Hostname: page-component-7c8c6479df-ws8qp Total loading time: 0 Render date: 2024-03-28T04:12:34.069Z Has data issue: false hasContentIssue false

Psychopharmacology and adverse effects of antipsychotic long-acting injections: A review

Published online by Cambridge University Press:  02 January 2018

David Taylor*
Affiliation:
Pharmacy Department, Maudsley Hospital, Denmark Hill, London SE5 8AZ, UK, email: David.taylor@slam.nhs.uk Division of Pharmaceutical Sciences, King's College London, UK
Rights & Permissions [Opens in a new window]

Abstract

Background

Depot antipsychotics are widely used in clinical practice. Long-acting formulations of second-generation antipsychotics are now being developed and introduced.

Aims

To review the pharmacology, pharmacokinetics and adverse effect profiles of currently available antipsychotic long-acting injections (LAIs).

Method

The psychopharmacological properties of first- and second-generation antipsychotic LAIs are reviewed using data available up to October 2008.

Results

First-generation antipsychotic (FGA) LAIs are associated with a high rate of acute and chronic movement disorders. Risperidone LAI is better tolerated in this respect, but is associated with hyperprolactinaemia and weight gain. Olanzapine LAI causes weight gain and other metabolic effects but appears not to be associated with an important incidence of movement disorders.

Conclusions

Dosing of LAIs is complicated by delayed release of drug, changes in plasma levels without change in dose, and by the lack of data establishing clear dose requirements. All LAIs offer the prospect of assured adherence (although patients may still default on treatment) but their use is complicated by adverse effects, complex pharmacokinetics and confusion over dose–response relationships.

Type
Review Articles
Copyright
Copyright © Royal College of Psychiatrists, 2009 

There are currently eight antipsychotic long-acting injections (LAIs) in widespread use around the world. Six of these are LAI preparations of first-generation antipsychotics (FGAs), including flupentixol, fluphenazine, haloperidol, perphenazine, pipotiazine and zuclopenthixol, and the remaining two are LAI formulations of the second-generation antipsychotics (SGAs) risperidone and olanzapine. The dosing characteristics of these LAIs are summarised in Table 1.

Table 1 Pharmacokinetic and pharmacodynamic characteristics of antipsychotic long-acting injections

Time to peak, days Plasma half-life, days Time to steady state, months Test dose, mg Typical clinical dose per 2 weeks, mg Licensed dosing intervals (UK), weeks Comments
First-generation antipsychotics
   Flupentixol decanoate 3–7 ∼17 2 20 60 2–4 Available as low-volume injection
   Fluphenazine decanoate 1 7–14 2 12.5 50 2–5 Available as concentrate
   Haloperidol decanoate 7 ∼21 2–3 Not stated 100 2–4
   Perphenazine decanoate 1–7 ∼14 3 Not stated 150 2–4 Not used in UK or USA
   Pipotiazine palmitate 7–14 ∼14 2 25 50 4
   Zuclopenthixol decanoate 7 ∼14 2 100 300 1–4 Available as concentrate
Second-generation antipsychotics
   Olanzapine pamoate 2–4 14–28 2–3 Not recommended 300 2 or 4 Limited clinical experience as recently introduced
   Risperidone microspheres 28 4–6 2 Not appropriate 37.5 2 Drug release delayed for 2–3 weeks

Pharmacokinetic characteristics

Almost all antipsychotics have short plasma half-lives of 1–2 days at most. This means that antipsychotics have to be administered frequently in order to attain and maintain therapeutic effects. Although the elimination half-lives of these drugs cannot be altered to any important degree, chemical manipulation of the parent compound and changes in pharmaceutical formulation can significantly alter drug release or absorption characteristics. When absorption and/or release are extensively prolonged, effective plasma half-life can be extended (half-life is dependent on release rather than elimination).

Some FGAs possess a terminal alcohol (–OH) group which allows them to be combined with carboxylic acids by a process of esterification. Long-chain esters show high oil solubility and low water solubility. Long-acting injectable formulations of FGAs contain long-chain drug esters (e.g. decanoate, palmitate) dissolved in a vegetable oil. When injected intramuscularly, the oil forms a depot of drug: the drug ester slowly diffuses into the blood stream and is then rapidly hydrolysed to release the parent drug. Reference Barnes and Curson1 Most SGAs lack terminal –OH groups suitable for esterification. Modification of release characteristics is brought about by other means: encapsulating the drug into a biodegradable polymer (risperidone) or injecting a suspension (i.e. water-insoluble particulate matter in water) of drug compound (olanzapine pamoate).

First-generation LAIs

Flupentixol

Flupentixol is a thioxanthine antipsychotic. Flupentixol LAI is formulated as flupentixol decanoate in a low-viscosity vegetable oil (fractionated coconut oil). Flupentixol decanoate provides peak plasma levels 3–7 days after intramuscular injection and shows an apparent half-life of 17 days. Reference Jann, Ereshefsky and Saklad2,Reference Stauning, Kirk and Joorgensen3 Steady-state plasma levels can be expected to be achieved after 2 months or so of regular dosing. In practice, plasma levels may show marked variability independent of dose changes. Reference Tuninger and Levander4

Fluphenazine

Fluphenazine is a piperazine phenothiazine compound. Fluphenazine decanoate is available as an LAI in sesame oil. Plasma levels peak within 24 h of intramuscular injection, Reference Wiles and Gelder5 and the half-life is approximately 7–14 days. Reference Barnes and Curson1,Reference Curry, Whelpton, De Schepper, Vranckx and Schiff6 Plasma levels obtained vary up to 40-fold in patients receiving the same dose. Reference Wiles and Gelder5 Smoking significantly reduces plasma fluphenazine levels. Reference Ereshefsky, Jann, Saklad, Davis, Richards and Burch7

Haloperidol

Haloperidol is a butyrophenone and is available as haloperidol decanoate in sesame oil. Peak plasma levels are seen up to 7 days after intramuscular injection and plasma half-life is around 3 weeks. Reference Barnes and Curson1,Reference Jann, Ereshefsky and Saklad2 Steady-state plasma levels can be expected to be reached after 2–3 months of regular dosing. Reference Kissling, Moller, Walter, Wittmann, Krueger and Trenk8 As with fluphenazine, clearance of haloperidol is significantly increased by smoking. Reference Jann, Saklad, Ereshefsky, Richards, Harrington and Davis9 Nonetheless, variation in plasma levels is smaller in patients receiving haloperidol decanoate than in those receiving oral haloperidol. Reference Nayak, Doose and Nair10

Perphenazine

Perphenazine is a piperazine phenothiazine available as perphenazine decanoate in sesame oil, used mainly in northern Europe and Scandinavia. After intramuscular injection, peak plasma levels are obtained in 1–7 days and the half-life is approximately 2 weeks. Reference Knudsen, Hansen and Larsen11 Steady-state levels are obtained after 3 months. Reference Larsen and Hansen12 Variations in plasma levels during regular dosing are small, Reference Knudsen, Hansen and Larsen11 and plasma levels are directly correlated with dose. Reference Kistrup, Gerlach, Aaes-Jorgensen and Larsen13

Pipotiazine

Pipotiazine is a piperidine phenothiazine antipsychotic. The LAI formulation contains pipotiazine palmitate in coconut oil. This provides peak plasma levels after 1–2 weeks although no drug is released for at least 3 days. Reference Girard, Granier, Schmitt, Cotonat, Escande and Blanc14 Other sources suggest peak levels are seen within 24 h. Reference Barnes and Curson1,Reference Altamura, Sassella, Santini, Montresor, Fumagalli and Mundo15 Plasma half-life is around 2 weeks and the time to steady state is 2 months. Reference Barnes and Curson1,Reference Girard, Granier, Schmitt, Cotonat, Escande and Blanc14,Reference Altamura, Sassella, Santini, Montresor, Fumagalli and Mundo15

Zuclopenthixol

Like flupentixol, zuclopenthixol is a thioxanthine compound. Zuclopenthixol LAI is formulated as the decanoate ester dissolved in thin vegetable oil (fractionated coconut oil). Peak plasma levels of zuclopenthixol are achieved a week after injection. Reference Viala, Hou, Ba, Durand, Dufour and D'Agostino16 Plasma half-life has been estimated at 7.4 days and 19 days. Reference Poulsen, Olesen and Larsen17,Reference Jorgensen and Overo18 Zuclopenthixol LAI shows moderate inter- and intra-individual differences in plasma levels, Reference Viala, Ba, Durand, Gouezo, Hou and Jorgensen19 and marked differences between peak and trough plasma levels when given every 2 weeks (peak levels more than 3 times higher than trough). Reference Poulsen, Olesen and Larsen17 Steady-state plasma levels are achieved after around 2 months of regular dosing. Reference Viala, Ba, Durand, Gouezo, Hou and Jorgensen19

Second-generation LAIs

Olanzapine

Olanzapine LAI is a salt of pamoic acid and olanzapine (olanzapine pamoate) suspended in water. This formulation provides peak plasma levels some 2–4 days after intramuscular administration and a plasma half-life of 2–4 weeks. Reference Kurtz, Bergstrom, McDonnell and Mitchell20 Plasma levels obtained are directly proportional to dose given and time to steady state is around 2–3 months. Reference Kurtz, Bergstrom, McDonnell and Mitchell20 In 2-weekly dosing trough levels are around 50% of peak level; in monthly dosing trough levels are 75% lower than peak. Reference Kurtz, Bergstrom, McDonnell and Mitchell20 Olanzapine pamoate dissociates readily in aqueous environments (e.g. plasma) so its pharmacokinetic properties depend on the method of administration (see below).

Risperidone

As with olanzapine LAI, the formulation of risperidone LAI is materially different from the ester-in-oil preparations of first-generation antipsychotics so far described. Its LAI formulation contains risperidone encapsulated into polymeric microspheres which requires cold storage to retain its intended release characteristics. These microspheres are made up of biodegradable copolymer which is slowly hydrolysed in vivo to release risperidone. Reference Ramstack, Grandolfi, Mannaert, D'Hoore and Lasser21 After intramuscular injection a small amount of risperidone is released from the surface of the microspheres, Reference Ramstack, Grandolfi, Mannaert, D'Hoore and Lasser21 but then further release is delayed for 2–3 weeks, during which time erosion of the microspheres takes place. In 2-weekly dosing peak plasma levels are seen at around 4 weeks after the first injection and the plasma half-life is of the order of 4–6 days. Reference Gefvert, Eriksson, Persson, Helldin, Bjorner and Mannaert22 Steady-state plasma levels are obtained after 8 weeks, at which time peak-to-trough levels vary threefold. Reference Gefvert, Eriksson, Persson, Helldin, Bjorner and Mannaert22

Dose and administration

As with oral antipsychotics, dosing requirements for LAIs in individuals vary enormously. Examination of population data (as below) can provide some guidance on optimum dosing, but individual variation should always be borne in mind and dose adjustments should be according to patient response and tolerability.

Flupentixol decanoate is given initially as a test dose of 20 mg. After 7 days a further dose of 20–40 mg is given. Dosing is then at intervals of 2–4 weeks, at doses ranging from 50 mg every 4 weeks to 400 mg weekly. Dose–response relationships are less than clear. One study showed 40 mg every 2 weeks to be as effective in relapse prevention as 40 mg every 4 weeks. Reference Agrup-Andersson, Bengtsson, Erlandsson, Gottfries and Witzell-Ostlund23 Other trials lend some support to the use of 40 mg every 2 weeks as the optimal dose of this compound. Reference McCreadie, Flanagan, McKnight and Jorgensen24,Reference Johnson, Ludlow, Street and Taylor25 Both studies were double-blind. In one study, 40 mg every 2 weeks was shown to be as effective in improving ‘ward behaviour’ and better tolerated than 200 mg every 2 weeks in female patients over 13 weeks' treatment. Reference McCreadie, Flanagan, McKnight and Jorgensen24 In the other study a 50% dose reduction to 6 mg per week led to substantially increased risk of relapse compared with participants not undergoing dose reduction receiving on average 9 mg per week. Reference Johnson, Ludlow, Street and Taylor25 In contrast, another study in patients stabilised on higher doses showed that reducing the dose of flupentixol below 200 mg every 2 weeks significantly increased the risk of relapse. Reference Cookson26 This study was also a double-blind randomised trial but included only 18 participants.

Fluphenazine decanoate is initiated with a test dose of 12.5 mg. Maintenance doses are in the range 12.5–100 mg given every 2–5 weeks – the maximum dose is thus 100 mg every 2 weeks. Much lower doses appear to be effective – 25 mg every 2 weeks is no more effective than 25 mg every 6 weeks. Reference Carpenter, Buchanan, Kirkpatrick, Lann, Breier and Summerfelt27 There is some support for even lower doses, Reference Geller28 but controlled trials suggest that doses substantially less than 25 mg every 2 weeks greatly increase the risk of relapse. Reference Marder, Van Putten, Mintz, Lebell, McKenzie and May29,Reference Capstick30

For haloperidol decanoate no formal test dose is specified. Treatment is usually begun with a dose of 50 mg and maintenance doses are usually in the range 50–300 mg every 4 weeks (in the USA up to 20 times the previous oral haloperidol dose can be used). Doses of around 100 mg every 4 weeks are probably optimal; Reference Kane, Davis, Schooler, Marder, Casey and Brauzer31,Reference Taylor32 relapse prevention is not improved by higher doses. A dose of 25 mg every 4 weeks is probably ineffective, Reference Kane, Davis, Schooler, Marder, Casey and Brauzer31 whereas doses of around 50 mg every 4 weeks may be effective in some patients. Reference Kane, Davis, Schooler, Marder, Casey and Brauzer31,Reference Eklund and Forsman33

Perphenazine plasma levels of 1–5 nmol/l have been associated with good therapeutic response, Reference Omerov, Wistedt, Bolvig-Hansen and Larsen34 although a higher therapeutic range has been suggested. Reference Mazure, Nelson, Jatlow, Kincare and Bowers35 In a study that aimed to discover the minimum effective dose of perphenazine decanoate, it was found that this was around 100 mg every 2 weeks (range 21.6–270.5 per 2 weeks). Reference Kistrup, Gerlach, Aaes-Jorgensen and Larsen13 Plasma levels ranged from around 2 nmol/l to 18 nmol/l depending on dose. In a further study of responding patients receiving 108.5 mg perphenazine decanoate every 2 weeks, plasma levels averaged 5 nmol/l and ranged from around 2 nmol/l to 6 nmol/l. Reference Knudsen, Hansen and Larsen11 Other studies have shown that doses of around 100 mg every 2 weeks are effective and provide plasma levels in the range 3–11 nmol/l. Reference Knudsen, Hansen, Hojholdt and Larsen36,Reference Knudsen, Hansen, Hojholdt and Larsen37

Pipotiazine palmitate is usually first given as a test dose of 25 mg. Maintenance doses are usually within the range 50–200 mg every month (up to 250 mg in Canada). The dose–response relationship is poorly defined. An early study suggested that the optimal dose of pipotiazine palmitate was between 100 mg and 600 mg every 4 weeks. Reference Gallant, Mielke, Bishop, Oelsner and Guerrero-Figueroa38 Later trials found doses of 50–200 mg every 4 weeks to be adequately effective. Reference Schmidt39,Reference Burch and Ayd40

Zuclopenthixol decanoate is initiated with a test dose of 100 mg and maintenance doses vary from 200 mg every 4 weeks to 600 mg a week. In an open dose-reduction study of 23 patients, the minimum effective dose was shown to be, on average, 200 mg every 2 weeks (range 60–400 mg). Reference Solgaard, Kistrup, Aaes-Jorgensen and Gerlach41 In practice zuclopenthixol is often given at weekly or 2-weekly intervals. However, doses of 100–600 mg (mean 284 mg) given every 4 weeks have been shown to be as effective in preventing relapse as haloperidol decanoate (38–200 mg per month). Reference Wistedt, Koskinen, Thelander, Nerdrum, Pedersen and Molbjerg42 Fortnightly or monthly administration thus seems reasonable in practice in the out-patient setting for maintenance treatment.

Olanzapine pamoate is licensed to be given in doses ranging from 150 mg every 2 weeks to 300 mg every 2 weeks. Supplementation with oral olanzapine is not usually required owing to prompt early release of olanzapine from the formulation. In a placebo-controlled trial, doses of 210 mg per 2 weeks, 300 mg per 2 weeks and 405 mg per 4 weeks were equally effective and well tolerated. Reference Lauriello, Lambert, Andersen, Lin, Taylor and McDonnell43

Risperidone LAI is usually begun at a dose of 25 mg or 37.5 mg according to the dose of antipsychotic previously received. Ideally, patients should first be treated with oral risperidone to assess dose requirements and tolerability. Supplementary oral antipsychotic treatment is required for 4–6 weeks after the first risperidone LAI. Dosing is licensed only at 2-weekly intervals, although there is emerging evidence that monthly dosing might be effective. Reference Gharabawi, Gearhart, Lasser, Mahmoud, Zhu and Mannaert44 Doses of 25 mg, 37.5 mg, 50 mg and 75 mg every 2 weeks have been shown to be broadly equal in effectiveness in clinical trials. Reference Moller45Reference Keks, Ingham, Khan and Karcher47 The maximum licensed dose is 50 mg every 2 weeks (a dose of 75 mg per 2 weeks causes significantly more extrapyramidal symptoms). Reference Kane, Eerdekens, Lindenmayer, Keith, Lesem and Karcher46 In practice, the lowest licensed dose of 25 mg every 2 weeks may be relatively ineffective. Reference Taylor, Young and Patel48,Reference Bai, Ting, Chen, Chang, Wu and Hung49

Pharmacodynamics

Both first-generation and second-generation antipsychotics administered by any route are presumed to exert their antipsychotic action by blocking central dopamine receptors of the D2 receptor subtype. All LAIs are potent in vitro antagonists of D2 receptors. In vivo, potency in humans is usually determined by measuring striatal D2 occupancy using methods such as positron emission tomography (PET) and single photon emission computerised tomography (SPECT). Therapeutic response seems to be associated with striatal D2 occupancies of 65% or greater, whereas hyperprolactinaemia and extrapyramidal side-effects occur at occupancies of 72% and 78% respectively, Reference Kapur, Zipursky, Jones, Remington and Houle50 although these observations are based on a single study including a limited number of individuals. Antipsychotic action may not be derived from activity in the striatum, but striatal occupancies correlate closely with response to antipsychotic treatment. Reference Agid, Mamo, Ginovart, Vitcu, Wilson and Zipursky51

Few studies have examined striatal D2 occupancy afforded by the use of LAIs. The earliest PET study found occupancies of 50–80% for patients receiving haloperidol decanoate 50–200 mg a month, pipotiazine palmitate 100 mg a month and fluphenazine decanoate 200 mg a month. Reference Baron, Martinot, Cambon, Boulenger, Poirier and Caillard52 Occupancy levels appeared to increase in the 20 days following injection. A later study of four patients receiving 30–50 mg haloperidol decanoate a month showed that peak D2 occupancies were in the range 66–82% a week after injection. Reference Nyberg, Farde and Halldin53 In an extension of this study (n=8, including two patients who participated in the previous study), D2 occupancies peaked at 60–82% a week after injection (doses 30–50 mg every 4 weeks). Reference Nyberg, Farde, Halldin, Dahl and Bertilsson54 Four weeks later occupancies were in the range 20–74%. The patients remained well despite these low occupancies.

A relatively recent study of patients responding to perphenazine decanoate found that at plasma levels of 1.8–9 nmol/l striatal D2 occupancy ranged from 66% to 82%. Reference Talvik, Nordstrom, Larsen, Jucaite, Cervenka and Halldin55 Doses used were in the range 38–108 mg per 2 weeks.

One study has examined D2 occupancy produced by olanzapine pamoate. Reference Mamo, Kapur, Keshavan, Laruelle, Taylor and Kothare56 Participants were switched from oral olanzapine to the LAI (300 mg every 4 weeks) and followed up for 6 months. Dopamine D2 receptor occupancy averaged 69% for oral olanzapine (5–20 mg per day) but then fell to around 55% in the first month before rising to just over 60% (all trough samples). There was no change in patients' clinical condition over the study period.

Two studies have determined D2 occupancy associated with the use of risperidone LAI. In the first, 28 patients received 2-weekly injections of 25 mg, 50 mg or 75 mg risperidone and D2 occupancy was estimated 2 weeks after the fifth injection (i.e. at steady-state trough plasma levels): the D2 occupancies were 25–48% for 25 mg, 59–83% for 50 mg and 62–72% for 75 mg. Reference Gefvert, Eriksson, Persson, Helldin, Bjorner and Mannaert22 In the second study D2 occupancies were determined for nine participants up to 3 days after injection and less than 5 days before the next injection. Reference Remington, Mamo, Labelle, Reiss, Shammi and Mannaert57 All participants had at least five injections before PET determinations were carried out. Occupancies were 53–75% for two patients receiving 25 mg, 59–85% for five patients receiving 50 mg and 71–85% for two patients receiving 75 mg.

Data on D2 occupancies associated with the use of LAIs are somewhat perplexing. They appear to show apparently subtherapeutic D2 occupancies associated with some doses, at least at some time points, without attendant relapse. This may mean that persistently high striatal D2 receptor occupancy is not required for full therapeutic effect, or that other D2 activity outside the striatum or other receptor activities are relevant. On the other hand, the low occupancies seen with low doses of haloperidol and risperidone do fit with clinical data which suggest that these doses are suboptimal. Reference Kane, Davis, Schooler, Marder, Casey and Brauzer31,Reference Eklund and Forsman33,Reference Taylor, Young and Patel48,Reference Bai, Ting, Chen, Chang, Wu and Hung49

Adverse effects

Long-acting injection formulations of FGAs might be expected to give rise to acute extrapyramidal symptoms, tardive dyskinesia and symptoms related to hyperprolactinaemia. Reference Haddad, Taylor and Niaz58 The frequency of movement disorders and tardive dyskinesia with FGA–LAIs is similar to that seen with oral FGAs. Reference Adams, Fenton, Quraishi and David59 Consequently the clinical utility of the LAIs is often seen to be severely limited by the likelihood of these important adverse effects (Appendix 1).

Flupentixol decanoate typically causes extrapyramidal symptoms, sedation and dry mouth. Reference Eberhard and Hellbom60 In one study of patients receiving 40 mg every 3 weeks, 97% showed signs of extrapyramidal symptoms and 82% received anti-Parkinsonian medication. Reference Knights, Okasha, Salih and Hirsch61 Fluphenazine decanoate (25 mg every 3 weeks) in the same trial caused extrapyramidal symptoms in 97% of patients and 93% required anti-Parkinsonian medication. Rates of other important side-effects such as incident depression were virtually identical for flupentixol and fluphenazine (53% v. 55%). Reference Knights, Okasha, Salih and Hirsch61 Fluphenazine decanoate is also associated with blurred vision and excess salivation. Reference Johnson62 Weight gain of more than 1 kg is common. Reference Chouinard, Annable and Campbell63 Changes in prolactin levels seem to be dose-related. Reference Bechelli, Iecco, Acioli and Pontes64

Haloperidol decanoate is also associated with a dose-dependent rise in plasma prolactin and in one study caused extrapyramidal symptoms severe enough to warrant anti-Parkinsonian drugs in around 90% of patients (dose averaging around 250 mg every 2–4 weeks). Reference Chouinard, Annable and Campbell63 Although haloperidol decanoate is often assumed to give the highest risk of extrapyramidal symptoms, this has not been demonstrated in comparative studies. Reference Kissling, Moller, Walter, Wittmann, Krueger and Trenk8,Reference Wistedt, Koskinen, Thelander, Nerdrum, Pedersen and Molbjerg42,Reference Wistedt, Persson and Hellbom65 Weight gain may be less common than with fluphenazine decanoate. Reference Wistedt66

Perphenazine decanoate, like other first-generation drugs, is associated with acute extrapyramidal symptoms. Reported prevalences of such symptoms or use of anticholinergic drugs range from 29% Reference Knudsen, Hansen, Hojholdt and Larsen36 to 55%. Reference Kistrup, Gerlach, Aaes-Jorgensen and Larsen13 Other studies suggest high rates of extrapyramidal symptoms and hyperprolactinaemia with perphenazine enanthate, Reference Lindholm, Gullberg, Ohman and Sedvall67,Reference Ahlfors, Dencker, Gravem and Remvig68 but this ester of perphenazine is known to result in much higher plasma levels of perphenazine than perphenazine decanoate. Reference Knudsen, Hansen, Auken, Waehrens, Hojholdt and Larsen69 The total number of patients examined in well-conducted trials with either ester appears to be only 313. Reference David, Quraishi and Rathbone70 Injection site abscesses have been reported for perphenazine enanthate. Reference Starmark, Forsman and Wahlstrom71 Weight gain seems not to occur with perphenazine decanoate. Reference Knudsen, Hansen, Hojholdt and Larsen36

Pipotiazine palmitate (mean dose 65 mg per month) has been shown to cause similar rates of extrapyramidal symptoms as haloperidol decanoate (mean dose 100 mg per month): 36% and 29% respectively. Reference Bechelli, Iecco, Acioli and Pontes64 In the same study weight gain of more than 5 kg was more common in those receiving pipotiazine (39% v. 16%). As a piperidine phenothiazine, pipotiazine might be expected to show a low incidence of extrapyramidal symptoms. There is limited evidence that pipotiazine has a relatively low potential for causing these symptoms compared with other LAIs. Reference Burch and Ayd40

Zuclopenthixol typically causes extrapyramidal symptoms and mild autonomic effects such as postural dizziness and blurred vision. Reference Wistedt, Koskinen, Thelander, Nerdrum, Pedersen and Molbjerg42 In one study of doses ranging from 50 mg to 300 mg every 2 weeks, 61% of participants experienced extrapyramidal symptoms and 78% required anti-Parkinsonian medication. Reference Solgaard, Kistrup, Aaes-Jorgensen and Gerlach41

A meta-analysis of comparative trials of FGA–LAI doses revealed no differences in the rates of extrapyramidal symptoms and tardive dyskinesia compared with oral medication and few important differences between individual LAIs. Reference Adams, Fenton, Quraishi and David59 Other sources suggest an increased incidence of extrapyramidal symptoms and tardive dyskinesia with LAIs compared with oral antipsychotics. Reference Barnes and Curson1 Both reviews reported an increased risk of movement disorders with fluphenazine decanoate compared with other FGA–LAIs. Reference Haddad, Taylor and Niaz58 All FGA–LAIs are associated with acute and chronic local reactions at injection sites, particularly when used in high doses or injection volumes. Reference Hay72

Adverse effects seen with olanzapine pamoate are similar to those seen with oral olanzapine. Common side-effects include increased plasma triglycerides, weight gain, sedation and increased appetite. Reference Lauriello, Lambert, Andersen, Lin, Taylor and McDonnell43 Extrapyramidal symptoms have not been reported more frequently than with oral olanzapine and the incidence of tardive dyskinesia is not known.

There has been no report of any important difference in adverse events between oral olanzapine and the LAI formulation, with the exception of a safety risk that emerged in clinical trials: post-injection syndrome. Reference McDonnell, Sorsaburu, Brunner, Detke, Anderson and Bergstrom73 This came to light when an unanticipated degree of sedation was observed in a small number of patients following an injection. This adverse event has occurred in 24 patients (up to end September 2007), an incidence of 1.2% of patients treated with olanzapine pamoate or 0.07% of injections given. Post-injection syndrome consists of sedation, confusion, dizziness, altered speech/dysarthria, somnolence and/or unconsciousness. These effects usually occur within an hour of injection, but the median time ranged from 20 min to 3 h after injection. To date all patients have fully recovered from this adverse event, usually within 3–72 h, without permanent sequelae and the majority (67%) have continued to receive the LAI formulation. Blood samples of olanzapine plasma concentrations were taken during the events and were found to be substantially elevated (Fig. 1).

Fig. 1 Olanzapine plasma concentration profiles for a period during which six injections were administered to a patient who experienced an inadvertent intravascular (IAIV) event after the second injection. Reference Lilly75

Solubility experiments have revealed that when olanzapine pamoate LAI is injected into the muscle as intended, the dissolution of the salt is gradual and results in a slow release of drug into the blood stream. However, if the salt comes into contact with a considerable amount of blood or plasma, as occurs if the needle punctures a vessel or enters a rich capillary bed during administration, the salt dissolves and therefore dissociates more quickly. Other factors thought to affect the dissolution rate of the pamoate salt are the volume and rate of blood flow and the degree of vascular injury. Reference Lilly74

The patient should be advised about the potential risk of post-injection syndrome and needs to be observed for 3 h in a healthcare facility each time that the injection is administered. Reference Lilly74 Prior to giving the injection the clinician should determine that the patient will not have to travel alone to his or her next destination. During the post-injection observation period it should be confirmed that the patient is alert and oriented and that there is no sign or symptom of overdose. If post-injection syndrome is suspected, close medical supervision and monitoring should continue until examination indicates that signs and symptoms have resolved. For the remainder of the day after injection, patients should be advised to be vigilant for signs and symptoms of overdose secondary to post-injection adverse reactions, should be able to obtain assistance if needed and should not drive or operate machinery.

Risperidone LAI is associated with relatively low rates of movement disorder. Reference Moller45 In one study extrapyramidal symptoms were seen in 10% of participants receiving 25 mg every 2 weeks and in 24% of those receiving 50 mg. Reference Kane, Eerdekens, Lindenmayer, Keith, Lesem and Karcher46 Other studies confirm this low rate of movement disorder. Reference Keks, Ingham, Khan and Karcher47,Reference Schmauss, Sacchetti, Kahn and Medori76 Weight gain averages around 3 kg in the longer term (up to 1 year). Reference Moller45 Risperidone LAI is associated with an incidence of increased plasma prolactin similar to that seen with oral risperidone but the magnitude of this increase is lower, Reference Moller45 perhaps because risperidone LAI is associated with lower plasma levels of the active drug moiety. Reference Bai, Ting, Chen, Chang, Wu and Hung49 Injection site pain and local reactions have been carefully evaluated – pain is not usually severe and injection site reactions are rare. Reference Moller45 Outside clinical trials, injection site pain leading to treatment withdrawal is uncommon. Reference Taylor, Young and Patel48,Reference Niaz and Haddad77 Tardive dyskinesia has been shown to emerge in 1.19% of treated individuals over 1 year. Reference Gharabawi, Bossie, Zhu, Mao and Lasser78 A similarly low incidence is also seen in more susceptible populations such as elderly patients. Reference Kissling, Glue, Medori and Simpson79 (Comparative data for FGA–LAIs are scant. One study observed an increase in the prevalence of oral tardive dyskinesia from 8% to 22% over a 3-year period during which fluphenazine LAI or flupentixol LAI were used.) Reference Gibson80

Initiation and switching to LAIs

The use of LAIs is confined to those who have already received antipsychotic medication: these injections should never be given to antipsychotic-naive patients. Treatment with LAIs is therefore initiated in those already taking antipsychotics or those who, for whatever reason, have recently stopped taking them. Initiation of LAIs is complicated by a number of factors (Appendix 2). First-generation antipsychotic LAIs are usually first given as a test dose, or at least at a dose at the lower end of the licensed range. These test doses are aimed at establishing patient tolerability of the drug and vehicle administered. However, test doses given in this way only really establish or otherwise the tolerability of a single, small dose of the LAI. Their use has little bearing on likely tolerability in multiple, higher-dose treatment because plasma levels at steady state are considerably higher than after a single dose. For example, in one study in which patients were given fluphenazine decanoate 50 mg every week, plasma levels quadrupled between the end of the first week and the end of the sixth week. Reference Ereshefsky, Saklad, Jann, Davis, Richards and Seidel81 This substantial rise in plasma levels despite there being no change in dose is likely to be seen with all LAIs because of their shared long plasma half-lives, and makes sensible dose titration almost impossible. An added difficulty here is that population dose–response relationships are poorly defined for most LAIs (olanzapine, risperidone and haloperidol are the exceptions). In practice, therefore, dose optimisation is a matter largely of conjecture, and patient tolerability is often used as a proxy for dose limitation. Furthermore, the use of oral antipsychotics as ‘cover’ during the early stages of LAI treatment often leads to prolonged polypharmacy (there is evidence that prescribers tend not to change regimens once patients improve, presumably because to do so would be to risk relapse). Reference Taylor, Mir, Mace and Whiskey82

Conclusion

All antipsychotic LAIs enable infrequent administration and assurance of adherence (or at least ready detection of non-adherence). However, appropriate prescribing of LAI preparations is clearly complicated by their shared prolonged apparent half-lives, their delayed release (particularly with risperidone) and the risk of post-injection syndrome (olanzapine). The use of test doses appears not to mitigate this complexity nor to minimise tolerability difficulties. Moreover, the lack of robust dose–response data for most preparations makes for rather approximate dosing in practice. Even with risperidone and haloperidol LAIs (for which fixed-dose studies have established the dose–response relationship) doubts persist over the clinical value of doses at the lower end of the dose range.

First-generation antipsychotic LAIs are clearly linked to high rates of extrapyramidal effects and tardive dyskinesia, among other adverse effects. There appear to be few differences between individual FGA–LAIs, although fluphenazine decanoate may be associated with a relatively higher rate of movement disorder and pipotiazine palmitate a relatively lower rate. Risperidone LAI has, on the face of it, important advantages over FGA drugs in respect of movement disorders, but its use is complicated – and clinical utility compromised – by excessively delayed release and by the requirement for cold storage of the product. Olanzapine LAI also appears to have advantages over FGA formulations but its use is complicated by the risk of post-injection syndrome and by the risk of metabolic adverse effects. The apparent advantages of SGA–LAIs need to be confirmed in large-scale randomised controlled trials. It is notable that the claimed advantages of oral SGAs become less apparent when these drugs are directly compared with oral FGAs in well-conducted trials. Reference Lieberman, McEvoy, Swartz, Rosenheck, Perkins and Keefe83,Reference Jones, Barnes, Davies, Dunn, Lloyd and Hayhurst84

Appendix 1

Adverse effects

Extrapyramidal symptoms

Symptoms are tremor, rigidity, mask-like face, reduced arm swing, drooling, dystonia (muscular spasm), oculogyric crisis, akathisia (severe inner restlessness).

Tardive dyskinesia

Tardive dyskinesia is characterised by:

  1. (a) abnormal masticatory movements (lip-smacking, licking);

  2. (b) tongue protrusion;

  3. (c) vermicular (worm-like) movement;

  4. (d) abnormal postures (tardive dystonia): opisthotonos (arching of back and neck), retrocollis (backward arching of head and neck), torticollis (sideways turn of head and neck);

  5. (e) digital abnormalities (‘pill-rolling’, ‘piano playing’, ‘guitar playing’);

  6. (f) grimacing.

Metabolic adverse effects

Effects include impaired glucose tolerance/diabetes mellitus, weight gain, increased triglyceride levels, increased cholesterol levels.

Appendix 2

Factors complicating long-acting injection dosing

  1. (a) The effect of any dose is likely to be prolonged. Test doses are usually required but may not properly assess tolerability in longer-term use.

  2. (b) Some long-acting injections (LAIs) show delayed as well as prolonged release. Cover with oral antipsychotics may be necessary.

  3. (c) Attainment of steady-state plasma levels is usually delayed for up to 2–3 months. During this time, plasma levels are likely to rise substantially even when dosages are not increased.

  4. (d) Dose–response relationships are poorly defined for most LAIs.

References

1 Barnes, TR, Curson, DA. Long-term depot antipsychotics. A risk–benefit assessment. Drug Saf 1994; 10: 464–79.CrossRefGoogle ScholarPubMed
2 Jann, MW, Ereshefsky, L, Saklad, SR. Clinical pharmacokinetics of the depot antipsychotics. Clin Pharmacokinet 1985; 10: 315–33.CrossRefGoogle ScholarPubMed
3 Stauning, JA, Kirk, L, Joorgensen, A. Comparison of serum levels after intramuscular injections of 2% and 10% cis(Z)-flupentixol decanoate in Viscoleo to schizophrenic patients. Psychopharmacology (Berl) 1979; 65: 6972.CrossRefGoogle ScholarPubMed
4 Tuninger, E, Levander, S. Large variations of plasma levels during maintenance treatment with depot neuroleptics. Br J Psychiatry 1996; 169: 618–21.Google Scholar
5 Wiles, DH, Gelder, MG. Plasma fluphenazine levels by radioimmunoassay in schizophrenic patients treated with depot injections of fluphenazine decanoate. Br J Clin Pharmacol 1979; 8: 565–70.Google Scholar
6 Curry, SH, Whelpton, R, De Schepper, PJ, Vranckx, S, Schiff, AA. Kinetics of fluphenazine after fluphenazine dihydrochloride, enanthate and decanoate administration to man. Br J Clin Pharmacol 1979; 7: 325–31.Google Scholar
7 Ereshefsky, L, Jann, MW, Saklad, SR, Davis, CM, Richards, AL, Burch, NR. Effects of smoking on fluphenazine clearance in psychiatric inpatients. Biol Psychiatry 1985; 20: 329–32.Google Scholar
8 Kissling, W, Moller, HJ, Walter, K, Wittmann, B, Krueger, R, Trenk, D. Double-blind comparison of haloperidol decanoate and fluphenazine decanoate effectiveness, side-effects, dosage and serum levels during a six months' treatment for relapse prevention. Pharmacopsychiatry 1985; 18: 240–5.Google Scholar
9 Jann, MW, Saklad, SR, Ereshefsky, L, Richards, AL, Harrington, CA, Davis, CM. Effects of smoking on haloperidol and reduced haloperidol plasma concentrations and haloperidol clearance. Psychopharmacology 1986; 90: 468–70.Google Scholar
10 Nayak, RK, Doose, DR, Nair, NP. The bioavailability and pharmacokinetics of oral and depot intramuscular haloperidol in schizophrenic patients. J Clin Pharmacol 1987; 27: 144–50.Google Scholar
11 Knudsen, P, Hansen, LB, Larsen, NE. Perphenazine decanoate in sesame oil vs. perphenazine enanthate in sesame oil: a comparative study of pharmacokinetic properties and some clinical implications. Acta Psychiatr Scand Suppl 1985; 322: 11–4.Google Scholar
12 Larsen, NE, Hansen, LB. Prediction of the optimal perphenazine decanoate dose based on blood samples drawn within the first three weeks. Ther Drug Monit 1989; 11: 642–6.Google Scholar
13 Kistrup, K, Gerlach, J, Aaes-Jorgensen, T, Larsen, NE. Perphenazine decanoate and cis(z)-flupentixol decanoate in maintenance treatment of schizophrenic outpatients. Serum levels at the minimum effective dose. Psychopharmacology (Berl) 1991; 105: 42–8.CrossRefGoogle ScholarPubMed
14 Girard, M, Granier, F, Schmitt, L, Cotonat, J, Escande, M, Blanc, M. [Initial results of a pharmacokinetic study of pipothiazine and its palmitic ester (Piportil L4) in a schizophrenic population]. Encephale 1984; 10: 171–6.Google Scholar
15 Altamura, AC, Sassella, F, Santini, A, Montresor, C, Fumagalli, S, Mundo, E. Intramuscular preparations of antipsychotics: uses and relevance in clinical practice. Drugs 2003; 63: 493512.CrossRefGoogle ScholarPubMed
16 Viala, A, Hou, N, Ba, B, Durand, A, Dufour, H, D'Agostino, N, et al. Blood and plasma kinetics of cis(Z)-clopenthixol and fluphenazine in psychiatric patients after intramuscular injection of their decanoic esters. Psychopharmacology (Berl) 1984; 83: 147–50.Google Scholar
17 Poulsen, JH, Olesen, OV, Larsen, NE. Fluctuation of serum zuclopenthixol concentrations in patients treated with zuclopenthixol decanoate in viscoleo. Ther Drug Monit 1994; 16: 155–9.Google Scholar
18 Jorgensen, A, Overo, KF. Clopenthixol and flupenthixol depot preparations in outpatient schizophrenics. III. Serum levels. Acta Psychiatr Scand Suppl 1980; 279: 4154.CrossRefGoogle Scholar
19 Viala, A, Ba, B, Durand, A, Gouezo, F, Hou, N, Jorgensen, A. Comparative study of the pharmacokinetics of zuclopenthixol decanoate and fluphenazine decanoate. Psychopharmacology (Berl) 1988; 94: 293–7.Google Scholar
20 Kurtz, D, Bergstrom, R, McDonnell, D, Mitchell, M. Pharmacokinetics (PK) of multiple doses of olanzapine long-acting injection (OLAI), an intramuscular (IM) depot formulation of olanzapine (OLZ), in stabilized patients with schizophrenia. Biol Psychiatry 2008; 63 (suppl 1): 228S.Google Scholar
21 Ramstack, M, Grandolfi, GP, Mannaert, E, D'Hoore, P, Lasser, RA. Long-acting risperidone: prolonged-release injectable delivery of risperidone using Medisorb microsphere technology. Schizophr Res 2003; 60 (suppl 1): 314.Google Scholar
22 Gefvert, O, Eriksson, B, Persson, P, Helldin, L, Bjorner, A, Mannaert, E, et al. Pharmacokinetics and D2 receptor occupancy of long-acting injectable risperidone (Risperdal Consta) in patients with schizophrenia. Int J Neuropsychopharmacol 2005; 8: 2736.Google Scholar
23 Agrup-Andersson, L, Bengtsson, A, Erlandsson, K, Gottfries, CG, Witzell-Ostlund, G. Flupenthixol decanoate. Controlled investigation concerning dosage. Acta Psychiatr Scand Suppl 1974; 255: 714.Google Scholar
24 McCreadie, RG, Flanagan, WL, McKnight, J, Jorgensen, A. High dose flupenthixol decanoate in chronic schizophrenia. Br J Psychiatry 1979; 135: 175–9.Google Scholar
25 Johnson, DA, Ludlow, JM, Street, K, Taylor, RD. Double-blind comparison of half-dose and standard-dose flupenthixol decanoate in the maintenance treatment of stabilised out-patients with schizophrenia. Br J Psychiatry 1987; 151: 634–8.Google Scholar
26 Cookson, IB. The effects of a 50% reduction of cis(z)-flupenthixol decanoate in chronic schizophrenic patients maintained on a high dose regime. Int Clin Psychopharmacol 1987; 2: 141–9.CrossRefGoogle ScholarPubMed
27 Carpenter, WT, Buchanan, RW, Kirkpatrick, B, Lann, HD, Breier, AF, Summerfelt, AT. Comparative effectiveness of fluphenazine decanoate injections every 2 weeks versus every 6 weeks. Am J Psychiatry 1999; 156: 412–8.CrossRefGoogle ScholarPubMed
28 Geller, J. Use of extremely low doses of fluphenazine decanoate. Am J Psychiatry 1990; 147: 672.Google Scholar
29 Marder, SR, Van Putten, T, Mintz, J, Lebell, M, McKenzie, J, May, PR. Low- and conventional-dose maintenance therapy with fluphenazine decanoate. Two-year outcome. Arch Gen Psychiatry 1987; 44: 518–21.Google Scholar
30 Capstick, N. Long-term fluphenazine decanoate maintenance dosage requirements of chronic schizophrenic patients. Acta Psychiatr Scand 1980; 61: 256–62.Google Scholar
31 Kane, JM, Davis, JM, Schooler, N, Marder, S, Casey, D, Brauzer, B, et al. A multidose study of haloperidol decanoate in the maintenance treatment of schizophrenia. Am J Psychiatry 2002; 159: 554–60.Google Scholar
32 Taylor, D. Establishing a dose–response relationship for haloperidol decanoate. Psychiatr Bull 2005; 29: 104–7.Google Scholar
33 Eklund, K, Forsman, A. Minimal effective dose and relapse – double-blind trial: haloperidol decanoate vs. placebo. Clin Neuropharmacol 1991; 14 (suppl 2): S712.Google Scholar
34 Omerov, M, Wistedt, B, Bolvig-Hansen, L, Larsen, NE. The relationship between perphenazine plasma levels and clinical response in acute schizophrenia. Prog Neuropsychopharmacol Biol Psychiatry 1989; 13: 159–66.Google Scholar
35 Mazure, CM, Nelson, JC, Jatlow, PI, Kincare, P, Bowers, MB. The relationship between blood perphenazine levels, early resolution of psychotic symptoms, and side effects. J Clin Psychiatry 1990; 51: 330–4.Google ScholarPubMed
36 Knudsen, P, Hansen, LB, Hojholdt, K, Larsen, NE. Long-term depot neuroleptic treatment with perphenazine decanoate. I. Efficacy and side effects in a 12 month study of 42 drug monitored psychotic patients. Acta Psychiatr Scand Suppl 1985; 322: 2940.Google Scholar
37 Knudsen, P, Hansen, LB, Hojholdt, K, Larsen, NE. Long-term depot neuroleptic treatment with perphenazine decanoate. II. Different depot intervals in the last 6 months of a 12 month study of 42 drug monitored psychotic patients. Acta Psychiatr Scand Suppl 1985; 322: 4150.CrossRefGoogle ScholarPubMed
38 Gallant, DM, Mielke, D, Bishop, G, Oelsner, T, Guerrero-Figueroa, R. Pipotiazine palmitate: an evaluation of a new long acting intramuscular antipsychotic agent in severely ill schizophrenic patients. Dis Nerv Syst 1975; 36: 193–6.Google Scholar
39 Schmidt, K. Pipothiazine palmitate: a versatile, sustained-action neuroleptic in psychiatric practice. Curr Med Res Opin 1986; 10: 326–9.Google Scholar
40 Burch, EA, Ayd, FJ. Depot pipotiazine 1970–1982: a review. J Clin Psychiatry 1983; 44: 242–7.Google Scholar
41 Solgaard, T, Kistrup, K, Aaes-Jorgensen, T, Gerlach, J. Zuclopenthixol decanoate in maintenance treatment of schizophrenic outpatients. Minimum effective dose and corresponding serum levels. Pharmacopsychiatry 1994; 27: 119–23.Google Scholar
42 Wistedt, B, Koskinen, T, Thelander, S, Nerdrum, T, Pedersen, V, Molbjerg, C. Zuclopenthixol decanoate and haloperidol decanoate in chronic schizophrenia: a double-blind multicentre study. Acta Psychiatr Scand 1991; 84: 1421.CrossRefGoogle ScholarPubMed
43 Lauriello, J, Lambert, T, Andersen, S, Lin, D, Taylor, CC, McDonnell, D. An 8-week, double-blind, randomized, placebo-controlled study of olanzapine long-acting injection in acutely ill patients with schizophrenia. J Clin Psychiatry 2008; 69: 790–9.Google Scholar
44 Gharabawi, GM, Gearhart, NC, Lasser, RA, Mahmoud, RA, Zhu, Y, Mannaert, E, et al. Maintenance therapy with once-monthly administration of long-acting injectable risperidone in patients with schizophrenia or schizoaffective disorder: a pilot study of an extended dosing interval. Ann Gen Psychiatry 2007; 6: 3.CrossRefGoogle ScholarPubMed
45 Moller, HJ. Long-acting injectable risperidone for the treatment of schizophrenia: clinical perspectives. Drugs 2007; 67: 1541–66.Google Scholar
46 Kane, JM, Eerdekens, M, Lindenmayer, JP, Keith, SJ, Lesem, M, Karcher, K. Long-acting injectable risperidone: efficacy and safety of the first long-acting atypical antipsychotic. Am J Psychiatry 2003; 160: 1125–32.Google Scholar
47 Keks, NA, Ingham, M, Khan, A, Karcher, K. Long-acting injectable risperidone v. olanzapine tablets for schizophrenia or schizoaffective disorder: randomised, controlled, open-label study. Br J Psychiatry 2007; 191: 131–9.Google Scholar
48 Taylor, DM, Young, C, Patel, MX. Prospective 6-month follow-up of patients prescribed risperidone long-acting injection: factors predicting favourable outcome. Int J Neuropsychopharmacol 2006; 9: 685–94.Google Scholar
49 Bai, YM, Ting, CT, Chen, JY, Chang, WH, Wu, B, Hung, CH, et al. Equivalent switching dose from oral risperidone to risperidone long-acting injection: a 48-week randomized, prospective, single-blind pharmacokinetic study. J Clin Psychiatry 2007; 68: 1218–25.CrossRefGoogle ScholarPubMed
50 Kapur, S, Zipursky, R, Jones, C, Remington, G, Houle, S. Relationship between dopamine D2 occupancy, clinical response, and side effects: a double-blind PET study of first-episode schizophrenia. Am J Psychiatry 2000; 157: 514–20.Google Scholar
51 Agid, O, Mamo, D, Ginovart, N, Vitcu, I, Wilson, AA, Zipursky, RB, et al. Striatal vs extrastriatal dopamine D2 receptors in antipsychotic response. A double-blind PET study in schizophrenia. Neuropsychopharmacology 2006; 32: 1209–15.Google Scholar
52 Baron, JC, Martinot, JL, Cambon, H, Boulenger, JP, Poirier, MF, Caillard, V, et al. Striatal dopamine receptor occupancy during and following withdrawal from neuroleptic treatment: correlative evaluation by positron emission tomography and plasma prolactin levels. Psychopharmacology (Berl) 1989; 99: 463–72.Google Scholar
53 Nyberg, S, Farde, L, Halldin, C. Delayed normalization of central D2 dopamine receptor availability after discontinuation of haloperidol decanoate. Preliminary findings. Arch Gen Psychiatry 1997; 54: 953–8.Google Scholar
54 Nyberg, S, Farde, L, Halldin, C, Dahl, ML, Bertilsson, L. D2 dopamine receptor occupancy during low-dose treatment with haloperidol decanoate. Am J Psychiatry 1995; 152: 173–8.Google Scholar
55 Talvik, M, Nordstrom, AL, Larsen, NE, Jucaite, A, Cervenka, S, Halldin, C, et al. A cross-validation study on the relationship between central D2 receptor occupancy and serum perphenazine concentration. Psychopharmacology (Berl) 2004; 175: 148–53.Google Scholar
56 Mamo, D, Kapur, S, Keshavan, M, Laruelle, M, Taylor, CC, Kothare, PA, et al. D2 receptor occupancy of olanzapine pamoate depot using positron emission tomography: an open-label study in patients with schizophrenia. Neuropsychopharmacology 2008; 33: 298304.Google Scholar
57 Remington, G, Mamo, D, Labelle, A, Reiss, J, Shammi, C, Mannaert, E, et al. A PET study evaluating dopamine D2 receptor occupancy for long-acting injectable risperidone. Am J Psychiatry 2006; 163: 396401.Google Scholar
58 Haddad, PM, Taylor, M, Niaz, OS. First-generation antipsychotic long-acting injections v. oral antipsychotric medication in schizophrenia: systematic review of randomised controlled trials and observational studies. Br J Psychiatry 2009; 195 (suppl 52): s208.Google Scholar
59 Adams, CE, Fenton, MKP, Quraishi, S, David, AS. Systematic meta-review of depot antipsychotic drugs for people with schizophrenia. Br J Psychiatry 2001; 179: 290–9.Google Scholar
60 Eberhard, G, Hellbom, E. Haloperidol decanoate and flupenthixol decanoate in schizophrenia. A long-term double-blind cross-over comparison. Acta Psychiatr Scand 1986; 74: 255–62.CrossRefGoogle ScholarPubMed
61 Knights, A, Okasha, MS, Salih, MA, Hirsch, SR. Depressive and extrapyramidal symptoms and clinical effects: a trial of fluphenazine versus flupenthixol in maintenance of schizophrenic out-patients. Br J Psychiatry 1979; 135: 515–23.Google Scholar
62 Johnson, DAW. The side-effects of fluphenazine decanoate. Br J Psychiatry 1973; 123: 519–22.Google Scholar
63 Chouinard, G, Annable, L, Campbell, W. A randomized clinical trial of haloperidol decanoate and fluphenazine decanoate in the outpatient treatment of schizophrenia. J Clin Psychopharmacol 1989; 9: 247–53.Google Scholar
64 Bechelli, LPC, Iecco, MC, Acioli, A, Pontes, MC. A double-blind trial of haloperidol decanoate and pipothiazine palmitate in the maintenance treatment of schizophrenics in a public out-patient clinic. Curr Ther Res 1985; 37: 662–71.Google Scholar
65 Wistedt, B, Persson, T, Hellbom, E. A clinical double-blind comparison between haloperidol decanoate and fluphenazine decanoate. Curr Ther Res 1984; 35: 804–14.Google Scholar
66 Wistedt, B. A comparative trial of haloperidol decanoate and fluphenazine decanoate in chronic schizophrenic patients. Int Clin Psychopharmacol 1986; 1 (suppl 1): 1523.Google Scholar
67 Lindholm, H, Gullberg, B, Ohman, A, Sedvall, G. Effects of perphenazine enanthate injections on prolactin levels in plasma from schizophrenic women and men. Psychopharmacology (Berl) 1978; 57: 14.Google Scholar
68 Ahlfors, UG, Dencker, SJ, Gravem, A, Remvig, J. Clopenthixol decanoate and perphenazine enanthate in schizophrenic patients. A double-blind Nordic multicentre trial. Acta Psychiatr Scand Suppl 1980; 279: 7791.Google Scholar
69 Knudsen, P, Hansen, LB, Auken, G, Waehrens, J, Hojholdt, K, Larsen, NE. Perphenazine decanoate vs. perphenazine enanthate: efficacy and side effects in a 6 week double-blind, comparative study of 50 drug monitored psychotic patients. Acta Psychiatr Scand Suppl 1985; 322: 1528.Google Scholar
70 David, A, Quraishi, S, Rathbone, J. Depot perphenazine decanoate and enanthate for schizophrenia. Cochrane Database Syst Rev 2005; (3): CD001717.Google Scholar
71 Starmark, JE, Forsman, A, Wahlstrom, . Abscesses following prolonged intramuscular administration of perphenazine enantate. Acta Psychiatr Scand 1980; 62: 154–7.Google Scholar
72 Hay, J. Complications at site of injection of depot neuroleptics. BMJ 1995; 311: 421.Google Scholar
73 McDonnell, D, Sorsaburu, S, Brunner, E, Detke, H, Anderson, S, Bergstrom, R, et al. Post-injection delirium/sedation syndrome observed with olanzapine long-acting injection: review of the first 25 events. Eur Neuropsychopharmacol 2008; 18 (suppl 4): S437.Google Scholar
74 Lilly, Eli. Summary of Product Characteristics. Zypadhera 210 mg, 300 mg, and 405 mg, powder and solvent for prolonged release suspension for injection. Lilly, 2009 (http://emc.medicines.org.uk/medicine/21361/SPC/ZYPADHERA+210+mg%2c+300+mg%2c+and+405+mg%2c+powder+and+solvent+for+prolonged+release+suspension+for+injection).Google Scholar
75 Lilly, Eli. Psychopharmacologic Drugs Advisory Committee Briefing Document. Zyprexa Olanzapine Pamoate (OP) Depot. Olanzapine Long-Acting Injection. Schizophrenia. Lilly, 2009 (http://www.fda.gov/ohrms/dockets/ac/08/briefing/2008-4338b1-03-Lilly.pdf).Google Scholar
76 Schmauss, M, Sacchetti, E, Kahn, JP, Medori, R. Efficacy and safety of risperidone long-acting injectable in stable psychotic patients previously treated with oral risperidone. Int Clin Psychopharmacol 2007; 22: 8592.Google Scholar
77 Niaz, OS, Haddad, PM. Thirty-five months experience of risperidone long-acting injection in a UK psychiatric service including a mirror-image analysis of in-patient care. Acta Psychiatr Scand 2007; 116: 3646.Google Scholar
78 Gharabawi, GM, Bossie, CA, Zhu, Y, Mao, L, Lasser, RA. An assessment of emergent tardive dyskinesia and existing dyskinesia in patients receiving long-acting, injectable risperidone: results from a long-term study. Schizophr Res 2005; 77: 129–39.Google Scholar
79 Kissling, W, Glue, P, Medori, R, Simpson, S. Long-term safety and efficacy of long-acting risperidone in elderly psychotic patients. Hum Psychopharmacol 2007; 22: 505–13.Google Scholar
80 Gibson, AC. Depot injections and tardive dyskinesia. Br J Psychiatry 1978; 133: 361–5.Google Scholar
81 Ereshefsky, L, Saklad, SR, Jann, MW, Davis, CM, Richards, A, Seidel, DR. Future of depot neuroleptic therapy: pharmacokinetic and pharmacodynamic approaches. J Clin Psychiatry 1984; 45: 50–9.Google Scholar
82 Taylor, D, Mir, S, Mace, S, Whiskey, E. Co-prescribing of atypical and typical antipsychotics – prescribing sequence and documented outcome. Psychiatr Bull 2002; 26: 170–2.Google Scholar
83 Lieberman, JA, McEvoy, JP, Swartz, MS, Rosenheck, RA, Perkins, DO, Keefe, RS, et al. Effectiveness of antipsychotic drugs in patients with chronic schizophrenia. N Engl J Med 2005; 353: 1209–23.Google Scholar
84 Jones, PB, Barnes, TR, Davies, L, Dunn, G, Lloyd, H, Hayhurst, KP, et al. Randomized controlled trial of the effect on Quality of Life of second- vs first-generation antipsychotic drugs in schizophrenia: Cost Utility of the Latest Antipsychotic Drugs in Schizophrenia Study (CUtLASS 1). Arch Gen Psychiatry 2006; 63: 1079–87.Google Scholar
Figure 0

Table 1 Pharmacokinetic and pharmacodynamic characteristics of antipsychotic long-acting injections

Figure 1

Fig. 1 Olanzapine plasma concentration profiles for a period during which six injections were administered to a patient who experienced an inadvertent intravascular (IAIV) event after the second injection.75

Submit a response

eLetters

No eLetters have been published for this article.