also failed to find an upper end of the therapeutic window. Coryell et al. (61) prospectively assigned patients to fixed HPDL doses for at least the first 2 weeks of
treatment to yield a distribution of plasma levels above and below a hypothesized therapeutic level of 18 ng/mL. They found a significant negative linear relationship (i.e., as dose increased, response decreased) between HPDL plasma levels and outcome at week 1, which did not persist into weeks 2, 3, and 4.








TABLE 5-4 EARLY FIXED-DOSE STUDIES FINDING A CURVILINEAR RELATIONSHIP BETWEEN HALOPERIDOL PLASMA LEVELS AND CLINICAL RESPONSE

















































Study


Assay Method


Therapeutic Range


Rating Scale


Study Duration


Garver (1984)/Mavroidis (1983)


GLC


4-11ng/mL


NHSI


14 days


Smith (1984)


GLC (RRA)


7-17 ng/mL


BPRS psychosis factor


24 days


Potkin (1985)


RIA


4-26 ng/mL


CGI


6 weeks


Van Putten (1985)


RIA


5-16 ng/mL


BPRS


7 days


Van Putten (1988)


RIA


2-12 ng/mL


BPRS psychosis factor


4 weeks


Santos (1989)


RIA


12-35.5 ng/mL (7.4-24.9 in subchronic group)


BPRS total score


21 days


BPRS, Brief Psychiatric Rating Scale; CGI, Clinical Global Impression; GLC, gas liquid chromatographic; NHSI, New Haven Schizophrenic Index; RIA, Radioimmunoassay; RRA, radioreceptor assay.


Adapted from Janicak PG, Javaid JI, Davis JM. Neuroleptic plasma levels: methodological issues, study design, and clinical applicability. In: Marder SR, Davis JM, Janicak PG, eds. Clinical Use of Neuroleptic Plasma Levels. Washington, DC: APPI Press; 1993:17-44.



TARGETED PLASMA LEVEL DESIGNS

One can pool data from several fixed-dose studies to achieve an adequate sample size that would define the optimal cutoff point for a lower end and possibly for an upper end as well. These data can then be used to design a targeted plasma level study.

Using such a design, Volavka et al. (62) failed to find evidence for a therapeutic window after randomly assigning 111 schizophrenia or schizoaffective patients to one of three HPDL plasma levels (i.e., 2 to 13, 13.1 to 24, or 24.1 to 35 ng/mL). However, too high a level for the low and perhaps middle ranges and a prolonged period to titrate to the middle and high HPDL plasma levels may have complicated interpretation of their results.

In a subsequent report, Volavka et al. (54) targeted acutely psychotic patients to a low (mean, 2.2 ng/mL) or a middle (mean, 10.5 ng/mL) HPDL plasma level group. Twenty-five patients with levels less than 3 ng/mL showed minimal response over 3 weeks, indicating that patients would benefit if titrated to higher plasma levels. There was no indication of diminished responding at higher levels.

Janicak et al. (63) attempted to address these issues by prospectively reassigning initial HPDL nonresponders to the putative therapeutic range (Fig. 5-4). During phase A, 25% to 30% of patients in the low, middle, and high HPDL plasma levels responded. Further, in the second phase of this study, initial partial or nonresponders to low or high plasma levels benefited from a dose adjustment (i.e., received an average dose of 25 mg/day) to achieve plasma levels of about 12 ng/mL (Fig. 5-5). When they analyzed only the phase A nonresponders in phase B, the sample size was smaller; but those in the middle range demonstrated significantly more improvement on both the total Brief Psychiatric Rating Scale (BPRS) and the positive symptom subscale scores in contrast to those who remained in the low or the high groups. Further, no benefit was achieved by raising plasma levels beyond the defined middle range.

In the two Case Examples, discussed, patients demonstrated clinically relevant and statistically significant decreases in psychotic symptoms after their plasma HPDL concentrations were targeted to within the 10 to 15 ng/mL range. Although it is possible that they improved during the second phase solely because of time on treatment, their scores on the BPRS dropped rapidly once switched into the middle range. In addition, they demonstrated minimal improvement during the first 3 weeks.

Further, scores on the Simpson-Angus side effect scale were in the mild range for both patients and did not change during either phase. Thus, this would not support decrease in toxicity
as the reason for improvement, especially when the dose and blood levels were reduced in the first patient.






Figure 5-4 Haloperidol (HPDL) plasma level ranges by targeted groups. (Adapted from Janicak PG, Javaid JI, Sharma RP, et al. A two-phase, double-blind randomized study of three haloperidol blood levels for acute psychosis with reassignment of initial non-responders. Acta Psychiatr Scand. 1997;95:343-350.)

As part of this study, a dose-prediction formula was developed to more rapidly achieve the desired HPDL Css. For this purpose, the first 28 patients, before receiving the initial assigned dose for achieving their targeted level, received a 15-mg “test” dose (orally) of HPDL, and blood samples were drawn 24 and 48 hours afterward. Data analysis indicated a strong linear relationship between the targeted log Css achieved and the dose required when the 24-hour log-transformed plasma level was included in a linear regression model (r = 0.933) (63). This formula was then prospectively tested and found to be valid in determining the dose required to achieve the desired HPDL Css (64).

The threshold data generated by McEvoy et al. (65) support the phase A findings of this study. This group determined what they called the NT dose (i.e., 3.7 ± 2.3 mg/day) for 106 schizophrenic and schizoaffective patients by starting with a low dose and increasing it until mild rigidity developed. The phase A results of the previous study were quite consistent with these data in that plasma levels as low as 2 ng/mL (mean dose, 3.3 mg/day) did not clearly fall below a therapeutic threshold for response. After a 2-week open trial at this dose, McEvoy et al. then randomly assigned the 95 remaining patients under double-blind conditions to remain at the NT dose or to be switched to a higher dose of HPDL (mean, 11.6 ± 4.7 mg/day) for an additional 2 weeks. Although there were similar rates of improvement on the total BPRS in phase A with McEvoy et al. at comparable
time points, the Janicak et al. (63) study found a greater improvement in phase B (8.1 BPRS points) compared with McEvoy et al.’s second phase (2.9 BPRS change score points). In the McEvoy study, although higher doses of HPDL did not produce greater improvement in measures of psychosis, they did lead to slightly more improvement in hostility.






Figure 5-5 Clinical response based on plasma leve l assignment in Phase B. (From Janicak PG, Javaid JI, Sharma RP, et al. A two-phase, double-blind randomized study of three haloperidol plasma levels for acute psychosis with reassignment of initial non-responders. Acta Psychiatr Scand. 1997;95:343-350, with permission.)


These results also complement a series of PET studies by Nordstrom et al. (66) and Nyberg et al. (67), which support a potential optimal striatal D2 occupancy level. Thus, below 60% D2 occupancy response may be suboptimal and above 80% D2 occupancy EPS may increase. In addition, Wolkin et al. (68) found that HPDL plasma levels of 5 to 15 ng/mL led to rapid increase in striatal D2 occupancy, up to 80%. In a sample of seven acutely psychotic, first-episode patients, Kapur et al. (69), in a prospective controlled trial, found that 2 mg of HPDL for 2 weeks produced a 66% ± 7% D2 receptor occupancy level. Patients achieved HPDL plasma concentrations ranging from 0.6 to 1.5 ng/mL (κ2 = 1.1 ± 0.4). At these levels, five patients showed “much improvement” per the Clinical Global Impression (CGI), including a 45% improvement in positive symptoms and a 55% improvement in negative symptoms.

Although some studies using higher plasma levels find a modest decrement in clinical effect and others find a response plateau, none finds evidence that higher plasma levels lead to enhanced efficacy. In addition, though not a marked phenomenon, with unnecessarily high plasma levels (or doses), there is an increased risk of adverse events (AEs). For example, there is evidence that rapid dose escalation may predispose to neuroleptic malignant syndrome (NMS) (70). In this context, we emphasize the consistent agreement among studies that there is no
additional benefit to using higher doses or plasma levels.



Clozapine

Clozapine Plasma Level-Drug Interactions. Clozapine is principally metabolized to N-desmethylclozapine (norclozapine). It is also metabolized to N-oxide, other hydroxyl metabolites, and a protein-reactive metabolite. The N-oxide can be converted back to clozapine. The enzyme responsible for the metabolism of clozapine to norclozapine is cytochrome P450 (CYP) 1A2 (71). This is consistent with a study showing that caffeine, a marker for 1A2, is cleared in relationship to the conversion of clozapine to norclozapine (72). Discontinuation of coffee intake can decrease clozapine plasma levels by more than 50%, and increasing caffeine intake can produce a reemergence of adverse effects (e.g., drowsiness, excess salivation). In addition, smoking, which induces 1A2, lowers clozapine plasma levels. Fluvoxamine, an inhibitor of 1A2, dramatically increases plasma levels, and on occasion, adverse effects are seen (73). This phenomenon can lead to clozapine intoxication in patients on high doses of fluvoxamine.

Phenytoin, an enzyme inducer, can reduce clozapine plasma levels. There have been two reported cases of RISP causing an increase in clozapine plasma levels; however, because this agent is not an inhibitor of 1A2, the mechanisms for this increase are unclear.

Clozapine Dose-Response Relationship. Data exits on dose-response curves for clozapine. In a blinded, controlled study, Simpson et al. (74) randomized patients to three doses of clozapine: 100, 300, and 600 mg. These researchers found that 600 mg was more effective than 300 mg and that doses up to 400 mg/day usually produce inadequate plasma levels. Thus, the results of this dose-response study are consistent with the plasma level studies. This provides some evidence that therapeutic drug monitoring (TDM) may help ensure that patients receive a clozapine dose high enough to reach an adequate plasma level.

Clozapine Plasma Level-Response Relationship. Dose-response and plasma level-response information is conceptually very similar. In dose-response terms, it is important to know the average therapeutic threshold dose, the minimal dose to produce a full response. When plasma levels correlate with clinical response and there is a wide range of plasma levels for a given dose, then it is important to know the minimal therapeutic plasma levels to produce the optimal response for most patients. Several reports have indicated that plasma concentrations of clozapine above 350 to 450 ng/mL may be required to maximize response in treatment-refractory schizophrenic patients (34,35,75). Clozapine plasma levels, however, vary substantially from patient to patient. For example, Potkin et al. (35) found a greater than 20-fold (40 to 911 mg/mL) range in plasma levels after a fixed dose of 400 mg/day. Initially, Perry et al. (34) did a plasma level study assigning patients to a dose of 400 mg/day for 4 weeks and
identified 350 ng/mL as a threshold for response. At or above this plasma level, 64% of patients responded as compared with only 23% of patients below that level. This group followed up these original results with a larger sample, and in a follow-up of the existing samples, five of seven patients had an unsatisfactory response to a dose of 400 mg/day. When the dose was increased to achieve a plasma level above the threshold 350 ng/mL, however, most then responded (76).

Potkin et al. (35) also used a fixed dose of 400 mg/day in a study of 58 schizophrenic patients and found that very few (8%) patients with low clozapine plasma levels (<420 ng/mL) responded, in comparison with 60% of those with plasma levels greater than 420 ng/mL. This group used a targeted plasma level design so that patients with originally low plasma levels had these levels increased above 420 ng/mL in a double-blind, random-assignment procedure. Response rate increased to 73%, compared with 29% of those whose plasma levels remained low. This result is particularly impressive because it was a randomized prospective change in dose, which provides important confirmation of the relationship between clozapine plasma levels and clinical efficacy. Kronig et al. (75) also found a therapeutic level of 350 ng/mL. Although the chosen level varies from study to study, there is general agreement that a threshold therapeutic plasma level exists somewhere between 350 and 450 ng/mL.

It is important to consider whether parent compound levels only or the sum of clozapine plus norclozapine plasma levels are measured because in the latter case, the threshold would be higher. Note that a quality control study has been done comparing different laboratories on the assay reliability of clozapine plasma levels, with split samples finding good agreement. Clinically, it is important to start with a small dose and gradually increase to avoid adverse effects. Even so, it is desirable to move rapidly to the correct level in patients who may need a slightly higher than average dose. Perry (78) has provided a prediction formula for plasma concentration that requires information about the patient’s clozapine dose, smoking status, and gender.



Risperidone

Riedel et al. (38) measured RISP and 9-OHRISP levels in patients receiving open-label treatment. Those with higher levels were less likely to be clinical responders. Although plasma concentrations and EPS were not correlated, a high concentration at week 2 predicted the development of EPS. The lack of a clear relationship between levels and response could be related to clinicians adjusting the dose to manage EPS. The poor response of patients with higher plasma levels could be due to mild EPS, which went unrated but may have compromised the overall clinical outcome. Another trial (39) did find a relationship between RISP plasma levels and EPS but not clinical response.



Olanzapine

Perry et al. (42) measured OLZ concentrations in patients who participated in a study of three OLZ dose ranges. Receiver-operated curve (ROC) analyses indicated that a level greater than 23.2 ng/mL was a useful predictor of response. Thus, among patients with levels above 23.2 ng/mL, 52% met response criteria, whereas only 25% of patients with lower levels met these criteria.



Antipsychotics for Acute Psychosis


ACUTE SCHIZOPHRENIA

Each class of antipsychotic was found superior to placebo for the treatment of acute schizophrenia. Some of the most informative studies regarding the effectiveness of these agents were carried out during the first years following their discovery, when there was considerable skepticism regarding their usefulness. For example, a classic, double-blind, random-assignment investigation by Cole et al. (79) compared the response of schizophrenia patients treated with antipsychotics with those treated with placebo. In a reexamination of the data (80), the authors evaluated the fate of the 10% to 20% patients who had the best outcome. A small percentage recovered completely without residual symptoms in 6 weeks, representing about 16% of those on an active drug but only about 1% of those on placebo. This is a remarkable difference, and although some did relatively well without medication, it is likely that they would have done even better with active drug therapy. Further, in the drug-treated group, only 2% deteriorated, in contrast to 33% in the placebo group. It is true that some patients showed little improvement with antipsychotics, but if they had been given only placebo, these patients most likely would have deteriorated even more. Thus, at both ends of the prognostic spectrum, there appears to be a substantial benefit from drugs. In some of the studies, the dose was too low to produce an effect, but when adequate these agents were consistently superior to placebo. The magnitude of improvement in the drug-treated group was considerable whether evaluated by the degree of change (e.g., worse, no change, slight improvement, marked improvement) or the degree of remission (e.g., full remission, only minimal symptoms, still mildly ill, moderately ill, or severely ill). Results from this National Institute of Mental Health (NIMH) Collaborative Study, combining both types of evaluation, are presented in Table 5-5 (79,80). These results can be summarized as follows:








TABLE 5-5 EFFICACY OF ANTIPSYCHOTICS








































Degree of Remission


Degree of Change


Drug (%)


Placebo (%)


Remitted


Very much improved


16


1


Only borderline symptoms remain


Improved


29


11


Mild symptoms still present


Improved


16


10


Moderately ill


Slightly improved


31


31


Moderately ill


Not improved


6


15


Severely ill


Worse


2


33


From Cole JO, Davis JM. Antipsychotic drugs. In: Bellak L, Loeb L, eds. The Schizophrenic Syndrome. New York, NY: Grune & Stratton; 1969:478-568, with permission.




  • 16% of the drug-treated versus only 1% of the placebo-treated group were in complete remission or very much improved



  • 29% of the drug-treated and 11% of the placebo-treated group were evaluated as improved, with only borderline symptoms


  • 16% of the drug-treated and 10% of the placebo-treated group were evaluated as improved, with mild symptoms remaining


  • 8% of patients did poorly on active drug (i.e., rated as not improved or still ill), compared with 48% on placebo


  • Only 2% of those on an antipsychotic versus 33% of the placebo patients deteriorated during 6 weeks of treatment


  • The largest differences between the drug- and the placebo-treated patients are seen at both ends of the outcome spectrum

This study also found that new schizophrenia symptoms often emerged during the 6 weeks of placebo treatment, whereas worsening of symptoms was prevented by antipsychotics. Lehmann (81) coined the term psychostatic to describe the ability of phenothiazine antipsychotics to prevent the reemergence of symptoms.

The time course of improvement for most acutely psychotic patients on these agents demonstrates that most therapeutic gain occurs during the first 6 weeks, although further progress can be realized much later (e.g., clozapine). Thus, some patients improve rapidly, within a few days, whereas others show gradual changes over several months. There is no evidence for clinical tolerance, for if there were, the dose would have to be increased over time. Fixed-dose studies, however, show that the initial effective amount of drug does not lose efficacy. In addition, patients do not require higher doses after several weeks of treatment; instead, the reverse appears to be true. It is also clear that antipsychotics do not produce dependency of the barbiturate, stimulant, or narcotic type.

In the hope of developing either a more effective agent or one with fewer adverse effects, medicinal chemists have synthesized a number of new compounds. Preclinical animal model studies are used to select potentially useful drugs, using a profile of pharmacological properties reflecting differential blockade of DA as well as other neurotransmitter systems (e.g., 5-HT). This research has led to the development of several new classes, as noted earlier in this chapter.

The question immediately arises as to whether any of the drugs in these various classes are superior to CPZ for the typical patient, for particular symptoms, or for subgroups of patients. With the exception of promazine and mepazine, all antipsychotics are clearly superior to placebo or nonspecific sedatives. Comparisons with CPZ in controlled trials found mepazine and promazine inferior to CPZ, but all other agents were equal to CPZ in therapeutic efficacy. Other controlled trials using thioridazine or trifluoperazine as standards also found all other FGAs to be comparable with these agents. Although there were trends at times favoring one agent over another, they were usually not statistically significant, and an inspection of the data finds no agent to be consistently superior to any other. Furthermore, all these drugs produced consistent changes in the same symptoms. These similarities are quite striking and support the theory that they work through a common mechanism of action (e.g., DA blockade). Furthermore, relevant differences are primarily related to their adverse effect profiles. Pimozide is FDA labeled for Tourette syndrome and is particularly interesting in that it is a highly specific DA antagonist that may produce fewer adverse effects than HPDL. In open studies with adequate doses, this agent has demonstrated efficacy for acute schizophrenia. Several double-blind trials comparing pimozide with other FGAs also found it to be an equally effective maintenance therapy (82,83 and 84). We consider this agent to be as effective as the other standard agents, with the same, potential for adverse effects.

When the SGAs were first introduced in the early 1990s, there was hope that these agents would prove to be more effective than FGAs for the acute symptoms of schizophrenia. This hope was encouraged by comparisons of clozapine with FGAs, which found clozapine more effective (85,86). However, there was an important source of bias in these studies. Among the patients included were those who could not be adequately treated with FGAs because they failed to improve on them or because they could not tolerate them. These studies clearly identified clozapine as a valuable alternative for those patients who failed on FGAs, but they did not address whether it had an advantage for the broad group of patients who could be treated with FGAs.

Support for an advantage for SGAs came from early studies of RISP and OLZ (reviewed in Leucht et al. (87). These were Phase II and III
studies that compared these SGAs with both a placebo and an FGA, usually HPDL. All of these early studies found that the SGA was superior to placebo, and some found that the SGA was also more effective than HPDL for psychotic symptoms. However, a broader look at these and studies of other SGAs raised questions about their advantages. A meta-analysis by Geddes et al. (88) concluded that the advantages of SGAs were apparent only when patients received excessive doses of the comparator FGA. More specifically, studies that used doses of HPDL greater than 12 mg daily were more likely to find advantages for the SGAs. This suggests that the greater severity of extrapyramidal symptoms in the HPDL-treated groups may have given the SGAs an apparent advantage, though it was not related to their intrinsic antipsychotic properties. In addition, another meta-analysis (87) indicated that even when there were statistically significant advantages for the newer drug over HPDL, the effect sizes were very small. This finding is disputed in a meta-analysis by Davis et al. (89) who found substantial advantages for clozapine, amisulpride, RISP, and OLZ when compared with HPDL. These advantages were not present with other SGAs. Cochrane reviews of RISP (90) and OLZ (91), however, point out design flaws such as high dropout rates that make it difficult to determine whether these SGAs are superior to FGAs.

Results of the first phase of the Clinical Antipsychotic Trial of Intervention Effectiveness (CATIE) provide useful information regarding the relative effectiveness of various antipsychotics (92). This was a double-blind, randomized, NIMH-sponsored trial conducted at 57 US sites in which 1,493 non-treatment-resistant patients with schizophrenia were randomized to perphenazine (8 to 32 mg/day), OLZ (7.5 to 30 mg/day), QTP (200 to 800 mg/day), RISP (1.5 to 6 mg/day), or ZPD (40 to 160 mg/day). Concomitant psychotropics other than another antipsychotic were allowed. The primary outcome measure was the time that patients remained on their assigned drug. Seventy-four percent of subjects discontinued treatment during an 18-month period. Rates of discontinuation were 64% for OLZ, 75% for perphenazine, 82% for QTP, 74% for RISP, and 79% for ZPD. This study (like others) indicates that FGAs and SGAs are similar in efficacy. There was a statistically better outcome with OLZ in that



  • Time to discontinuation for any cause was longer for OLZ (but this was nonsignificant for perphenazine and ZPD after adjustment for multiple comparisons)


  • Time to discontinuation for lack of efficacy was longer for OLZ (but this was nonsignificant for ZPD after adjustment for multiple comparisons)


  • Duration of successful treatment was significantly longer for OLZ

The mean modal doses of some of the antipsychotics were problematic in that they were lower than those typically used in clinical practice (e.g., QTP = 543 mg/day; ZPD = 113 mg/day; RISP = 3.9 mg/day). Monitoring of important AEs found that



  • Time to discontinuation for intolerable side effects did not differ, but rates were highest in the OLZ group and lowest in the RISP group


  • Metabolic issues were highest with OLZ, while ZPD was the only agent associated with improvement in metabolic variables


  • EPSs were highest with perphenazine


  • Hyperprolactinemia only occurred with RISP


  • Clinically relevant changes in the QTc interval did not occur with any agent

The interpretation of the results from the first phase of CATIE, however, is complex. Although OLZ had an advantage in efficacy, it also caused greater increases in weight, glucose levels, and lipids. Patients who received perphenazine were more likely to discontinue their medication due to EPS. The study was too brief to evaluate the relative risk of TD among the agents, and for ethical reasons, those with a history of TD could not be randomized to perphenazine. In addition, the patients who entered the study were probably not representative of typical patients who receive treatment in clinical settings. On average, patients had been ill for more than 15 years, had received multiple trials with SGAs, but still had not found a completely satisfactory agent. This suggests that the study was focused on patients who were partial responders to antipsychotics— a group likely to have higher rates of discontinuation. Nevertheless, the large sample size of this study and the fact that it was supported by NIMH rather than industry give the results considerable weight. The results also indicate that all currently available antipsychotics have serious limitations for many patients.


A study from the United Kingdom, Cost Utility of the Latest Antipsychotic Drugs in Schizophrenia Study (CUtLASS), also compared first- and second-generation antipsychotics (93). In this study, 227 patients with similar demographic characteristics to CATIE patients were randomized to a first- or secondgeneration antipsychotic. In contrast to CATIE, they were not assigned to a particular drug, but to a first- or second-generation agent selected by the subject’s psychiatrist. The study failed to find a difference between FGAs and SGAs. However, the findings are difficult to interpret since the most commonly chosen FGA in the study was sulpiride, an agent that is unfamiliar to US psychiatrists and one that is viewed as having less EPS than other FGAs.

Taken together, these large studies do not point to an efficacy advantage for the SGAs. On the other hand, many psychiatrists prefer the newer drugs because of their lower risk of EPS.


SPECTRUM OF ACTIVITY FOR ANTIPSYCHOTICS


Schizophrenia Versus Other Psychosis

Antipsychotics are effective for virtually every condition in which psychosis can be a symptom, including major depression, bipolar disorder, dementias, and psychoses secondary to medical conditions. In each of these illnesses, antipsychotics do not specifically treat all aspects of the underlying illness. Rather, these agents appear to target the psychotic manifestations of the illness.


Effects on Positive, Negative, and Neurocognitive Symptoms in Schizophrenia

Contemporary models for understanding schizophrenia have proposed that the illness includes a number of psychopathological dimensions. Different models accommodate different dimensions, but all include at least positive, negative, and neurocognitive symptoms (94,95 and 96). These dimensions are core features of the illness and are relatively independent of one another. Agitation/excitement and depression are often included as well (Table 5-6). If antipsychotics could be considered truly “antischizophrenic,” it would be expected that they would effectively improve each of these dimensions. However, treatment of an acute psychotic episode typically leads to improvement in positive symptoms such as hallucinations and delusions. As patients improve and enter remission, it is common for positive symptoms to be substantially reduced or eliminated and for negative (e.g., restricted affect, alogia, asociality, avolition, anhedonia) and neurocognitive symptoms to remain. This suggests that antipsychotics act primarily on the positive symptom component and that improvements in other dimensions are secondary.








TABLE 5-6 SCHIZOPHRENIA: SYMPTOM DIMENSIONS




















































Positive symptoms



Hallucinations



Delusions


Neurocognitive symptoms



Dissociative thinking



Disorganization of thought



Attentional impairment



Memory impairment


Negative symptoms (i.e., deficit syndrome)



Restricted affect



Alogia



Asociality



Apathy



Anhedonia


Excitement/agitation


Mood symptoms



Dysphoria



Depression


Impairments in neurocognition, including memory, attention, executive functioning, and psychomotor performance (97), are a core feature of schizophrenia. Unlike positive symptoms, which tend to be episodic, cognitive impairments are usually present at the onset of the illness and remain relatively stable over the course of the patient’s life. These impairments tend to be an independent dimension of the illness with little relationship to positive symptoms. The importance of neurocognitive symptoms relates to the ability of patients with schizophrenia to function in the community. That is, the relationship between functional outcomes—which include the ability to function vocationally and socially—and neurocognitive impairments is stronger than the relationship between functional outcomes and positive symptoms.

Both FGAs and SGAs may improve cognitive symptoms in psychotic individuals (98),
although their effects are relatively weak compared with the severity of the impairments. Although a number of studies suggest that SGAs are superior to FGAs in improving cognition (99), patients treated with the newer agents usually still retain these impairments. For example, it is estimated that individuals with schizophrenia perform one and one half to two standard deviations below the mean on neuropsychological tests, and SGAs may improve performance by about one half of a standard deviation (99). Although a number of studies have compared SGAs to one another, it is unclear if any SGA demonstrates advantages over others for neurocognitive impairment (100). The NIMH CATIE study compared the effects of five different antipsychotics on a neurocognitive battery (101). Each of the medications led to relatively small improvements in a composite score (z = 0.12 to 0.26) after 2 months. Moreover, there were no differences among the antipsychotics which included perphenazine, an FGA. Interestingly, at 18 months, there was a suggestion that perphenazine had an advantage when compared with the SGAs. This provides further evidence that SGAs do not have specific effects on neurocognition.

The lack of a substantial effect of available antipsychotics for both cognition and negative symptoms has led to a search for medications that would target these domains. These agents might be ineffective as antipsychotics but would be added as comedications with antipsychotics. A number of experimental drugs targeting cholinergic, glutamatergic, dopaminergic, and other systems are in development and may be effective adjuncts to existing APs (102,103). Although most of these agents focus on improving cognition, there are suggestions that some may be effective for treating negative symptoms as well (104).

Some models of psychopathology also include a component of excitement or agitation. Early impressions following the introduction of the first FGAs focused on the calming they induced and led observers to propose that they were “major tranquilizers.” Antipsychotics can diminish excitation, but they also lead to increased activity in withdrawn psychotic individuals. Therefore, “tranquilizer” is a misnomer. Antipsychotics do not, in any real sense, produce a state of tranquility in normal or psychotic individuals; in fact, normal control subjects often find their effects unpleasant. An appropriate analogy may be aspirin, which reduces an elevated temperature but typically does not alter normal temperature. Another example is insulin, which replaces the absent endogenous supply and restores a diabetic to normal glucostasis.

Dysphoria and depression are also prominent symptoms in schizophrenia. They may overlap with certain negative symptoms, are worsened by some FGAs, and contribute to the high suicide rate in these patients.



DOSING STRATEGIES

The goal of pharmacotherapy for acute schizophrenia is to minimize psychotic symptoms that impair the patient’s ability to function and cause substantial distress. For some patients, drug treatment results in a complete remission of psychotic symptoms. Others continue to experience symptoms that are attenuated. Premature attempts to treat the lingering psychosis by increasing the dose, changing antipsychotics, or adding a second agent may not lead to further improvement and may worsen adverse effects. As with most treatments in medicine, antipsychotics diminish symptoms but may not alter the
underlying disease process. The result is that patients may enjoy a robust antipsychotic effect for positive symptoms but still retain psychopathology in other domains such as negative symptoms or cognitive impairments. Although goals may include eliminating every trace of hallucinations or delusions, even a substantial improvement in these positive symptoms may be associated with reduced suffering, improved functioning, and a better quality of life.

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

Stay updated, free articles. Join our Telegram channel

Aug 27, 2016 | Posted by in PHARMACY | Comments Off on Treatment with Antipsychotics

Full access? Get Clinical Tree

Get Clinical Tree app for offline access