Файл: An.Atlas.of.Schizophrenia.eBook-EEn.pdf

ВУЗ: Не указан

Категория: Не указан

Дисциплина: Не указана

Добавлен: 02.10.2020

Просмотров: 1135

Скачиваний: 6

ВНИМАНИЕ! Если данный файл нарушает Ваши авторские права, то обязательно сообщите нам.
background image

©2002 CRC Press LLC

In early phase II placebo controlled trials,

iloperidone at 8 mg per day was superior to
placebo in improving positive and negative
symptoms with EPS at placebo levels. The most
frequently described side-effects were dizziness,
postural hypotension and nausea

76

.

A large scale multicenter trial comparing

iloperidone 4mg, 8mg and 12mg with haloperidol
15 mg per day and placebo in a total of 621
patients over 42 days has recently been
completed

77

. Initial data indicates clinical respo-

nse similar to haloperidol but with significantly
lower rates of EPS (similar to placebo).

Aripiprazole

Aripiprazole is a quinolinone derivative and differs
from other novel antipsychotics in that rather than
being an antagonist at dopamine receptors, it
appears to be a high affinity partial agonist at
presynaptic D

receptors but exhibits antagonist

effects on postsynaptic D

receptors

78

. It has low

affinity for D

and D

receptors and no appre-

ciable affinity for D

1

-like receptors. Its affinity for

5-HT

2A 

receptors is low and of a similar magni-

tude to that of clozapine.

In phase II clinical studies, aripiprazole is

significantly superior to placebo at improving the
total score of the PANSS

79

. A   recently completed

phase III clinical trial compared aripiprazole 15mg
or 30 mg per day with haloperidol 10 mg per day
and placebo in a total of 414 patients for 28 days.
The preliminary results from this study show that
both doses of aripiprazole are significantly better
than placebo at improving PANSS total score and
equivalent to haloperidol. Aripiprazole at both
doses lead to placebo rates of EPS which is
significantly lower than the rate seen in the
patients treated with haloperidol

80

.

ADVERSE EVENTS OF ATYPICAL
ANTIPSYCHOTICS

The issue of side-effects or adverse events is
closely linked to tolerability and acceptability and
therefore to both compliance and relapse preven-
tion. Often the most debilitating and obvious side-
effects of conventional antipsychotics are motor.

As described above, all of the newer antipsych-
otics demonstrate a lower propensity to cause EPS
at clinically effective doses. There is increasing
evidence that these drugs may additionally
provide benefits for patients who suffer from
akathisia and tardive dyskinesia.

Indeed, it is in the area of tardive dyskinesias

that clozapine appears to have its most marked
effect. Lieberman and associates

81 

reported a 50%

reduction of symptoms over 28 months of
treatment in 43% of patients, and Gerlach and
Peacock

82 

reported a resolution of tardive

dyskinesia in 54% of patients after 5 years of
clozapine treatment. Furthermore, Tamminga and
colleagues

83 

reported a significant difference in

reduction of tardive dyskinesia scores in a
clozapine group versus a haloperidol-treated
group and that this difference began after about 4
months of treatment. There have also been case
reports that switching to clozapine has been
effective in reducing tardive dystonia

84

.

Initial data from the studies quoted above on

the newer antipsychotics indicates that they too
produce very low rates of tardive dyskinesia. The
rate of other side-effects may vary between each of
the newer drugs, although fully adequate comparison
studies remain to be published (

Table 4.4

)

85

.

Negative symptoms

It is claimed that clozapine has an almost unique
action against the negative symptoms of schizo-
phrenia, out of proportion to its effect on positive
symptoms

86

, but the evidence for this is by no

means clear. Tandon and associates

87

found that

the improvement in negative symptoms covaried
with the improvement in positive symptoms, and
Hagger and co-workers

88 

found no improvement

in negative symptoms. A more recent finding is
that in comparison with haloperidol, clozapine
had a significant effect on negative symptoms in
patients with non-deficit schizophrenia, but not in
those with deficit schizophrenia, i.e. those with
enduring negative symptoms

89,90

. It has therefore

been suggested that clozapine’s apparently benefi-
cial effect on negative symptoms may simply be a
reflection of its reduced tendency to cause EPS

91

.

The weight of current evidence suggests that


background image

©2002 CRC Press LLC

clozapine has an excellent effect on the secondary,
but not primary, negative symptoms

92

. All of the

newer antipsychotics appear to have an effect on
secondary negative symptoms. Most of the
available trials have indicated a modest but
significant advantage for all of the newer
medications over conventional antipsychotics in
negative symptom improvement

54,58,87,93

.

Amisulpride at low doses (50–300 mg/day) is

claimed to have a unique effect in patients with
only negative symptoms. In comparison with
placebo, these low doses of amisulpride produced
significant reductions in negative symptom scores
with little change in positive symptom scores

94–96

.

However, in a study utilizing a different design but
similar patient population, low-dose amisulpride
was similar to low-dose haloperidol treatment in
terms of negative symptom improvement

97

.

Cognition

Neurocognitive deficits that are a core feature of
schizophrenia, are apparent at the onset of illness
and may deteriorate during the first few years of
illness. The older antipsychotics have limited

impact on these, although inconsistent long-term
improvements have been noted

98,99

. There has

been increasing interest in the possibility that the
newer antipsychotics may ameliorate these probl-
ems which are linked to poor outcome and future
unemployment. Clozapine may lead to improve-
ments in attention, memory and executive
function over 6–12 months

100

. Risperidone is

claimed to improve frontal function and spatial
working memory compared with halo-
peridol

101,102

. Olanzapine is claimed to improve a

variety of measures of function including psycho-
motor speed, verbal fluency and memory

103

. It has

recently been reported that quetiapine improves
attentional performance to the level of that seen
in a matched control group over 2 months of
treatment

104

. In a study comparing haloperidol 

1 mg and 2 mg and amisulpride 50 mg and 100 mg
with placebo in healthy volunteers,

the

haloperidol-treated groups showed greater cogni-
tive impairment on tasks measuring problem-
solving abilites

105

.

It is still not clear how relevant the modest

improvements or impairments reported in these
studies are to long-term outcome.

Table 4.4  Qualitative comparison of the relative side-effects of the newer medications.

These will be subject to change

over time, as new tolerability data are published and report forms returned. Adapted from reference 85

Typicals

Clozapine

Risperidone

Olanzapine

Quetiapine

Amisulpride

Ziprasidone

Anticholinergic

±

+++

±

+

±

±

±

Orthostatic

hypotension

± to +++

+++

+

±

+

±

±

Prolactin

elevation

++ to +++

0

++

±

±

++

±

QT prolongation

± to +

+

± to +

± to +

± to +

±

± to +

Sedation

+ to +++

+++

+

++

++

±

+

Seizures

±

++ to +++

±

±

±

±

±

Weight gain

± to ++

+++

+ to ++

+++

+ to ++

± to +

±


background image

©2002 CRC Press LLC

Affective symptoms

Patients with schizophrenia are significantly more
likely than the general population to suffer from
other psychiatric disorders such as depression,
and conventional antipsychotics are used to treat
other psychotic disorders outside of schizo-
phrenia

Clozapine has been reported to be effective in

patients with treatment-resistant schizoaffective
or manic illnesses

106

(see reference 107 for

review). In one study, clozapine reduced baseline
mania ratings by more than 50% in 72% of a
group of patients suffering from either mania or
schizoaffective disorder and 32% had a significant
improvement in BPRS scores. The latter finding
was more frequent in the patients with bipolar
disorder and the non-rapid cycling patients

108

. In

depressive disorders, clozapine had a more
equivocal response. Although it was seemingly
effective against depressive symptoms occurring
comorbidly with schizophrenia

109

, there has been

little work showing a particular use for clozapine
in treatment-refractory depression

110,111

.

Conventional antipsychotics may both

improve and contribute towards depressive symp-
toms. Clozapine reduces both depressive features
and suicidality

109

. Risperidone produces signifi-

cantly greater reduction in anxiety/depression
subscales in comparison with haloperidol

52

.

Olanzapine has significant antidepressant effects
in comparison with haloperidol

112

. Amisulpride

(50 mg/day) has been compared with imipramine
(100 mg/day) and placebo in patients with
dysthymia and major depressive disorder. In this
study (

= 219), both imipramine and amisulpride

produced similar and significant improvements on
all rating scales

113

. The implications of this in

patients with schizophrenia have yet to be
elucidated.

THE FUTURE

Despite the advances in schizophrenia pharmaco-
therapy since the early 1950s there are still many
limitations. EPS make use of high doses of the
older typical antipsychotics problematic and often

–0.5

–0.3

–0.1

0

–0.4

–0.2

0.1

<

12 mg 

haloperidol

>

12 mg 

haloperidol

Favors

atypical

Favors

haloperidol

COMPARISON OF ATYPICAL AND TYPICAL ANTIPSYCHOTICS

-0.5

-0.3

-0.1

0

-0.4

-0.2

0.1

<

12 mg 

haloperidol

>

12 mg 

haloperidol

Favors

atypical

Favors

haloperidol

<

<

Figure 4.19 

Results from a meta-analysis of trials comparing newer atypical antipsychotics and typical antipsychotics (mainly

haloperidol). The left graph suggests that the clinical superiority of atypical antipsychotics over typical antipsychotics is lost
if lower doses of typical antipsychotics are used (less than 12 mg per day of haloperidol equivalents). The right-hand graph,
suggests that any superiority in tolerability for the atypical antipsychotics is similarly lost if patients receive lower doses of
typical antipsychotics. These findings and interpretations have been criticized on a number of counts including selection bias
in the trials chosen for the meta-analysis, no similar control for the doses of atypicals used and using the drop-out rates from
clinical trials as a measure of medication tolerability. Figure reproduced with permission from Geddes J, Freemantle N,
Harrison P, Bebbington P. Atypical antipsychotics in the treatment of schizophrenia: systematic overview and meta-regression
analysis. 

Br Med J 

2000;321:1371–6


background image

©2002 CRC Press LLC

unpleasant for those taking them. Lower doses of
‘typicals’ and the newer ‘atypical’ antipsychotics
offer benefits in terms of reductions in EPS, but
the latter medications are not without their own
unpleasant side-effects. In a recent meta-analysis,
Geddes and colleagues

114 

described that in their

analysis of all of the trials of atypical anti-
psychotics versus typical antipsychotics there was
no treatment advantage for the newer medications
over doses of haloperidol below 12 mg/day and no
significant difference in total side-effect load
(

Figure 4.19

). Such meta-analyses have, however,

come under criticism for obscuring real treatment
effects by being overinclusive of available
trials

115,116

. Furthermore, a recent critical review

of treatment trials with low-dose typical anti-
psychotics could find no convincing evidence that
there is a low dose of typical antipsychotics which
is effective but does not produce EPS

117

.

Another failing in the pharmacotherapy of

schizophrenia is that there are still 40–50% of
patients with schizophrenia who do not have an
optimal response to medication, and some 20%
who are resistant to all forms of treatment,
including clozapine.

Future developments may include a new

generation of antipsychotic drugs which are
partial agonists (rather than antagonists) at D

2

and

D

2

-like receptors, such as aripiprazole

78

. As

described above, early clinical trials with this
medication show good efficacy with placebo rates
of EPS

80

.

In conjunction with developments in trial

methodology, combinations of neurochemical and
functional imaging may help to elucidate the
neural correlates of treatment response and
resistance and allow more rationale therapeutic
decisions.

Pharmacogenetics may further help to define

the parameters which predict response or non-
response to particular medications by analyzing
the allelic variations for individual receptors
which correlate with response

118

.

REFERENCES

1.

Carlsson A, Lindqvist M. Effect of chlorpromazine or
haloperidol on formation of 3-methoxytyramine and
normetanephrine in mouse brain. Acta Pharmacol
Toxicol 
1963;20:140–4

2.

Creese I, Burt DR, Snyder SH. Dopamine receptor
binding predicts clinical and pharmacological
potencies of antischizophrenic drugs. Science 1976;
19:481–3

3.

Angrist B, van Kammen DP. CNS stimulants as a tool
in the study of schizophrenia. Trends Neurosci 1984;
7:388–90

4.

Lieberman JA, Kane JM, Alvir J. Provocative tests
with psychostimulant drugs in schizophrenia.
Psychopharmacology 1987;91:415–33

5.

Laruelle M. Imaging dopamine transmission in
schizophrenia. A review and meta-analysis. Q J Nucl
Med 
1998;42:211–21

6.

Kapur S, Zipursky R, Jones C, et al. Relationship
between dopamine D(2) occupancy,

clinical

response, and side effects: a double-blind PET study
of first-episode  schizophrenia. Am J Psychiatry
2000;157:514–20

7.

Farde L, Nordstrom AL. PET analysis indicates
atypical central dopamine receptor occupancy in
clozapine-treated patients. Br J Psychiatry 1992;17
(Suppl.):30-3

8.

Pilowsky LS, Costa DC, Ell PJ, et al. Clozapine, single
photon emission tomography, and the D2 dopamine
receptor blockade hypothesis of schizophrenia.
Lancet 1992;340:199–202

9.

Broich K, Grunwald F, Kasper S, et al. D

2

-dopamine

receptor occupancy measured by IBZM-SPECT in
relation to extrapyramidal side effects. Pharmaco-
psychiatry 
1998;31:159–62

10.

Busatto GF, Kerwin RW. Perspectives on the role of
serotonergic mechanisms in the pharmacology of
schizophrenia. J Psychopharmacol 1997;11:3–12

11.

Laruelle M, Abi-Dargham A, Gil R, et al. Increased
dopamine transmission in schizophrenia: relation-
ship to illness phases. Biol Psychiatry 1999;46:56–72

12.

Woolley DW, Shaw E. A biological and pharmaco-
logical suggestion about certain mental disorders.
Proc Natl Acad Sci USA 1954;40:228–31

13.

Okubo Y, Suhara T, Suzuki K, et al. Serotonin 5-HT

2

receptors in schizophrenic patients studied by
positron emission tomography. Life Sci 2000;66:
2455–64

14.

Verhoeff NP, Meyer JH, Kecojevic A, et al. A voxel-
by-voxel analysis of [18F]setoperone PET data
shows no substantial serotonin 5-HT(2A) receptor
changes in schizophrenia. Psychiatry Res 2000;99:
123–35


background image

©2002 CRC Press LLC

15.

Nordstrom AL, Farde L, Halldin C. High 5-HT

2

receptor occupancy in clozapine treated patients
demonstrated by PET. Psychopharmacology  1993;
110:365–7

16.

Travis MJ, Busatto GF, Pilowsky LS, et al. 5-HT

2A

receptor blockade in patients with schizophrenia
treated with risperidone or clozapine. A SPET study
using the novel 5-HT

2A

ligand 123I-5-I-R-91150. Br

J Psychiatry 1998;173:236–41

17.

Kapur S, Zipursky RB, Remington G. Clinical and
theoretical implications of 5-HT

2

and D

2

receptor

occupancy of clozapine, risperidone, and olanzapine
in schizophrenia. Am J Psychiatry 1999;156:286–93

18.

Travis MJ, Busatto GF, Pilowsky LS, et al. 5-HT2A
receptor occupancy in schizophrenic patients:
Typical versus atypical antipsychotics. J Psycho-
pharmacol 
1998;12(Suppl. A54):215

19.

Benes FM. The role of stress and dopamine-GABA
interactions in the vulnerability for schizophrenia. J
Psychiatr Res 
1997;31:257–75

20.

Wassef AA, Dott SG, Harris A, et al. Critical review
of GABA-ergic drugs in the treatment of schizo-
phrenia. J Clin Psychopharmacol 1999;19:222–32

21.

Krystal JH, Karper LP, Seibyl JP, et al. Subanesthetic
effects of the noncompetitive NMDA antagonist,
ketamine, in humans. Psychotomimetic, perceptual,
cognitive, and neuroendocrine responses. Arch Gen
Psychiatry 
1994;51:199–214

22.

Moghaddam B, Adams B, Verma A, Daly D.
Activation of glutamatergic neurotransmission by
ketamine: a novel step in the pathway from NMDA
receptor blockade to dopaminergic and cognitive
disruptions associated with the prefrontal cortex. J
Neurosci 
1997;17:2921–7

23.

Farber NB, Newcomer JW, Olney JW. The glutamate
synapse in neuropsychiatric disorders. Focus on
schizophrenia and Alzheimer's disease. Prog Brain
Res 
1998;116:421–37

24.

National Institutes of Mental HealthPhenothiazine
treatment in schizophrenia. Arch Gen Psychiatry
1964;10:246–26

25.

Johnstone EC, Crow TJ, Frith CD, Owens DG. The
Northwick Park “functional” psychosis study: diag-
nosis and treatment response. Lancet 1988;2:119–25

26.

Davis JM, Andriukaitis S. The natural course of
schizophrenia and effective maintainence treatment.
J Clin Psychopharmacol 1986;6(Suppl.):2–10

27.

Marder SR, Wirsching WC, van Putten T. Drug
treatment of schizophrenia. Overview of recent
research. Schizoph Res 1991;4:81–90

28.

Caffey EM, Diamond LS, Frank TV, et al.
Discontinuation or reduction of chemotherapy in
chronic schizophrenics. J Chronic Dis 1964;17:
347–58

29.

Hogarty GE, Goldberg SC, Schooler NR, et al. Drug
and sociotherapy in the aftercare of schizophrenic

patients. II: two year relapse rates.

Arch Gen

Psychiatry 1974;31:603–8

30.

Davis JM. Overview: maintenance therapy in
psychiatry – I. Schizophrenia. Am J Psychiatry 1975;
132:1237–45

31.

Gilbert PL, Harris J, McAdams LA, Jeste DV.
Neuroleptic withdrawal in schizophrenic patients.
Arch Gen Psychiatry 1995;52:173–88

32.

Viguera AC, Baldessarini RJ, Hegarty JD, et al.
Clinical Risk following abrupt and gradual
withdrawal of maintenance neuroleptic treatment.
Arch Gen Psychiatry 1997;54:49–55

33.

Schooler NA. Reducing dosage in maintainence
treatment of schizophrenia. Br J Psychiatry 1993;
163(Suppl. 22):58–65

34.

Barbui C, Saraceno B, Liberati A, Garattini S. Low-
dose neuroleptic therapy and relapse in schizo-
phrenia: metaanalysis of randomised controlled
trials. Eur Psychiatry 1996;11:306–13

35.

Herz MI, Glazer WM, Mostert MA, et al.
Intermittent vs maintenance medication in schizo-
phrenia. Two-year results. Arch Gen Psychiatry
1991;48:333–9

36.

Carpenter WT Jr, Hanlon TE, Heinrichs DW,
Summerfelt AT.

Continuous versus targeted

medication in schizophrenic outpatients: outcome
results [erratum appears in Am J Psychiatry 1991;
148:819]. Am J Psychiatry 1990;147:1138–48

37.

Jolley AG, Hirsch SR, Morrison E, et al. Trial of brief
intermittent neuroleptic prophylaxis for selected
schizophrenic outpatients:

clinical and social

outcome at two years. Br Med J 1990;301:837–42

38.

Gaebel W, Frick U, Kopcke W, et al. Early neuroleptic
intervention in schizophrenia:

are prodromal

symptoms valid predictors of relapse? Br J Psychiatry
1993;21(Suppl.):8–12

39.

McCreadie RG, Robertson LJ, Wiles DH. The
Nithsdale schizophrenia surveys. IX: Akathisia,
parkinsonism,

tardive dyskinesia and plasma

neuroleptic levels. Br J Psychiatry 1992;160:793–9

40.

Glazer WM, Morgenstern H, Doucette JT. Predicting
the long-term risk of tardive dyskinesia in out-
patients maintained on neuroleptic medications. J
Clin Psychiatry 
1993;54:133–9

41.

Reilly JG, Ayis SA, Ferrier IN, et al. QTc-interval
abnormalities and psychotropic drug therapy in
psychiatric patients. Lancet 2000;355:1048–52

42.

Yap YG, Camm J. Risk of torsades de pointes with
non-cardiac drugs. Doctors need to be aware that
many drugs can cause qt prolongation [Editorial]. Br
Med J 
2000;320:1158–9

43.

Morganroth J, Brozovich FV, McDonald JT, Jacobs
RA. Variability of the QT measurement in healthy
men, with implications for selection of an abnormal
QT value to predict drug toxicity and proarrhyth-
mia. Am J Cardiol 1991;67:774–6