Next Article in Journal
Bioactive Peptides and Exercise Modulate the AMPK/SIRT1/PGC-1α/FOXO3 Pathway as a Therapeutic Approach for Hypertensive Rats
Previous Article in Journal
Synthesis of Quinolones and Zwitterionic Quinolonate Derivatives with Broad-Spectrum Antibiotic Activity
Previous Article in Special Issue
Depression and Autoimmune Hypothyroidism—Their Relationship and the Effects of Treating Psychiatric and Thyroid Disorders on Changes in Clinical and Biochemical Parameters Including BDNF and Other Cytokines—A Systematic Review
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Systematic Review

Adverse Drug Reactions in Relation to Clozapine Plasma Levels: A Systematic Review

by
Maria Skokou
1,†,
Eleni A. Karavia
2,†,
Zoi Drakou
1,2,†,
Vassiliki Konstantinopoulou
2,†,
Christina-Anna Kavakioti
1,
Philippos Gourzis
1,
Kyriakos E. Kypreos
2,3 and
Ourania Andreopoulou
1,*
1
Department of Psychiatry, General University Hospital of Patras, School of Medicine, University of Patras, 26504 Patras, Greece
2
Department of Pharmacology, School of Medicine, University of Patras, 26504 Patras, Greece
3
Department of Life Sciences, School of Sciences, European University of Cyprus, Nicosia 2404, Cyprus
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Pharmaceuticals 2022, 15(7), 817; https://doi.org/10.3390/ph15070817
Submission received: 17 May 2022 / Revised: 22 June 2022 / Accepted: 27 June 2022 / Published: 1 July 2022
(This article belongs to the Special Issue Psychopharmacology of Affective Disorders)

Abstract

:
Clozapine is the gold standard for treatment-resistant schizophrenia. Serious and even life-threatening adverse effects, mostly granulocytopenia, myocarditis, and constipation, are of great clinical concern and constitute a barrier to prescribing clozapine, thus depriving many eligible patients of a lifesaving treatment option. Interestingly, clozapine presents variable pharmacokinetics affected by numerous parameters, leading to significant inter- and intra-individual variation. Therefore, therapeutic drug monitoring of plasma clozapine levels confers a significant benefit in everyday clinical practice by increasing the confidence of the prescribing doctor to the drug and the adherence of the patient to the treatment, mainly by ensuring effective treatment and limited dose-related side effects. In the present systematic review, we aimed at identifying how a full range of adverse effects relates to plasma clozapine levels, using the Jadad grading system for assessing the quality of the available clinical evidence. Our findings indicate that EEG slowing, obsessive-compulsive symptoms, heart rate variability, hyperinsulinemia, metabolic syndrome, and constipation correlate to plasma clozapine levels, whereas QTc, myocarditis, sudden death, leucopenia, neutropenia, sialorrhea, are rather unrelated. Rapid dose escalation at the initiation of treatment might contribute to the emergence of myocarditis, or leucopenia. Strategies for managing adverse effects are different in these conditions and are discussed accordingly.

1. Introduction

Efficacy and safety represent core aspects of medicinal treatments, and Hippocrates was probably the first to employ these concepts, in his aphorism “first, do no harm”, as also quoted in the Hippocratic Oath in the 5th century B.C.: “And I will use regimens to the benefit of the ill in accordance with my ability and my judgment, but from what is to their harm or injustice I will keep them” [1]. Some centuries later, Paracelsus stressed the significance of the drug dose for the emergence of adverse outcomes, saying, “What is there that is not poison? All things are poison and nothing is without poison. Solely the dose determines that a thing is not a poison” [2]. For modern psychiatrists, the case of clozapine encompasses the quest for optimal efficacy and safety in a dramatic mode, since this drug combines superior antipsychotic efficacy with serious and sometimes life-threatening adverse effects. Clozapine use has significantly improved the quality of life of treated patients and a strong association between clozapine use and reduced risk of mortality from natural causes [3], and all-cause mortality [4] has been reported, in those with treatment-resistant schizophrenia (TRS). For many patients, it has proved to be a lifesaving medicine.
Clozapine (CLOZ) is the first of a class of antipsychotic compounds collectively termed as “atypical”, i.e., risperidone, olanzapine, quetiapine, aripiprazole, among others, all of which (with some exceptions) share D2 and 5-HT2A blockage properties and are associated with fewer extrapyramidal side effects (EPSE) [5,6]. In fact, clozapine has a minimal incidence of EPSE, which was at first surprising, since researchers in the past related antipsychotic efficacy with the emergence of extrapyramidal symptoms [7]. It is proven superior to all other antipsychotics for the treatment of multiepisode schizophrenia and represents a first-line and gold standard option for TRS [8,9]. TRS is defined by consensus criteria as having positive and negative symptoms of at least moderate severity and at least moderate functioning impairment, with failure to respond to at least two different antipsychotic medications administered for at least 6 weeks at a minimum dosage equivalent to 600 mg chlorpromazine; this situation is encountered in about 30% of schizophrenia cases [10,11]. Clozapine is also indicated for recurrent suicidality in patients with schizophrenia or schizoaffective disorder and psychosis in Parkinson’s disease, irresponsive to other antipsychotics. Off-label uses include tardive dyskinesia and dystonia, treatment-resistant bipolar disorder, and schizophrenia with severe hostility [12]. According to the anatomical therapeutic chemical classification system (ATC) clozapine belongs to the N05AH02 class of drugs with a daily defined dose (DDD) of 300 milligrams per os [13].
Despite extensive preclinical research on its pharmacodynamics, its mechanism of action remains elusive. It is a tricyclic dibenzodiazepine, known to have weak dopamine-receptor-blocking activity at D1, D2, D3, and D5 receptors, but exerts highly potent D4 receptor antagonism [14]. Blockade of D4 is considered important for clozapine’s antipsychotic action, because of its abundance in the hippocampus and mesolimbic system [15]. Additionally, clozapine displays potent anti-adrenergic (α1 and α2), anticholinergic (Μ1, Μ2, Μ3, Μ5), antihistaminic (H1), antiserotoninergic (potent 5-HT2A and 5-HT2C receptor antagonist) and anti-GABAergic properties (GABAA) [14]. It also exerts partial agonism on M4 receptors and possibly modulates glutamatergic and GABAB neurotransmission [14,16]. Remarkably, there is currently insufficient explanation for clozapine’s superior efficacy. Apart from its unique binding profile to various receptors, presumably over 35 different receptors, there has been demonstrated a positive effect on neuroprotection, through modulation of microglia activation [17]. On the other hand, the drug’s affinity for a variety of receptors accounts for several of its adverse effects, at least those that are considered dose-related. Muscarinic antagonism produces anticholinergic actions, in turn leading to tachycardia, constipation, urinary retention, and enuresis. Antagonism of α1 norepinephrine receptors may produce hypotension, sedation, bradycardia, and syncope. Antihistaminic action is associated with sedation and weight gain [18].
In terms of its pharmacokinetics, clozapine is almost thoroughly absorbed by the gut and undergoes extensive hepatic metabolism [19], mainly by CYP1A2 and to a lesser extent by CYP3A5 and CYP3A43 [20,21,22]. In vitro studies indicated that CYP2D6 and CYP2C19 have a minor role in clozapine metabolism [19,23,24]. Its most studied metabolites are norclozapine, (N-desmethyl-clozapine), considered active but to a lower extent than the mother molecule, and N-oxide -clozapine, which is inactive, but can convert back to clozapine [19]. Norclozapine (NCLOZ) has potent M4 cholinergic actions and is thought to possibly enhance cognition [25].
Clozapine demonstrates highly variable pharmacokinetics and for a given dose, plasma clozapine levels present large inter-and intra-individual differences affected by age, sex, genetics, dietary, clinical, pharmacological, drug interactions, and other parameters [26,27]. Therefore, prediction of plasma levels according to dose is impossible. In general, the dose regimen should be personalized, with the desirable plasma concentrations set for most patients at 350 ng/mL for optimal efficacy and safety [28].
Significant toxicity is generally associated with higher clozapine plasma levels. For example, existing evidence indicates that the incidence of seizures increases significantly at doses above 600 mg/day. Importantly, plasma levels over 1000 ng/mL have been linked to adverse central nervous system (CNS) effects and are not recommended due to a perceived risk of developing full-blown toxicity [29]. In the United Kingdom’s National Health Service (UK’s NHS) specific plasma monitoring protocols have been developed to this effect (i.e., https://www.hpft.nhs.uk/media/4116/clozapine-plasma-level-monitoring-guide-with-logos-mar19.pdf (accessed on 20 June 2022)) and dedicated clozapine clinical protocols have been established (i.e., http://www.rdash.nhs.uk/wp-content/uploads/2014/04/Clozapine-Clinic-Protocol-v3.pdf (accessed on 20 June 2022)). Recent reviews have investigated adverse effects in relation to clozapine plasma levels [30,31,32], such as peripheral adverse effects, neutropenia, EEG changes, and epileptic seizures, helping the search for a best-informed approach to clozapine use [33].
In the present systematic review, we discuss a full range of adverse effects of clozapine, in relation to plasma drug levels, classified by organ system. Our aim is to provide comprehensive and concise guidance to clinicians, researchers, and clinical pharmacologists on the safe use of clozapine, helping to overcome barriers responsible for its underutilization.

2. Results

2.1. General Considerations during Literature Search and Evaluation

The search strategy followed by the exclusion process yielded a total of 60 articles out of which 47 were original research papers and 13 were case reports, as shown in the flowchart of Figure 1. Although case reports were not included in the review, they are listed in Supplementary Table S1A, because rare adverse effects are not likely to be reported in the usually employed sample sizes. For most of the studies, clinical evidence was classified as low quality, due to numerous limitations. In most studies, blood samples were drawn immediately prior to the morning dose of clozapine, or 8 to 16 h after the night-time dose. Eleven studies did not mention explicitly the time of sampling. Many studies were performed in a small or small to medium size sample, ranging from 6 to 190 patients receiving clozapine. Correlation of clozapine or metabolite levels with dose was calculated in thirteen studies [34,35,36,37,38,39,40,41,42,43,44,45,46]; nine reported a positive correlation between dose and plasma clozapine levels alone or with metabolite levels as well [34,35,36,37,39,41,42,43,44] and two reported no correlation with clozapine and norclozapine levels [45,46]. Sporn et al. [40] found a positive correlation of clozapine dose with norclozapine but not clozapine plasma levels, and Mauri et al. [38] reported that clozapine dosage correlated with clozapine but not norclozapine plasma levels. Furthermore, 27 studies examined the correlation of clozapine dose with adverse effects; of them, 19 found no correlation between dose and side effects studied [22,35,36,37,38,41,47,48,49,50,51,52,53,54,55,56,57,58,59,60,61]. The rest [34,43,62,63,64,65,66,67] reported positive correlations with side effects, albeit weaker or to a lesser extent than correlations with plasma clozapine levels. The time point in the patients’ treatment with clozapine was variable (initiation of clozapine, maintenance treatment, years after clozapine initiation), and the mode of titration in patients starting therapy ranged from rapid to slow. Differences existed in terms of comedications, which varied from none, in some prospective studies with wash out periods, up to different classes of psychotropics (mood stabilizers, antipsychotics, antidepressants, benzodiazepines). Many, but not all studies, used scales for detecting side effects and for measuring their severity, instead of rating adverse effects as present vs. absent. These data are presented in Supplementary Table S1B, except for comedications, which are reported in the results sections below.

2.2. Method of Plasma Clozapine Measurement

Eleven of the selected research papers do not specify how plasma clozapine concentrations were quantified. In the remaining, which describe a method of clozapine measurement, high-performance liquid chromatography (HPLC) and reversed-phase HPLC (RP-HPLC) combined with ultraviolet (UV) detection were reported as the most common methods. Gas chromatography (GC) was also used. Liquid chromatography (LC), LC coupled with mass spectrometry (LC-MS), LC coupled with tandem mass spectrometry (LC-MS/MS), and gas chromatography-mass spectrometry (GC-MS) were utilized in fewer studies (Table 1).

2.3. Overall or Combined Adverse Effects

Four studies examined associations between clozapine levels and a total number of side effects or combinations of side effects (Table 2, Table S2). Frazier et al. [43] found an association between norclozapine (r = 0.6, p = 0.002), and norclozapine + clozapine levels (r = 0.4, p = 0.002) and the total number of moderate and severe side effects, in a small (n = 6) pediatric sample, at 6 weeks of therapy. Yusufi et al. [56], by applying the Antipsychotic Non-Neurological Side Effects Rating Scale (ANNSERS) to a sample of adult patients at maintenance therapy with clozapine, found that the drug levels correlated weakly with the number of moderate and severe side effects (r = 0.23, p < 0.03) and the total scale score (r = 0.29, p < 0.004). More than three-quarters (77%) of the patients had at least one and 28% had at least three side effects that were rated as moderate or severe. Further, Spina et al. [46], showed a small but not significant increase in the incidence of side effects, in patients with higher clozapine levels (440 ± 125 ng/mL) at 12 weeks of treatment. Another study employing a different scale, the Systematic Assessment for Treatment Emergent Effects (SAFTEE), reported no correlation between total scale score and drug levels [41].

2.4. Nervous System Adverse Effects

2.4.1. Εlectroencephalographic (EEG) Abnormalities and Seizures

Seven studies addressed the issue of electroencephalographic (EEG) changes and/or seizures in relation to clozapine levels. Of these, two [35,47] examined patients at the maintenance phase, a few years after therapy initiation, while all others refer to patients at the early treatment phase (a maximum of 12 weeks). In a prospective, randomized clinical trial (Jadad score = 2) [34], 50 patients with treatment-resistant schizophrenia or schizoaffective disorder, were randomly assigned into three groups according to their clozapine plasma levels: group I: 50–150 ng/mL, group II: 200–300 ng/mL, group III: 350–450 ng/mL. Three patients (6%) exhibited seizures, with two having a prior history of seizures (assigned to group II) and one to group III. After excluding five patients who were on valproate, in the rest of the patients (n = 45) slowing was reported in 53% of them and was significantly more frequent in group III (82%), compared to groups I (20%) and II (26%). Spike/sharp activity (13%) and sleepiness (31%) were not significantly different among groups. EEG changes were not significantly different between responders and non-responders. Sleepiness correlated significantly with slowing, but not with clozapine serum levels, possibly due to a lower sensitivity of nurse observation, according to the authors.
Haring et al. [58] studied the occurrence of EEG changes in a group of 29 hospitalized patients treated with clozapine, in the context of a prospective clozapine drug monitoring program. Patients were classified into two subgroups based on EEG findings: in the first (53% of patients) pathological changes on the EEG were noted, while in the second (47% of patients), minimal or no changes were observed. This difference correlated with marked differences in plasma clozapine levels between groups (81.6 ± 64.6 ng/mL vs. 235.7 ± 169.8 ng/mL for patients with minimal versus pathological changes in EEG, respectively, p = 0.0009). No seizures or severe alterations were observed during the study. Administered dose, duration of treatment, sex, age, and weight had no influence on the induction of pathological EEG patterns. Similar results were found in a more recent retrospective study, where patients with clozapine levels over 600 ng/mL had a higher rate of EEG abnormalities than those below 600 ng/mL (93.8% vs. 65.5%, p = 0.02); again, dose, duration of illness or treatment, age, norclozapine levels did not correlate with EEG abnormalities [47]. In another study [35] (n = 30, chronic schizophrenia) with higher plasma clozapine levels (median: 1076 ng/mL, range: 706 ng/mL–1882 ng/mL), the severity of EEG changes was positively correlated with plasma clozapine but not norclozapine levels. Eighty-three percent of the subjects presented EEG abnormalities, similarly to the higher serum level group (group III) of the study by Freudenreich et al. [34].
In a Chinese study [42] (n = 51), similar incidence of EEG abnormalities (81.58%) was found on week 6 of observation but the severity was not assessed, and no correlation was found between incidence and clozapine levels (range: 100–1220 ng/mL, mean: 470.20 ± 234.23 ng/mL). Centorrino et al. [41] (n = 44, chronic patients) reported the absence of seizures in their sample but no EEG recordings were conducted (median clozapine levels: 291 ng/mL, range: 15–726 ng/mL). Finally, in a pediatric population [40] (n = 54) with childhood-onset schizophrenia (COS) reporting side effects at week 6 of observation, seizures were found in 6%, EEG abnormalities and slowing in 11%, and epileptiform discharges in 11% of study participants. Again, severity was not assessed and correlation with clozapine levels was not found (range of clozapine levels: 455 ± 285.1 ng/mL, and during follow-up: 395.7 ± 206.0 ng/mL).

2.4.2. Cognition

Rajji et al. [59] found that high cognitive impairment as measured by an assessment tool for global cognition was more frequently encountered in patients with higher age, higher clozapine levels (534.7 ± 237.7 ng/mL), and a higher clozapine/norclozapine ratio. Causality could not be established due to several limitations in the study design, but the finding could be supported by the partial agonism exerted by norclozapine on muscarinic receptors, contrary to the antagonistic muscarinic properties of clozapine. In another study, however [68], no significant correlation between plasma clozapine levels and cognitive performance could be established, except for a trend, on executive tests. Similarly, Yusufi et al. [56] found memory problems and lack of concentration in 38% of patients, but no correlation with plasma clozapine levels could be established. The Jadad score for all three studies suggested a low quality of clinical evidence.

2.4.3. Extrapyramidal Side Effects

VanderZwaag et al. [39] reported improvement of bradykinesia/rigidity, tremor, and akathisia, during a 12-week treatment period, irrespective of plasma levels; as for dyskinesia, 80% of patients who had scores >1 on the Abnormal Involuntary Movement Scale (AIMS) showed improvement over time. Liu et al. [82] reported mild rigidity in 8.1% and tremor in 3.2% of patients but as these might have been due to delayed effects of the previous therapy with haloperidol they were not reported as side effects of clozapine (mean levels: 598 ± 314, range 111–1585 ng/mL).
Yusufi et al. [56] found parkinsonism, akathisia, and tardive dyskinesia in 18%, 4%, and 5%, respectively, with no significant correlation with plasma levels (530 ± 370 ng/mL). In a pediatric sample [40] akathisia was reported on week 6 (15%), with no correlation with levels (455 ± 285.1 ng/mL).
A cohort study (n = 37) [83] was conducted for a period of 6 weeks and the mean doses of clozapine differed each week. Similarly, plasma clozapine levels were also different (week 1:450 ng/mL, week 2:650 ng/mL, week 3:450 ng/mL, week 4:575 ng/mL, week 5:575 ng/mL, week 6:663 ng/mL). The study indicates that none of the 7 patients without tardive dyskinesia (TD) developed TD in an average of ~6.9 months and there was no significant increase observed in the Hillside Modified version of the Simpson Dyskinesia Scale (SDS) for the non-TD patients. Nervous system adverse effects in relation to CLOZ levels are shown in Table 3 and Supplemental Table S3.

2.5. Psychiatric Adverse Effects

Obsessive-compulsive symptoms (OCS) in relation to plasma clozapine levels were examined in one retrospective study conducted in Taiwan [60] (n = 102). Thirty-nine patients (38.2%) manifested OCS. In 29 patients (28.4%) OCS was classified as clozapine-induced, with an average latency period of 39.8 ± 22.5 months of time-to-event. Plasma clozapine levels were remarkably higher in patients with OCS than in those without (595.1 ± 364.9 vs. 433.5 ± 252.8 ng/mL, p = 0.001) though no significant difference existed in the daily administered clozapine dose. By a quartile odds ratio comparison, patients with the highest plasma clozapine levels presented a four-fold higher probability of developing OCS.
Moreover, a cross-sectional study of 237 patients where serum clozapine and norclozapine plasma levels were available for 190 subjects, showed that the depression/anxiety factor score (comprising items such as difficulty in concentrating, tension, difficulty remembering things, depression, restlessness, difficulty getting to sleep) was significantly higher in patients with combined clozapine and norclozapine serum concentrations of at least 719 ng/mL (n = 92) compared to patients with lower concentration (n = 95). The correlation was not significant for sedation, dreaming, lack of emotion, and sympatichotonia factor scores [75]. In another study, night-time sleep problems were found in 32% of patients [56]. Mean clozapine levels were 530 ± 370 ng/mL and no correlation with levels was found [56].
In summary, a positive correlation between clozapine plasma levels and OCS has been reported; a correlation with depression/anxiety constellation of symptoms was noted, but the causality remains unclear (Table 3).

2.6. Cardiovascular System

2.6.1. QTc Prolongation

No significant correlation was found in the study of Wong et al. [42], where mean clozapine levels were 470.20 ± 234.23 ng/mL, but a caveat of the work was that baseline measurement was conducted during the tailing off of previous antipsychotic medications that could have an impact on QTc as well. Similarly, Grande et al. [79] found no significant correlation between clozapine levels > 400 ng/mL and QTc prolongation, in a retrospective study of 82 patients. The best predictors of QTc prolongation were QTc before treatment, age, and heart rate > 95 beats/min.

2.6.2. Sudden Death, Myocardial Function, Myocarditis, and Pericarditis

More recently, results of a longitudinal follow-up cohort study in Australia (n = 503) were reported [80]. The endpoints of the study were discontinuation of clozapine, all-cause mortality, sudden death, the incidence of clinical myocarditis, and time to myocarditis. Myocarditis was diagnosed by troponin I elevation and evidence of left ventricular systolic dysfunction. After clozapine initiation, incidence of sudden death was 2% (n = 10, time-to-event: 5 ± 4 years, and of myocarditis 3% (n = 14, time-to-event: 15 ± 7 days). Average plasma clozapine levels for the entire cohort were 475 ± 236 ng/mL compared to 439 ± 198 ng/mL in subjects who suffered a sudden death and 297 ± 152 ng/mL in subjects with myocarditis. Based on these, myocarditis or sudden death cannot directly be linked to clozapine blood levels. Nevertheless, plasma levels found in patients with myocarditis, in relation to the duration of clozapine exposure, point to a rapid mode of dose escalation. On the other hand, sudden death was correlated with recent weight gain and morbid obesity (70% of the patients gained weight) but not with prolonged QTc which was found normal, although ventricular arrhythmias were documented in half of those patients (n = 5).
Echocardiographic measures have been performed in a cohort of 15 patients with schizophrenia, receiving a daily clozapine dose titrated to 100 mg. Titration took place over a period of 4 weeks, starting at 25 mg per day at week 1 and increasing dose weekly by 25 mg increments [76]. The authors reported less efficient left ventricular function, without overt clinical manifestations in 60–80% of the sample, at rather low clozapine and norclozapine plasma levels (124 ± 70.8 ng/mL and 52.3 ± 35.7 ng/mL, respectively). No correlation between echocardiographic findings and drug or metabolite levels could be established.

2.6.3. Pulse and Blood Pressure Irregularities

Oyewumi et al. [64] reported a trend toward autonomic dysregulation during titration of clozapine in the first 8 weeks of therapy (n = 37). Small changes in temperature, pulse rate, and blood pressure, could relate to increased cardiovascular risk. Average plasma clozapine and norclozapine concentration and the norclozapine/clozapine ratio were 63.5 ± 46.0 ng/mL, 38.7 ± 30.8 ng/mL, and 0.65 ± 0.31 ng/mL, respectively, at week 1, and 379.5 ± 156.5 ng/mL, 249.6 ± 94.7 ng/mL and 0.70 ± 0.16 ng/mL, respectively, at week 8. The norclozapine/clozapine ratio was a significant covariate of the erect and supine systolic blood pressure (BP) (p < 0.001) and the standing diastolic BP (p < 0.002). A higher dose of clozapine and higher norclozapine levels were correlated with lower temperatures, (mean temperature was 36.6 ± 0.9 °C and 35.9 ± 0.6 °C at weeks 2 and 7, respectively) and higher norclozapine/clozapine ratio was significantly but weakly related to higher BP measures. Another study [44] on a very small subset of patients (n = 5) with known clozapine levels between 200–300 ng/mL found a positive relationship with severity of orthostatic dysregulation at day 6 (p < 0.05) and day 10 (p < 0.05) of treatment. The mean severity score was higher on day 3, whereas an adaptation occurred after 10–20 days of treatment. In the rest of the studies [35,39,56,82], orthostatic hypotension was found in 5–27% of patients, with no significant correlation with plasma clozapine concentration.
Many studies reported tachycardia (>100 bpm) in 23–51% of patients [35,39,41,56,82]. In one of these [35], in adult patients with chronic schizophrenia (n = 30), increased pulse rate (>80 bpm) was also reported in 73% of the participants in contrast to Spina et al. [46], who reported a much lower rate of 4.4%. Nevertheless, no association with drug levels was found. A more recent study employing 24 h ambulatory ECG recording (Holter) in 30 patients with HR >100 bpm did not find any relationship between clozapine levels and HR variables; HR was consistently very high during day- and night-time. Data concerning patients without baseline tachycardia were not included [48].
Heart rate variability (HRV) correlated negatively with cardiovascular risk. Clozapine levels appeared to inversely correlate with HRV parameters, and this influence was stronger than the influence of age [72]. This finding was later replicated by Eschweiler et al. [73], who reported that all patients with clozapine levels below 350 ng/mL had CV > 3.2%, contrasting patients with higher levels who all had CV < 3.2% (p < 0.001), setting this level as a separating point.
In pediatric populations, tachycardia was reported in 28–67% of patients [40,43] with no apparent correlation to plasma clozapine levels. Furthermore, Sporn et al. [40] reported hypertension (>140/90 mmhg) at a rate of 6% and orthostatic hypotension at a rate of 7%, which again did not present any correlation with plasma drug levels, 6 weeks after treatment initiation. In this study orthostatic hypotension was defined as a fall in systolic/diastolic BP >20/10 mmhg or an increase in heart rate >30 bpm within 3 min of obtaining the upright position.
In summary, QTc prolongation appears independent of plasma clozapine levels. Myocarditis, usually an idiosyncratic reaction, could relate to higher plasma clozapine levels, or rapid escalation of these levels, during initiation of therapy. Sudden death better correlates with other factors rather than plasma clozapine concentration. Importantly, HRV is negatively correlated to plasma clozapine levels. Finally, orthostatic dysregulation or hypotension and temperature dysregulation, but not tachycardia, show a positive correlation with clozapine plasma levels. Studies reporting cardiovascular adverse effects are presented in a tabular form in Table 4 and complementary Table S4.

2.7. Metabolic Adverse Effects

Eleven studies reported metabolic side effects in relation to plasma clozapine levels. Specifically, these studies assessed weight gain, metabolic syndrome, glucose, and lipid abnormalities.

2.7.1. Weight Gain

Three studies evaluated weight gain in relation to clozapine levels and found no significant association [50,63,71]. The sample size in all three studies was small to moderate (n = 61–74) and the duration of clozapine exposure varied from 12 weeks [71] to 16–48 weeks [63] to 6 months [50]. Similar results were obtained in another small pediatric sample [43].
However, a significant positive association between norclozapine levels and weight gain was reported by Lu and coworkers [71] who studied fluvoxamine add-on therapy as a way of reducing this adverse effect. The rationale of this approach is based on the observation that fluvoxamine lowers norclozapine levels. Similarly, norclozapine levels were significantly and positively associated with weight gain in nonsmokers (n = 8) in the study of De Leon et al. [63] after controlling for baseline BMI, sex, and clozapine levels.
Two studies reported the existence of excess body weight or BMI, as common physical features among their subjects and considered them an indirect indication of weight gain [41,51]. However, no direct evaluation of weight gain could be performed since baseline weight was not available. Vasudev et al. [51] found that clozapine levels correlated with metabolic syndrome. The authors calculated an 11% odds ratio increase per 100 ng/mL of clozapine blood level. On the other hand, Anderson et al. [74] reported a positive correlation between clozapine levels and BMI, in a retrospective study investigating sex differences in clozapine levels and metabolic parameters as a primary focus. Females had significantly higher BMI and clozapine levels compared to men.

2.7.2. Dyslipidemia

According to the study by Subramaniam et al. [65], clozapine levels could not predict lipid levels, total cholesterol (TC), or triglyceride (TRG) levels. Along the same line, a prospective observational study that examined the associations between therapeutic response to clozapine and lipid dysregulation in 49 patients [81], showed that increases in TRG levels were associated with clinical response to clozapine treatment (p < 0.001), but plasma clozapine concentrations did not correlate with TRG increase. Similarly, no correlation between TRG and clozapine levels was found in two other studies [71,77]
On the contrary, TRG was found to correlate to plasma clozapine concentration (p = 0.03) in another, cross-sectional study [70]. Hyperlipidemia was found in 40–60%, but again no correlation was found between cholesterol, high-density lipoprotein cholesterol (HDL-C), or LDL cholesterol (LDL-C) and plasma clozapine levels. Anderson et al. [74] found no association between clozapine levels and HDL-C. Lu et al. [71] reported a positive correlation between serum TRG and norclozapine but not clozapine concentration. Lipid elevation was found to associate with clozapine levels and norclozapine/clozapine ratios by Melkersson et al. [45]. Further, TC, but not HDL-C or LDL-C, correlated positively with plasma clozapine levels in a case-control study [77].

2.7.3. Hyperglycemia, Hyperinsulinemia, Insulin Resistance

Hyperglycemia has been found to positively correlate [74] or not related [65,70,71,77] to clozapine plasma levels. In the study of Subramaniam et al. [65], female gender (p = 0.02) and age (p = 0.04) but not clozapine concentrations were found to be significant predictors of glucose levels. Anderson et al. [74] have compared clozapine plasma levels and hyperglycemia between the two sexes and found that women had significantly higher clozapine levels and blood glucose than men, therefore indirectly pointing to a positive correlation between drug levels and levels of glucose. The findings of the two studies should be interpreted with caution because the groups compared differ in race [65] and sex [70]. Lu et al. [71] found that serum glucose was positively correlated with norclozapine, but not clozapine levels.
Insulin levels were positively correlated to clozapine serum concentration [49]. In the study by Melkersson and Dahl [70], hyperinsulinemia was present in 30–60% of patients and hyperglycemia in 10–30%. Plasma levels of insulin and C-peptide were positively correlated with plasma clozapine levels (p = 0.03 and p = 0.04, respectively). Insulin elevation and insulin resistance positive correlation with clozapine levels as well as norclozapine/clozapine ratios were also reported in a later study [45].
All studies, with the exception of one [63], were of low quality.

2.8. Endocrine System

Compared to typical antipsychotics, the effect of clozapine on prolactin (PRL) secretion is 5 to 10 times lower, according to a double-blind dose–response study. For every 100 ng/mL increase in plasma clozapine levels (clozapine plasma levels: 400–1600 ng/mL), average increments in prolactin levels of 0.45 ng/mL in females and 0.15 ng/mL in males were recorded. These effects were more evident in females, but no symptoms associated with hyperprolactinemia were recorded [66]. In another study, menstrual disturbances (period pains, reduced frequency of periods) were not associated with combined clozapine + norclozapine concentrations [75]. A negative correlation between clozapine levels and fT3 (r = −0.385, p = 0.021), but no correlation with TSH, fT4, or PRL, was more recently reported [77].
Studies relevant to metabolic and endocrine system side effects are shown in Table 5 and Supplemental Table S5.

2.9. Gastrointestinal System

Studies relevant to gastrointestinal adverse effects are shown in Table 6 and Supplemental Table S6.

2.9.1. Hypersalivation, Nocturnal Sialorrhea, Drooling

Two studies examined hypersalivation in relation to clozapine levels and failed to establish a significant association. According to the first study, (cross-sectional, n = 44) nocturnal sialorrhea (observed in 80% of the subjects), drooling and parotid swelling were among the most common side effects [41], with no significant correlation between these and mean clozapine and norclozapine levels in serum. In the other study, (prospective, n = 45), hypersalivation was observed in just a small proportion of the cohort (n = 3) [46]. Mean clozapine levels in patients with hypersalivation were higher than those in patients without the side effect (440 ± 125 ng/mL versus 359 ± 205 ng/mL, p = 0.212), but the difference did not reach statistical significance (p = 0.199).

2.9.2. Hepatotoxicity

Hummer et al. [52], in their prospective study, found a significantly positive correlation between plasma clozapine levels (165.4 ± 163.4 ng/mL) and increases in serum glutamic-pyruvic transaminase (SGPT). Increases in SGPT were more prominent during weeks 1–6, and thereafter generally decreased. Yet, three other, cross-sectional, studies [35,65,77], failed to find such an association. Incidence of increased γ-GT was 60% and the incidence of increased AST, ALT, and ALP was 15–25%, irrespective of duration of treatment, clozapine, norclozapine, or norclozapine/clozapine ratio levels [35]. Liver enzyme activity was also not correlated with clozapine serum concentration, in two studies on pediatric samples [40,43] with childhood-onset schizophrenia. In the latter (very small sample, n = 6), one patient demonstrated increased hepatic transaminase concentrations.

2.9.3. Constipation

Centorrino et al. [41], did not find a statistically significant association between constipation and clozapine concentration. A very interesting, cross-sectional study found a strong association between clozapine use and gastrointestinal hypomotility, and, in fact, higher clozapine levels correlated with longer colonic transit times (CTT) [53]. On the other hand, in a study where laxative use served as an indirect marker for constipation [54], it was demonstrated that laxative users had 29% higher norclozapine concentrations compared with non-users. Clozapine levels did not differ significantly between the two groups.

2.10. Hematological System

Four studies examining leucocyte (WBC) and neutrophil (NEU) counts in relation to clozapine levels yielded negative results [35,36,37,69]. No case of agranulocytosis was reported in an open-label study with seven chronic paranoid schizophrenic patients [84]. Similarly no correlation was found with eosinophilia [37], increased erythrocyte folate concentration [35], lymphocyte number [37,77], hemoglobin (HGB) levels and hematocrit [37]. A weak positive correlation was noted between clozapine levels and platelet count (PLT) between week 2 and 4 after clozapine initiation and the clinical significance is unclear [37]; in contrast, Gharab et al. [77] found an inverse correlation between clozapine levels and PLT (r = −0.362, p = 0.025), HGB (r = −0.342, p = 0.025) and NEU (r = −0.385, p = 0.017), during chronic therapy. Red blood cell count (RBC) (p = 0.020), mean corpuscular hemoglobin (MCH) (p = 0.036), HGB (p = 0.015), NEU (p = 0.034), PLT (p = 0.005), mean platelet volume (MPV) (p = 0.003) were significantly lower in clozapine-treated patients compared to controls, whereas mean corpuscular volume (MCV) was significantly higher (p = 0.002). Mauri et al. found a positive correlation between clozapine levels and NEU (r = 0.26, p = 0.001). Still, WBC showed no correlation [38,77]. On the other hand, Vaquero-Baez et al. [67] reported a negative correlation between clozapine levels and WBC (r = −0.725, p = 0.001) and NEU (r = −0.631, p = 0.004).
A few studies examined the correlation of hematological disorders with norclozapine plasma levels. Of these, three reported no correlation to WBC or NEU counts [36,41,67]. Smith et al. [78] found a positive correlation between NEU count and norclozapine as well as norclozapine/clozapine ratio (p = 0.002, p = 0.04, respectively, in mixed-model analyses). Mauri et al. [38] showed that norclozapine was positively (r = 0.20, p = 0.01) and norclozapine/clozapine ratio negatively correlated (r = −0.26, p = 0.002) with NEU count. Turning to pediatric samples, neutropenia, observed in a significant proportion of the sample (6–16.5%) was not correlated to clozapine or norclozapine levels [40,43] (Table 7 and Table S7).

2.11. Genitourinary System

One study [62] that examined anticholinergic side effects in relation to clozapine and resultant antimuscarinic activity serum levels, found no correlation between enuresis with antimuscarinic activity. Other studies of lower quality also do not report any relevant association in adults [41] or in samples of children and adolescents with childhood-onset schizophrenia [40,43] (Table 7 and Supplemental Table S7). Sexual disturbances, namely diminished sexual desire and functional disturbances were influenced by clozapine levels, in males, but not in females; furthermore, such influence was not found for menstrual disturbances, dry vagina, gynecomastia, galactorrhea [57].

2.12. Other Adverse Effects

Clozapine-treated patients had higher anticardiolipin antibodies (aCL) (IgG and IgM isotypes) levels compared to healthy controls while there was a significant positive relationship between serum IgM aCL and serum clozapine level (r = 0.461, p = 0.001) [55] (Table 7). Further, no correlation was noted between B12, Na, K, urea, creatinine levels, and clozapine concentrations [77].

3. Discussion

Usually, clozapine ADRs are type A (augmented) reactions that result from an exaggeration of the drug’s normal pharmacological actions when given at the usual therapeutic dose and are normally dose-dependent. This may be due to elevated plasma levels of clozapine or its active metabolite norclozapine. Nevertheless, some adverse reactions are type B, meaning they are unpredictable, rare, and idiosyncratic reactions, largely independent of plasma drug levels. Table 8 summarizes adverse drug reactions and their relation to plasma clozapine levels.
The interaction of clozapine with various receptor systems in the central nervous system justifies not only its increased effectiveness but also the induction of type A ADRs related to its use. Indeed, its competitive effects on α-adrenergic receptors are linked to the frequent occurrence of nocturnal enuresis in patients, while, a competition specifically of α1 receptors is associated with the occurrence of orthostatic hypotension, sedation, and dizziness [85,86]. Blockade of the α1 receptor in combination with the anticholinergic properties of clozapine contributes to the occurrence of tachycardia [86,87]. Binding to adrenergic α2 and muscarinic M4 receptors contributes to salivation, while binding to muscarinic M3 may be related to insulin regulation and the development of hyperglycemia [86,88]. Similarly, binding to muscarinic M1 receptors is mainly associated with the occurrence of constipation, but also suppression and paralytic ileus [86]. Binding to H1 receptors is also associated with the occurrence of suppression, while blockade of both H1 and serotonin 1A, 2A, and 2C receptors contribute to increased appetite and weight gain [85]. Finally, blockade of D2 receptors may contribute to the development of malignant neuroleptic syndrome and, rarely, motor disorders [85,86].
The induction of clozapine type B ADRs remains a mystery. One such ADR is agranulocytosis which appears even at very low plasma levels of clozapine during the first few days or weeks of its use. In this case, the metabolism of clozapine to nitrenium ion by liver microsomes, peripheral blood neutrophils, and their bone marrow precursors is believed to play a crucial role. This toxic metabolite has been shown to covalently bind to neutrophil proteins and this interaction is believed to trigger toxicity, though the precise mechanism remains unclear [89]. Another potential reason for type B ADRs may be a local build-up of clozapine concentration within certain cells and tissues which may be idiosyncratic and may not correlate directly with plasma levels. This mechanism may explain leukocytopenia in some patients even at very low plasma levels of clozapine [90].
In practical terms, for type A ADRs, TDM can offer quite valuable guidance, aiding dose adjustments for optimal efficacy and safety. For some level-related adverse effects, such as severe constipation or EEG slowing, pointing to some degree of encephalopathy, levels should be lowered, by reducing the administered dose. For those type A ADRs where, although level-related, reducing dose is not possible or mandatory—for example, because efficacy is compromised—or for type B ADRs that are not related to levels, clinical strategies for their management must be a priority. Such situations are, for instance, adding on an SSRI for obsessive compulsive symptoms [91], or advising behavioral measures of the morning activity to counteract sedation [92]. Slow and careful titration is best in all cases, especially for neutropenia and myocarditis, which may involve immune mechanisms [33,93]. Needless to say, the art of the pharmacological treatment of each individual patient rests on the treating clinician, who uses TDM in the context of a unique therapeutic relationship.
Most of the studies in our work suffered significant limitations when evaluated by the Jadad grading system, preventing concrete conclusions. This fact partially mirrors the difficulty of conducting studies designed as randomized and double-blind, since serious ethical issues are raised with randomization and blindness [94]. On the other hand, real-world design of clinical trials can also offer useful insights into various clinical matters, including efficacy in real-world settings, add-on strategies, comorbidities, and drug safety [95].
During our search and clinical evidence evaluation, a considerable heterogeneity among studies was observed with regards to patient demographics (race, gender, age), duration, and type of treatment (chronic or acute phase, initiation, or maintenance treatment, early or late in therapy), use of scales and severity measures for side effect assessment, mode of clozapine dose titration (rapid or slow), etc. All these factors can impact differently adverse effect development, identification, and evaluation, thus influencing outcomes set in different trials. In most studies, there is a great preponderance of men over women, oftentimes higher than three-fold. Gender differences in terms of side effects is an underestimated field, and most studies, with the exception of Anderson et al. [74], do not stratify samples by sex. Age also presents a source of side effect variability, ranging from 21 to 75, with a mean value between 30 and 45 years in adult samples. This is an additional factor affecting side effect profiles since women and the elderly are generally more susceptible to side effects than men or younger people [96,97]. Race is sometimes not reported; in some studies, samples are uniform in terms of race while others have mixed samples, or race differs significantly between samples (i.e., an excess of patients are of a certain race). Notably, ethnic pharmacokinetic differences exist and should be taken into consideration [98]. Moreover, some side effects may be dependent on the duration of the treatment, for example, leukopenia and myocarditis or myocardiopathy are more frequent in the first months of therapy than later [31,99]. Nonetheless, these two studies reported higher rates of adverse effects with the escalation of clozapine plasma levels [40, 43]]. The rate of occurrence of adverse effects may also differ depending on the duration of clozapine exposure which also varied among studies. Moreover, the use of validated instruments for side effect identification and severity evaluation as well as, laboratory biomarkers used in some studies may result in more rigorous detection of adverse effects compared to patients’ self-reports or routine clinical interrogations used in others. We also noted that most studies were conducted in adults with only two studies in pediatric samples, thus limiting our understanding of the role of plasma clozapine levels on adverse effects in children.
It would be desirable to set specific thresholds for each type A ADRs, in order to facilitate an informed decision by the treating psychiatrist. However, from our research, it became apparent that different studies set different thresholds for the same side effect and these thresholds may vary significantly from one another. This could be due to the fact that different investigators employed different methodologies for plasma level determination (as summarized in Table 1) or collected samples at different time points from different populations under different comedications, as described in paragraph 2.1. As a result of this heterogeneity in plasma clozapine level measurements, it would be misleading and largely inaccurate to proceed with threshold recommendations.
To this date, trough plasma levels (Ctrough) of clozapine are used and almost all studies relating plasma clozapine levels to side effects use these metrics. Nonetheless, peak plasma levels, usually around 2–3 h after drug administration may also be valuable since they may relate to the severity of dose-dependent side effects. As a future direction, a more detailed pharmacokinetic evaluation that will include not only trough but peak levels may be more informative. In that case, Ctrough/AUC would be a handy parameter to determine and relate to ADR frequency and severity.

4. Materials and Methods

4.1. Protocol and Registration

The Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) statement recommendations were followed for background, search strategy, methods, results, discussion, and conclusions [100]. Ethical approval was not required for this systematic review since all included data have been previously published with ethical approval.

4.2. Eligibility Criteria

Studies included were conducted in humans, and article language was limited to English. The main screening criterion at all levels of our search was the identification of publications reporting data of clozapine side effects in relation to its plasma/serum levels. Original research papers and case reports were included. Review papers relevant to our main screening criterion were used as a source of additional original research papers by manually searching their reference lists. Studies that used experiments or animals, or did not provide information on side effects in relation to plasma clozapine levels, as well as letters reporting no primary data, conference abstracts, and articles that analyzed drug-drug interactions were excluded. ZD, CAK, and VK performed the literature review and collected all relevant studies. MS, EAK, PG, KEK, and OA participated in the independent appraisal of the collected manuscripts, article selection, and the evaluation of reported data.

4.3. Search Strategy and Study Selection

A literature search in SCOPUS and PubMed databases was conducted from inception to August 2021. In both databases, we used the terms ‘Clozapine Levels’, ‘Side Effects’, ‘Adverse Effects’, ‘Adverse Drug Reaction’, and ‘Toxicity’ in different combinations. No limitation or other filters were applied to the searches. Studies that fulfilled the inclusion criteria, after exclusion of duplicates, were screened at the title and abstract level, and then selected articles were screened at the full-text level.

4.4. Data Collection Process

Each publication was analyzed for its type (i.e., review, original paper, metanalysis, etc.), date of publication, names of authors and their affiliations, quality of evidence according to the Jadad scoring system [101], the country where the study took place, sample size, cohort status, sex, race, mean age, diagnosis, and diagnostic tools, the analytical technique used for clozapine plasma level determination, duration of clozapine administration, mean dose administered, time of clozapine level measurement (peak or trough), diagnoses and comedications, the reported plasma levels, the recorded side effects and the assessment tools for identifying side effects, and the main conclusions of each publication.

4.5. Outcomes

Side effects in relation to plasma clozapine levels were categorized and presented by the physiological organ system.

4.6. Evaluation of Study Quality Using the Jadad Scoring System

The Jadad score [101] was used for the assessment of the quality of evidence of each study as low or high. This is a simplified quality assessment tool that allows easy and quick evaluation of the quality of clinical evidence. Briefly, the assessment was based on the type and protocol of randomization and blinding procedures and the description and accountability of withdrawals and dropouts. Studies with a score equal to or lower than 2 were classified as low quality, while studies with a score equal to or greater than 3 were characterized as high quality. The classification of evidence based on the Jadad score is briefly described in Table 9.

5. Conclusions

In conclusion, monitoring clozapine and its metabolites plasma levels is a reasonable strategy for personalization of treatment, improvement of drug safety, and enhancement of tolerance, in TRS patients. This may further contribute to overcoming the underutilization of clozapine, a drug with superior antipsychotic efficacy and the only effective in TRS. There is plenty of room for more and better-designed studies, emphasizing more the influences of gender, age, race, specific diagnoses, comorbidities, genomics, and other factors. Further exploring the intriguing interrelationship of clozapine plasma levels with inflammation, and the effects of the drug on the cardiac muscle, the autonomous and endocrine systems, the gastrointestinal tract, and energy metabolism, are important directions for future research.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/ph15070817/s1, Table S1A: Case Reports referring to adverse effects of clozapine in relation to drug levels; Tables S1B–S7: Additional data from all included studies (cited in the main manuscript). References [90,102,103,104,105,106,107,108,109,110,111,112,113] are cited in the supplementary materials.

Author Contributions

Conceptualization, M.S., K.E.K. and O.A.; methodology, M.S., K.E.K. and O.A.; software, E.A.K., V.K. and Z.D.; validation, M.S., K.E.K. and O.A.; formal analysis, Z.D., E.A.K., V.K. and M.S.; investigation, Z.D., E.A.K., V.K. and C.-A.K.; resources, K.E.K. and O.A.; data curation, V.K., E.A.K., Z.D., M.S. and P.G.; writing—original draft preparation, E.A.K., Z.D., V.K., M.S. and O.A.; writing—review and editing, M.S., O.A. and K.E.K.; visualization, Z.D. and K.E.K.; supervision, K.E.K.; project administration, O.A.; funding acquisition, K.E.K. and M.S. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded in part by the project “INSPIRED” (MIS 5002550), under the Action “Reinforcement of the Research and Innovation Infrastructure”, funded by the Operational Program “Competitiveness, Entrepreneurship and Innovation” (NSRF 2014–2020).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Miles, S.H. The Hippocratic Oath and the Ethics of Medicine; Oxford University Press: Oxford, UK, 2005; 232p. [Google Scholar]
  2. Grandjean, P. Paracelsus Revisited: The Dose Concept in a Complex World. Basic Clin. Pharmacol. Toxicol. 2016, 119, 126. [Google Scholar] [CrossRef]
  3. Hayes, R.D.; Downs, J.; Chang, C.K.; Jackson, R.G.; Shetty, H.; Broadbent, M.; Hotopf, M.; Stewart, R. The effect of clozapine on premature mortality: An assessment of clinical monitoring and other potential confounders. Schizophr. Bull. 2015, 41, 644–655. [Google Scholar] [CrossRef] [Green Version]
  4. Cho, J.; Hayes, R.D.; Jewell, A.; Kadra, G.; Shetty, H.; MacCabe, J.H.; Downs, J. Clozapine and all-cause mortality in treatment-resistant schizophrenia: A historical cohort study. Acta Psychiatr. Scand. 2019, 139, 237–247. [Google Scholar] [CrossRef] [PubMed]
  5. Bente, D.; Engelmeier, M.P.; Heinrich, K.; Hippius, H.; Schmitt, W. Clinical studies with a neuroleptically active dibenzothiazepine derivative. Arzneim.-Forsch. Drug Res. 1966, 16, 314–316. [Google Scholar]
  6. Ereshefsky, L.; Watanabe, M.D.; Tran-Johnson, T.K. Clozapine: An atypical antipsychotic agent. Clin. Pharm. 1989, 8, 691–709. [Google Scholar] [PubMed]
  7. Shen, W.W. A history of antipsychotic drug development. Compr. Psychiatry 1999, 40, 407–414. [Google Scholar] [CrossRef]
  8. Lobos, C.A.; Komossa, K.; Rummel-Kluge, C.; Hunger, H.; Schmid, F.; Schwarz, S.; Leucht, S. Clozapine versus other atypical antipsychotics for schizophrenia. Cochrane Database Syst. Rev. 2010, 83, CD006633. [Google Scholar] [CrossRef] [Green Version]
  9. Huhn, M.; Nikolakopoulou, A.; Schneider-Thoma, J.; Krause, M.; Samara, M.; Peter, N.; Arndt, T.; Bäckers, L.; Rothe, P.; Cipriani, A.; et al. Comparative efficacy and tolerability of 32 oral antipsychotics for the acute treatment of adults with multi-episode schizophrenia: A systematic review and network meta-analysis. Lancet 2019, 394, 939–951. [Google Scholar] [CrossRef] [Green Version]
  10. Howes, O.D.; McCutcheon, R.; Agid, O.; De Bartolomeis, A.; Van Beveren, N.J.M.; Birnbaum, M.L.; Bloomfield, M.A.P.; Bressan, R.A.; Buchanan, R.W.; Carpenter, W.T.; et al. Treatment-ResistantSchizophrenia: Treatment Response and Resistance in Psychosis (TRRIP) Working Group Consensus Guidelines on Diagnosis and Terminology. Am. J. Psychiatry 2017, 174, 216–229. [Google Scholar] [CrossRef]
  11. Kane, J.M.; Agid, O.; Baldwin, M.L.; Howes, O.; Lindenmayer, J.P.; Marder, S.; Olfson, M.; Potkin, S.G.; Correll, C.U. Clinical Guidance on the Identification and Management of Treatment-Resistant Schizophrenia. J. Clin. Psychiatry 2019, 80, 2783. [Google Scholar] [CrossRef] [Green Version]
  12. Rubio, J.M.; Kane, J.M. How and when to use clozapine. Acta Psychiatr. Scand. 2020, 141, 178–189. [Google Scholar] [CrossRef]
  13. WHOCC—ATC/DDD Index. Available online: https://www.whocc.no/atc_ddd_index/ (accessed on 20 June 2022).
  14. Daskalakis, Z.J.; George, T.P. Clozapine, GABAB, and the treatment of resistant schizophrenia. Clin. Pharmacol. Ther. 2009, 86, 442–446. [Google Scholar] [CrossRef]
  15. Woods, A.S. The dopamine D4 receptor, the ultimate disordered protein. J. Recept. Signal Transduct. Res. 2010, 30, 331. [Google Scholar] [CrossRef] [Green Version]
  16. Nair, P.C.; McKinnon, R.A.; Miners, J.O.; Bastiampillai, T. Binding of clozapine to the GABA B receptor: Clinical and structural insights. Mol. Psychiatry 2020, 25, 1910–1919. [Google Scholar] [CrossRef]
  17. Caruso, G.; Grasso, M.; Fidilio, A.; Tascedda, F.; Drago, F.; Caraci, F. Antioxidant properties of second-generation antipsychotics: Focus on microglia. Pharmaceuticals 2020, 13, 457. [Google Scholar] [CrossRef]
  18. Young, C.R.; Bowers, M.B.; Manure, C.M. Management of the Adverse Effects of Clozapine. Schizophr. Bull. 1998, 24, 381–390. [Google Scholar] [CrossRef] [Green Version]
  19. Pirmohamed, M.; Williams, D.; Madden, S.; Templeton, E.; Park, B.K. Metabolism and bioactivation of clozapine by human liver in vitro. J. Pharmacol. Exp. Ther. 1995, 272, 984–990. [Google Scholar]
  20. Dragovic, S.; Gunness, P.; Ingelman-Sundberg, M.; Vermeulen, N.P.E.; Commandeur, J.N.M. Characterization of human cytochrome P450s involved in the bioactivation of clozapines. Drug Metab. Dispos. 2013, 41, 651–658. [Google Scholar] [CrossRef] [Green Version]
  21. Tóth, K.; Csukly, G.; Sirok, D.; Belic, A.; Kiss, Á.; Háfra, E.; Déri, M.; Menus, Á.; Bitter, I.; Monostory, K. Potential Role of Patients’ CYP3A-Status in Clozapine Pharmacokinetics. Int. J. Neuropsychopharmacol. 2017, 20, 529–537. [Google Scholar] [CrossRef] [Green Version]
  22. Brandl, E.J.; Chowdhury, N.I.; Tiwari, A.K.; Lett, T.A.P.; Meltzer, H.Y.; Kennedy, J.L.; Müller, D.J. Genetic variation in CYP3A43 is associated with response to antipsychotic medication. J. Neural Transm. 2015, 122, 29–34. [Google Scholar] [CrossRef]
  23. Zhang, W.V.; D’Esposito, F.; Edwards, R.J.; Ramzan, I.; Murray, M. Interindividual variation in relative CYP1A2/3A4 phenotype influences susceptibility of clozapine oxidation to cytochrome P450-specific inhibition in human hepatic microsomes. Drug Metab. Dispos. 2008, 36, 2547–2555. [Google Scholar] [CrossRef] [PubMed]
  24. Olesen, O.V.; Linnet, K. Contributions of five human cytochrome P450 isoforms to the N-demethylation of clozapine in vitro at low and high concentrations. J. Clin. Pharmacol. 2001, 41, 823–832. [Google Scholar] [CrossRef] [PubMed]
  25. Georgiou, R.; Lamnisos, D.; Giannakou, K. Anticholinergic Burden and Cognitive Performance in Patients with Schizophrenia: A Systematic Literature Review. Front. Psychiatry 2021, 12, 779607. [Google Scholar] [CrossRef] [PubMed]
  26. Olsson, E.; Edman, G.; Bertilsson, L.; Hukic, D.S.; Lavebratt, C.; Eriksson, S.V.; Ösby, U. Genetic and clinical factors affecting plasma clozapine concentration. Prim. Care Companion J. Clin. Psychiatry 2015, 17. [Google Scholar] [CrossRef] [Green Version]
  27. Albitar, O.; Harun, S.N.; Zainal, H.; Ibrahim, B.; Maisharah, S.; Ghadzi, S.; Baynes, R.E. Population Pharmacokinetics of Clozapine: A Systematic Review. BioMed Res. Int. 2020, 2020, 9872936. [Google Scholar] [CrossRef]
  28. Suzuki, T.; Uchida, H.; Watanabe, K.; Kashima, H. Factors associated with response to clozapine in schizophrenia: A review. Psychopharmacol. Bull. 2011, 44, 32–60. [Google Scholar]
  29. Greenwood-Smith, C.; Lubman, D.I.; Castle, D.J. Serum clozapine levels: A review of their clinical utility. J. Psychopharmacol. 2003, 17, 234–238. [Google Scholar] [CrossRef]
  30. Tan, M.S.A.; Honarparvar, F.; Falconer, J.R.; Parekh, H.S.; Pandey, P.; Siskind, D.J. A systematic review and meta-analysis of the association between clozapine and norclozapine serum levels and peripheral adverse drug reactions. Psychopharmacology 2021, 238, 615–637. [Google Scholar] [CrossRef]
  31. Myles, N.; Myles, H.; Xia, S.; Large, M.; Kisely, S.; Galletly, C.; Bird, R.; Siskind, D. Meta-analysis examining the epidemiology of clozapine-associated neutropenia. Acta Psychiatr. Scand. 2018, 138, 101–109. [Google Scholar] [CrossRef]
  32. Varma, S.; Bishara, D.; Besag, F.M.C.; Taylor, D. Clozapine-related EEG changes and seizures: Dose and plasma-level relationships. Ther. Adv. Psychopharmacol. 2011, 1, 47–66. [Google Scholar] [CrossRef] [Green Version]
  33. De Leon, J.; Ruan, C.J.; Schoretsanitis, G.; De Las Cuevas, C. A Rational Use of Clozapine Based on Adverse Drug Reactions, Pharmacokinetics, and Clinical Pharmacopsychology. Psychother. Psychosom. 2020, 89, 200–214. [Google Scholar] [CrossRef]
  34. Freudenreich, O.; Weiner, R.D.; McEvoy, J.P. Clozapine-induced electroencephalogram changes as a function of clozapine serum levels. Biol. Psychiatry 1997, 42, 132–137. [Google Scholar] [CrossRef]
  35. Olesen, O.V.; Thomsen, K.; Jensen, P.N.; Rosenberg, R.; Wulff, C.H.; Rasmussen, N.A.; Refshammer, C.; Bysted, M.; Sørensen, J.; Christensen, J. Clozapine serum levels and side effects during steady state treatment of schizophrenic patients: A cross-sectional study. Psychopharmacology 1995, 117, 371–378. [Google Scholar] [CrossRef]
  36. Combs, M.D.; Perry, P.J.; Bever, K.A. N-desmethylclozapine, an insensitive marker of clozapine-induced agranulocytosis and granulocytopenia. Pharmacotherapy 1997, 17, 1300–1304. [Google Scholar] [CrossRef]
  37. Kola Oyewumi, L.; Cernovsky, Z.Z.; Freeman, D.J.; Streiner, D.L. Relation of blood counts during clozapine treatment to serum concentrations of clozapine and nor-clozapine. Can. J. Psychiatry 2002, 47, 257–261. [Google Scholar] [CrossRef] [Green Version]
  38. Mauri, M.C.; Rudelli, R.; Bravin, S.; Gianetti, S.; Giuliani, E.; Guerrini, A.; Orlandi, R.; Invernizzi, G. Clozapine metabolism rate as a possible index of drug-induced granulocytopenia. Psychopharmacology 1998, 137, 341–344. [Google Scholar] [CrossRef]
  39. VanderZwaag, C.; McGee, M.; McEvoy, J.P.; Freudenreich, O.; Wilson, W.H.; Cooper, T.B. Response of patients with treatment-refractory schizophrenia to clozapine within three serum level ranges. Am. J. Psychiatry 1996, 153, 1579–1584. [Google Scholar] [CrossRef]
  40. Sporn, A.L.; Vermani, A.; Greenstein, D.K.; Bobb, A.J.; Spencer, E.P.; Clasen, L.S.; Tossell, J.W.; Stayer, C.C.; Gochman, P.A.; Lenane, M.C.; et al. Clozapine treatment of childhood-onset schizophrenia: Evaluation of effectiveness, adverse effects, and long-term outcome. J. Am. Acad. Child Adolesc. Psychiatry 2007, 46, 1349–1356. [Google Scholar] [CrossRef]
  41. Centorrino, F.; Baldessarini, R.J.; Kando, J.C.; Frankenburg, F.; Volpicelli, S.A.; Flood, J.G. Clozapine and metabolites: Concentrations in serum and clinical findings during treatment of chronically psychotic patients. J. Clin. Psychopharmacol. 1994, 14, 119–125. [Google Scholar] [CrossRef]
  42. Oi-Yin Wong, J.; Leung, S.P.; Mak, T.; Man-Kin Ng, R.; Chan, K.T.; Hon-Kee Cheung, H.; Choi, W.K.; Lai, J.; Wai-Kiu Tsang, A. Plasma clozapine levels and clinical response in treatment-refractory Chinese schizophrenic patients. Prog. Neuro-Psychopharmacol. Biol. Psychiatry 2006, 30, 251–264. [Google Scholar] [CrossRef]
  43. Frazier, J.A.; Cohen, L.G.; Jacobsen, L.; Grothe, D.; Flood, J.; Baldessarini, R.J.; Piscitelli, S.; Kim, G.S.; Rapoport, J.L. Clozapine pharmacokinetics in children and adolescents with childhood-onset schizophrenia. J. Clin. Psychopharmacol. 2003, 23, 87–91. [Google Scholar] [CrossRef]
  44. Ackenheil, M. Clozapine—Pharmacokinetic investigations and biochemical effects in man. Psychopharmacology 1989, 99, S32–S37. [Google Scholar] [CrossRef]
  45. Melkersson, K.I.; Scordo, M.G.; Gunes, A.; Dahl, M.L. Impact of CYP1A2 and CYP2D6 polymorphisms on drug metabolism and on insulin and lipid elevations and insulin resistance in clozapine-treated patients. J. Clin. Psychiatry 2007, 68, 697–704. [Google Scholar] [CrossRef]
  46. Spina, E.; Avenoso, A.; Facciolà, G.; Scordo, M.G.; Ancione, M.; Madia, A.G.; Ventimiglia, A.; Perucca, E. Relationship between plasma concentrations of clozapine and norclozapine and therapeutic response in patients with schizophrenia resistant to conventional neuroleptics. Psychopharmacology 2000, 148, 83–89. [Google Scholar] [CrossRef]
  47. Kim, H.S.; Youn, T.; Kim, S.H.; Jeong, S.H.; Jung, H.Y.; Jeong, S.W.; Kim, K.K.; Kim, Y.S.; Chung, I.W. Association between electroencephalogram changes and plasma clozapine levels in clozapine-treated patients. Int. Clin. Psychopharmacol. 2019, 34, 131–137. [Google Scholar] [CrossRef]
  48. Nilsson, B.M.; Lindström, L.; Mohsen, I.; Holmlöv, K.; Bodén, R. Persistent tachycardia in clozapine treated patients: A 24-hour ambulatory electrocardiogram study. Schizophr. Res. 2018, 199, 403–406. [Google Scholar] [CrossRef]
  49. Melkersson, K.I.; Hulting, A.L.; Brismar, K.E. Different influences of classical antipsychotics and clozapine on glucose-insulin homeostasis in patients with schizophrenia or related psychoses. J. Clin. Psychiatry 1999, 60, 783–791. [Google Scholar] [CrossRef]
  50. Meltzer, H.Y.; Perry, E.; Jayathilake, K. Clozapine-induced weight gain predicts improvement in psychopathology. Schizophr. Res. 2003, 59, 19–27. [Google Scholar] [CrossRef]
  51. Vasudev, K.; Choi, Y.H.; Norman, R.; Kim, R.B.; Schwarz, U.I. Genetic Determinants of Clozapine-Induced Metabolic Side Effects. Can. J. Psychiatry. 2017, 62, 138–149. [Google Scholar] [CrossRef] [Green Version]
  52. Hummer, M.; Kurz, M.; Kurzthaler, I.; Oberbauer, H.; Miller, C.; Wolfgang Fleischhacker, W. Hepatotoxicity of clozapine. J. Clin. Psychopharmacol. 1997, 17, 314–317. [Google Scholar] [CrossRef]
  53. Every-Palmer, S.; Nowitz, M.; Stanley, J.; Grant, E.; Huthwaite, M.; Dunn, H.; Ellis, P.M. Clozapine-treated Patients Have Marked Gastrointestinal Hypomotility, the Probable Basis of Life-threatening Gastrointestinal Complications: A Cross Sectional Study. EBioMedicine 2016, 5, 125–134. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Bailey, L.; Varma, S.; Ahmad, N.; Gee, S.; Taylor, D.M. Factors predicting use of laxatives in outpatients stabilized on clozapine. Ther. Adv. Psychopharmacol. 2015, 5, 256–262. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Shen, H.; Li, R.; Xiao, H.; Zhou, Q.; Cui, Q.; Chen, J. Higher serum clozapine level is associated with increased antiphospholipid antibodies in schizophrenia patients. J. Psychiatr. Res. 2009, 43, 615–619. [Google Scholar] [CrossRef] [PubMed]
  56. Yusufi, B.; Mukherjee, S.; Flanagan, R.; Paton, C.; Dunn, G.; Page, E.; Barnes, T.R.E. Prevalence and nature of side effects during clozapine maintenance treatment and the relationship with clozapine dose and plasma concentration. Int. Clin. Psychopharmacol. 2007, 22, 238–243. [Google Scholar] [CrossRef]
  57. Hummer, M.; Kemmler, G.; Kurz, M.; Kurzthaler, I.; Oberbauer, H.; Fleischhacker, W.W. Sexual disturbances during clozapine and haloperidol treatment for schizophrenia. Am. J. Psychiatry 1999, 156, 631–633. [Google Scholar] [CrossRef]
  58. Haring, C.; Neudorfer, C.; Schwitzer, J.; Hummer, M.; Saria, A.; Hinterhuber, H.; Fleischhacker, W.W. EEG alterations in patients treated with clozapine in relation to plasma levels. Psychopharmacology 1994, 114, 97–100. [Google Scholar] [CrossRef]
  59. Rajji, T.K.; Uchida, H.; Ismail, Z.; Ng, W.; Mamo, D.C.; Remington, G.; Pollock, B.G.; Mulsant, B.H. Clozapine and global cognition in schizophrenia. J. Clin. Psychopharmacol. 2010, 30, 431–436. [Google Scholar] [CrossRef]
  60. Lin, S.K.; Su, S.F.; Pan, C.H. Higher plasma drug concentration in clozapine-treated schizophrenic patients with side effects of obsessive/compulsive symptoms. Ther. Drug Monit. 2006, 28, 303–307. [Google Scholar] [CrossRef]
  61. Pinto, D.; Pagnamenta, A.T.; Klei, L.; Anney, R.; Merico, D.; Regan, R.; Conroy, J.; Magalhaes, T.R.; Correia, C.; Abrahams, B.S.; et al. Functional Impact of Global Rare Copy Number Variation in Autism Spectrum Disorder. Nature 2010, 466, 368. [Google Scholar] [CrossRef] [Green Version]
  62. De Leon, J.; Odom-White, A.; Josiassen, R.C.; Diaz, F.J.; Cooper, T.B.; Simpson, G.M. Serum antimuscarinic activity during clozapine treatment. J. Clin. Psychopharmacol. 2003, 23, 336–341. [Google Scholar] [CrossRef]
  63. De Leon, J.; Diaz, F.J.; Josiassen, R.C.; Cooper, T.B.; Simpson, G.M. Weight gain during a double-blind multidosage clozapine study. J. Clin. Psychopharmacol. 2007, 27, 22–27. [Google Scholar] [CrossRef]
  64. Oyewumi, L.K.; Cernovsky, Z.Z.; Freeman, D.J. Autonomic signs and dosing during the initial stages of clozapine therapy. Med. Sci. Monit. 2004, 10, 19–23. [Google Scholar]
  65. Subramaniam, M.; Ng, C.; Chong, S.-A.; Mahendran, R.; Lambert, T.; Pek, E.; Huak, C.Y. Metabolic differences between Asian and Caucasian patients on clozapine treatment. Hum. Psychopharmacol. Clin. Exp. 2007, 22, 217–222. [Google Scholar] [CrossRef]
  66. De Leon, J.; Diaz, F.J.; Josiassen, R.C.; Simpson, G.M. Possible individual and gender differences in the small increases in plasma prolactin levels seen during clozapine treatment. Eur. Arch. Psychiatry Clin. Neurosci. 2004, 254, 318–325. [Google Scholar] [CrossRef]
  67. Vaquero-Baez, M.; Diáz-Ruíz, A.; Tristán-López, L.; Avinã-Cervantes, C.; Torner, C.; Ramírez-Bermúdez, J.; Montes, S.; Riós, C. Clozapine and desmethylclozapine: Correlation with neutrophils and leucocytes counting in Mexican patients with schizophrenia. BMC Psychiatry 2019, 19, 295. [Google Scholar] [CrossRef] [Green Version]
  68. Carceller-Sindreu, M.; Portella, M.J.; Carmona, C.; Rametti, G.; Puigdemont, D.; Figueras, M.; Fernández-Vidal, A.; Villalta, L.; Alvarez, E. Neuropsychological effects of maintenance treatment with Clozapine in treatment-resistant psychotic disorder. Actas Esp. Psiquiatr. 2014, 42, 68–73. [Google Scholar]
  69. Hummer, M.; Kurz, M.; Barnas, C.; Saria, A.; Fleischhacker, W.W. Clozapine-induced transient white blood count disorders. J. Clin. Psychiatry 1994, 55, 429–432. [Google Scholar]
  70. Melkersson, K.I.; Dahl, M.L. Relationship between levels of insulin or triglycerides and serum concentrations of the atypical antipsychotics clozapine and olanzapine in patients on treatment with therapeutic doses. Psychopharmacology 2003, 170, 157–166. [Google Scholar] [CrossRef]
  71. Lu, M.L.; Lane, H.Y.; Lin, S.K.; Chen, K.P.; Chang, W.H. Adjunctive fluvoxamine inhibits clozapine-related weight gain and metabolic disturbances. J. Clin. Psychiatry 2004, 65, 766–771. [Google Scholar] [CrossRef]
  72. Rechlin, T.; Beck, G.; Weis, M.; Kaschka, W.P. Correlation between plasma clozapine concentration and heart rate variability in schizophrenic patients. Psychopharmacology 1998, 135, 338–341. [Google Scholar] [CrossRef]
  73. Eschweiler, G.W.; Bartels, M.; Längle, G.; Wild, B.; Gaertner, I.; Nickola, M. Heart-rate variability (HRV) in the ECG trace of routine EEGs: Fast monitoring for the anticholinergic effects of clozapine and olanzapine? Pharmacopsychiatry 2002, 35, 96–100. [Google Scholar] [CrossRef]
  74. Anderson, S.G.; Livingston, M.; Couchman, L.; Smith, D.J.; Connolly, M.; Miller, J.; Flanagan, R.J.; Heald, A.H. Sex differences in plasma clozapine and norclozapine concentrations in clinical practice and in relation to body mass index and plasma glucose concentrations: A retrospective survey. Ann. Gen. Psychiatry 2015, 14, 39. [Google Scholar] [CrossRef]
  75. Seppälä, N.; Leinonen, E.; Viikki, M.; Solismaa, A.; Nuolivirta, T.; Kampman, O. Factors associated with subjective side-effects during clozapine treatment. Nord. J. Psychiatry 2015, 69, 161–166. [Google Scholar] [CrossRef]
  76. Curto, M.; Comparelli, A.; Ciavarella, G.M.; Gasperoni, C.; Lionetto, L.; Corigliano, V.; Uccellini, A.; Mancinelli, I.; Ferracuti, S.; Girardi, P.; et al. Impairment of left ventricular function early in treatment with clozapine: A preliminary study. Int. Clin. Psychopharmacol. 2015, 30, 282–289. [Google Scholar] [CrossRef]
  77. Kamil Gharab, K.M.; Onmaz, D.E.; Abusoglu, S.; Aydin, M.; Sivrikaya, A.; Tok, O.; Abusoglu, G.; Unlu, A. The relationship between serum clozapine concentrations and hematological parameters by a validated mass spectrometric method. J. Pharm. Biomed. Anal. 2020, 180, 113056. [Google Scholar] [CrossRef]
  78. Smith, R.L.; Haslemo, T.; Andreassen, O.A.; Eliasson, E.; Dahl, M.L.; Spigset, O.; Molden, E. Correlation between Serum Concentrations of N-Desmethylclozapine and Granulocyte Levels in Patients with Schizophrenia: A Retrospective Observational Study. CNS Drugs 2017, 31, 991–997. [Google Scholar] [CrossRef]
  79. Grande, I.; Pons, A.; Baeza, I.; Torras, Ú.; Bernardo, M. QTc prolongation: Is clozapine safe? Study of 82 cases before and after clozapine treatment. Hum. Psychopharmacol. 2011, 26, 397–403. [Google Scholar] [CrossRef]
  80. Khan, A.A.; Ashraf, A.; Baker, D.; Al-Omary, M.S.; Savage, L.; Ekmejian, A.; Singh, R.S.H.; Brienesse, S.; Majeed, T.; Gordon, T.; et al. Clozapine and incidence of myocarditis and sudden death—Long term Australian experience. Int. J. Cardiol. 2017, 238, 136–139. [Google Scholar] [CrossRef]
  81. Lally, J.; Gallagher, A.; Bainbridge, E.; Avalos, G.; Ahmed, M.; McDonald, C. Increases in triglyceride levels are associated with clinical response to clozapine treatment. J. Psychopharmacol. 2013, 27, 401–403. [Google Scholar] [CrossRef]
  82. Liu, H.C.; Chang, W.H.; Wei, F.C.; Lin, S.K.; Lin, S.K.; Jann, M.W. Monitoring of plasma clozapine levels and its metabolites in refractory schizophrenic patients. Ther. Drug Monit. 1996, 18, 200–207. [Google Scholar] [CrossRef]
  83. Lieberman, J.; Johns, C.; Cooper, T.; Pollack, S.; Kane, J. Clozapine pharmacology and tardive dyskinesia. Psychopharmacology 1989, 99, S54–S59. [Google Scholar] [CrossRef] [PubMed]
  84. Aymard, N.; Baldacci, C.; Leyris, A.; Smagghe, P.O.; Tribolet, S.; Vacheron, M.N.; Viala, A.; Caroli, F. Neuroleptic-resistant schizophrenic patients treated by clozapine: Clinical evolution, plasma and red blood cell clozapine and desmethylclozapine levels. Therapie 1997, 52, 227–232. [Google Scholar] [PubMed]
  85. Fitzsimons, J.; Berk, M.; Lambert, T.; Bourin, M.; Dodd, S. A review of clozapine safety. Expert Opin. Drug Saf. 2005, 4, 731–744. [Google Scholar] [CrossRef] [PubMed]
  86. Stahl, S.M. Prescriber’s Guide: Stahl’s Essential Psychopharmacology, 6th ed.; Cambridge University Press: Cambridge, UK, 2017. [Google Scholar]
  87. Rechlin, T.; Claus, D.; Weis, M. Heart rate variability in schizophrenic patients and changes of autonomic heart rate parameters during treatment with clozapine. Biol. Psychiatry 1994, 35, 888–892. [Google Scholar] [CrossRef]
  88. Davydov, L.; Botts, S.R. Clozapine-induced hypersalivation. Ann. Pharmacother. 2000, 662. [Google Scholar] [CrossRef]
  89. Pirmohamed, M.; Park, K. Mechanism of clozapine-induced agranulocytosis: Current status of research and implications for drug development. CNS Drugs 1997, 7, 139–158. [Google Scholar] [CrossRef]
  90. Bergemann, N.; Abu-Tair, F.; Aderjan, R.; Kopitz, J. High clozapine concentrations in leukocytes in a patient who developed leukocytopenia. Prog. Neuropsychopharmacol. Biol. Psychiatry 2007, 31, 1068–1071. [Google Scholar] [CrossRef]
  91. Kim, D.D.; Barr, A.M.; White, R.F.; Honer, W.G.; Procyshyn, R.M. Clozapine-induced obsessive-compulsive symptoms: Mechanisms and treatment. J. Psychiatry Neurosci. 2019, 44, 71–72. [Google Scholar] [CrossRef]
  92. Citrome, L.; McEvoy, J.P.; Saklad, S.R. A guide to the management of clozapine-related tolerability and safety concerns. Clin. Schizophr. Relat. Psychoses 2016, 10, 163–177D. [Google Scholar] [CrossRef]
  93. Wiciński, M.; Wȩclewicz, M.M. Clozapine-induced agranulocytosis/granulocytopenia: Mechanisms and monitoring. Curr. Opin. Hematol. 2018, 25, 22–28. [Google Scholar] [CrossRef]
  94. Mann, C.J. Observational research methods. Research design II: Cohort, cross sectional, and case-control studies. Emerg. Med. J. 2003, 20, 54–60. [Google Scholar] [CrossRef]
  95. Verster, J.C.; van de Loo, A.J.A.E.; Adams, S.; Stock, A.K.; Benson, S.; Scholey, A.; Alford, C.; Bruce, G. Advantages and Limitations of Naturalistic Study Designs and Their Implementation in Alcohol Hangover Research. J. Clin. Med. 2019, 8, 2160. [Google Scholar] [CrossRef] [Green Version]
  96. Whitley, H.P. Sex-Based Differences in Drug Activity. Am. Fam. Physician 2009, 80, 1254–1258. [Google Scholar]
  97. Lavan, A.H.; Gallagher, P. Predicting risk of adverse drug reactions in older adults. Ther. Adv. Drug Saf. 2016, 7, 11. [Google Scholar] [CrossRef]
  98. Kim, K.; Johnson, J.A.; Derendorf, H. Differences in Drug Pharmacokinetics Between East Asians and Caucasians and the Role of Genetic Polymorphisms. J. Clin. Pharmacol. 2004, 44, 1083–1105. [Google Scholar] [CrossRef]
  99. Siskind, D.; Sidhu, A.; Cross, J.; Chua, Y.T.; Myles, N.; Cohen, D.; Kisely, S. Systematic review and meta-analysis of rates of clozapine-associated myocarditis and cardiomyopathy. Aust. N. Z. J. Psychiatry 2020, 54, 467–481. [Google Scholar] [CrossRef]
  100. Page, M.J.; McKenzie, J.E.; Bossuyt, P.M.; Boutron, I.; Hoffmann, T.C.; Mulrow, C.D.; Shamseer, L.; Tetzlaff, J.M.; Akl, E.A.; Brennan, S.E.; et al. The PRISMA 2020 statement: An updated guideline for reporting systematic reviews. BMJ 2021, 372, n71. [Google Scholar] [CrossRef]
  101. Jadad, A.R.; Moore, R.A.; Carroll, D.; Jenkinson, C.; Reynolds, D.J.M.; Gavaghan, D.J.; McQuay, H.J. Assessing the quality of reports of randomized clinical trials: Is blinding necessary? Control. Clin. Trials 1996, 17, 1–12. [Google Scholar] [CrossRef]
  102. Simpson, G.M.; Cooper, T.A. Clozapine Plasma Levels and Convulsions. Am. J. Psychiatry 1978, 135, 99–100. [Google Scholar] [CrossRef]
  103. Khan, A.Y.; Preskorn, S.H. Examining Concentration-Dependent Toxicity of Clozapine: Role of Therapeutic Drug Monitoring. J. Psychiatr. Pract. 2005, 11, 289–301. [Google Scholar] [CrossRef]
  104. McCollum, B.; Barclay, J.; de Leon, J. Unexpected Falls during Clozapine Treatment Explained by Myoclonus. Prim. Care Companion CNS Disord. 2018, 20, 26969. [Google Scholar] [CrossRef]
  105. Zink, M.; Knopf, U.; Kuwilsky, A. Management of Clozapine-Induced Obsessive-Compulsive Symptoms in a Man with Schizophrenia. Aust. N. Z. J. Psychiatry 2007, 41, 293–294. [Google Scholar] [CrossRef]
  106. Cadeddu, G.; Deidda, A.; Stochino, M.E.; Velluti, N.; Burrai, C.; Del Zompo, M. Clozapine Toxicity Due to a Multiple Drug Interaction: A Case Report. J. Med. Case Rep. 2015, 9, 1–6. [Google Scholar] [CrossRef] [Green Version]
  107. Pieroni, M.; Cavallaro, R.; Chimenti, C.; Smeraldi, E.; Frustaci, A. Clozapine-Induced Hypersensitivity Myocarditis. Chest 2004, 126, 1703–1705. [Google Scholar] [CrossRef] [Green Version]
  108. Flanagan, R.J.; Ball, R.Y. Gastrointestinal Hypomotility: An under-Recognised Life-Threatening Adverse Effect of Clozapine. Forensic Sci. Int. 2011, 206, e31–e36. [Google Scholar] [CrossRef]
  109. Chopra, A.; Rai, A.; Philbrick, K.; Das, P. A Dangerous GI Complication. Curr. Psychiatr. 2011, 10, 68–72. [Google Scholar]
  110. Pelizza, L.; De Luca, P.; La Pesa, M.; Borella, D. Clozapine-Induced Intestinal Occlusion: A Serious Side Effect. Acta Biomed. l’Ateneo Parm. 2007, 78, 144–148. [Google Scholar]
  111. Abou Farha, K.; van Vliet, A.; Knegtering, H.; Bruggeman, R. The Value of Desmethylclozapine and Serum CRP in Clozapine Toxicity: A Case Report. Case Rep. Psychiatry 2012, 2012, 1–4. [Google Scholar] [CrossRef]
  112. Haack, S.; Seeringer, A.; Thürmann, P.A.; Becker, T.; Kirchheiner, J. Sex-Specific Differences in Side Effects of Psychotropic Drugs: Genes or Gender? Pharmacogenomics 2009, 10, 1511–1526. [Google Scholar] [CrossRef]
  113. Yadav, D.; Burton, S.; Sehgal, C. Clozapine-Induced Neutropenia Reversed by Lithium. Prog. Neurol. Psychiatry 2016, 20, 13–15. [Google Scholar] [CrossRef] [Green Version]
Figure 1. Flowchart of search strategy and article selection.
Figure 1. Flowchart of search strategy and article selection.
Pharmaceuticals 15 00817 g001
Table 1. Methods of plasma clozapine measurement.
Table 1. Methods of plasma clozapine measurement.
Methods for Measurement of Plasma CLOZ LevelsReferences
High-Performance Liquid Chromatography (HPLC)Combs et al. [36], Carceller-Sindreu et al. [68], Olesen et al. [35], Oyewumi et al. [37], Hummer et al. [69], Rajii et al. [59], Wong et al. [42], Oyewumi et al. [64], Melkersson et al. [49], Yusufi et al. [56], Spina et al. [46], Melkersson and Dahl [70], Melkersson et al. [45], Lu et al. [71], Rechlin et al. [72], Eschweiler et al. [73], Mauri et al. [38], Vaquero-Baez et al. [67], Anderson et al. [74]
Reversed-Phase High-Performance Liquid Chromatography (RP-HPLC)Haring et al. [58], Lin et al. [60], Shen et al. [55]
Liquid Chromatography (LC)Seppala et al. [75], Kim et al. [47]
Reversed-Phase Liquid ChromatographyCentorrino et al. [41], Frazier et al. [43]
Gas Chromatography (GC)Ackenheil [44], De Leon et al. [66], De Leon et al. [62], De Leon et al. [63], Freudenreich et al. [34], VanderZwaag et al. [39]
Liquid Chromatography coupled with Mass Spectrometry (LC-MS)Curto et al. [76], Vasudev et al. [51]
Liquid Chromatography coupled with tandem Mass Spectrometry (LC-MS/MS)Gharab et al. [77], Smith et al. [78]
Not specifiedHummer et al. [52], Every-Palmer et al. [53], Grande et al. [79], Sporn et al. [40], Khan et al. [80], Subramaniam et al. [65], Lally et al. [81], Nilsson et al. [48], Meltzer et al. [50], Bailey et al. [54], Hummer et al. [57]
Table 2. General scores of adverse effect scales or combinations of adverse effects in relation to clozapine plasma levels.
Table 2. General scores of adverse effect scales or combinations of adverse effects in relation to clozapine plasma levels.
ReferenceType of StudyAge (Mean)
(Years)
ComedicationAveraged Serum
Levels of Clozapine (ng/mL)
Duration of CLOZ ExposureReported Side EffectsCorrelation to CLOZ Plasma LevelsJadad Score
[56]Cross-sectional39.3 ± 8.8Mood stabilizer (31%), Anticholinergic (18%), antidepressant (16%), other antipsychotic (5%), anxiolytic or hypnotic (5%)CLOZ: 530 ± 370,
NCLOZ: 310 ± 190
Median (range): 30 (3–156) monthsParkinsonism, akathisia, tardive dyskinesia, non-neurological side effects including cardiovascular, gastrointestinal, sexual genitourinary and othersCLOZ levels vs. total ANSSERS score: r = 0.29, p < 0.004
CLOZ levels vs. moderate and severe ADRs: r = 0.23, p < 0.03
1
[43]Open-label13.3 ± 2.7 (range 9–16)No concomitant medicationsCrude (ng/mL):
289 ± 116, Normalized (ng/mL-mg-Kg):
99 ± 37.3
6 weeksAdverse effects included sedation, enuresis, tachycardia, sialorrhea, reduced neutrophil count, increased hepatic transaminasesModerate and severe side effects vs. CLOZ + NCLOZ: r = 0.4, p = 0.002)
Moderate and severe side effects vs. NCLOZ: r = 0.6, p = 0.002
Moderate and severe side effects vs. CLOZ + NCLOZ + NOX levels: r = 0.4, p = 0.03
0
[46]Prospective, open follow-up19–65NoCLOZ: 385 ± 183 (range 147–974)
NCLOZ: 174 ± 84 (range: 43–445)
12 weeksHypersalivation, constipation, tachycardia, dizziness, sedation, weight gainNo1
[41]Cross-sectional36.6 ± 9.1 (range 20–54)Benzodiazepines, lithium, antidepressantsCLOZ: mean = 297 (median: 291), among 68 samples.
Subsample not exposed to fluoxetine or valproate (n = 27): 239 ± 159
2.15 ± 2.30 yearsSAFTEE scale ADRsNo0
CLOZ: clozapine, NCLOZ: norclozapine, ANNSERS: Antipsychotic Non-Neurological Side Effects Rating Scale, SAFTEE: Systematic Assessment for Treatment Emergent Effects.
Table 3. Nervous system and psychiatric adverse effects in relation to CLOZ plasma levels.
Table 3. Nervous system and psychiatric adverse effects in relation to CLOZ plasma levels.
ReferenceType of StudyAge (Mean)
(Years)
ComedicationAveraged Serum Levels of Clozapine (ng/mL)DurationReported Side EffectsCorrelation to Clozapine Plasma LevelsJadad Score
[34]Prospective, randomized38 (range: 21–56)Rarely given doses of haloperidol or fluphenazinegroup I (n = 16): 50–150
group II (n = 22): 200–300
group III (n = 12): 350–450
12 weeks
Seizures, EEG changes, sleepinessEEG abnormalities, more severe than borderline, rate: group III: 73% vs. group I: (20%) and group II: 21% (p = 0.006)
Severity: group I 0.9 ± 1.8, group II 1.0 ± 1.5, group III 3.4 ± 1.9 (p < 0.001)
Spike/sharp activity: no correlation
Slowing: more slowing in group III vs. group II and I (p = 0.049), positive correlation with levels (r = 0.44, p = 0.002), possible cutoff: 300 ng/mL
Sleepiness: positive correlation with levels (r = 0.33, p = 0.029). The EEG slowing correlated with observed sleepiness
2
[58]Prospective, observational31.7 ± 10.2NoWhole sample (n = 29): 161.3 ± 150.0 group 1 (n = 14): 81.6 ± 64.6, group 2 (n = 15): 235.7 ± 169.820.2 ± 16.8 daysEEG changesPlasma levels significantly different among groups according to severity of EEG changes.
Group 1 (n = 14): degree 0–1, plasma levels: 81.6 ± 64.6 ng/mL (95% CI = 44.3–118.9). Group 2 (n = 15): degree 2–4, plasma levels: 235.7 ± 169.8 mg.mL (95% CI: 141.7–329.7) (p = 0.0009)
0
[35]Cross-sectional, blinded for EEG measures37.6 ± 1.67
Levomepromazine or chlorprothixene for sedation up to 100 mg/day. Other medication as usualMedian CLOZ: 351 (231–615) (range: 64–1824)2.5 (1.0–9.0) yearsEEG changesSeverity correlated to plasma CLOZ (r = 0.43; p < 0.05) but not NCLOZ levels.
CLOZ concentrations ≥1306 ng/mL lead to progressive gradual EEG changes
0
[39]Prospective, randomized, double-blind3
(range 21–56)
All psychoactive medication tapered off. Valproate in two patients with a history of seizuresLow: 91 ± 15 (50–150), medium: 251 ± 13 (200–300), high: 396 ± 16 (350–450)12 weeksSleepiness,
EPS
Trend of sleepiness and serum level to correlate at week 6 (p = 0.08), but no significance at week 12.
EPS improved over time, with no group-by-time interactions.
2
[40]Prospective, longitudinal, observational, partly double-blind (n = 22) partly open-label (n = 32)Range: 8–18NoWeek 6: CLOZ = 455 ± 285.1,
NCLOZ + CLOZ 302.4 ± 142.2
6 week treatment at first and then 2–6 years follow-upEEG changes seizures,
akathisia
Rates of side effects were not directly associated with CLOZ or nor CLOZ blood levels or their ratio0
[41]Cross-sectional36.6 ± 9.1 (range 20–54)Benzodiazepines, lithium, antidepressants, other medically indicated agentsCLOZ: mean = 297 (median: 291), among 68 samples.
Subsample not exposed to fluoxetine or valproate (n = 27): 239 ± 159
2.15 ± 2.30 yearsSedationNo0
[42]Prospective, non-randomized, double-blind, observational37.61 ± 8.68 (range 21–63)Chloral hydrate,
Lorazepam, Valproate
Week 6:
cloz:
470.20 ± 234.2, range 100–1220
norclozapine: 233.06 ± 105.56, range 70–670, Week 12: cloz 681 ± 390.71, range 220–1920,
NCLOZ:
297.8 ± 146.49 range 8–720
12 weeksEEG changes, akathisia, EPSNo significant correlation between plasma levels and EEG abnormalities on week 6, BARS, AIMS, SAS scores on weeks 6 and 12, negative correlation with sedation at week 6, which was clinically implausible0
[47]Retrospective, observational37.7 ± 11.7Valproate (n = 25, 35.2%)
Mood stabilizers (lamotrigine, valproate, lithium, topiramate) (n = 31)
Antipsychotics, antidepressants, benzodiazepines, or mood stabilizers in combination (n = 68)
CLOZ: 429.4 ± 264.1
NCLOZ: 197.8 ± 132.6
4.6 ± 4.9 yearsEEG changesPositive correlation with CLOZ levels (p = 0.008). No correlation with NCLOZ levels (p = 0.12).
Patients with CLOZ levels > 600 ng/mL had higher rate of EEG abnormalities (93.8%) than those with levels <600 ng/mL (65.5%) (p = 0.02)
0
[75]Cross-sectionalAge: 42.5 (range: 20–65)Clozapine monotherapy: 65.4%,
CLOZ + atypical: 22.5%, ClOZ + typical: 9.7%, CLOZ + typical +
atypical: 1.7%
Men: 722 ± 366, Women:
886 ± 480 (p = 0.03) Total sample:
778 ± 444.57
3–12 months (1.7%),
1–5 years (32.5%),
5 years (57.8%), not specified (8%)
Difficulty in concentrating, tension, difficulty remembering things, depression, restlessness, difficulty getting to sleepYes, only with the Depression/Anxiety score0
[68]Single-blind, cross-sectionalGroup I: 45 ± 10.3, Group II
47.2 ± 7.5
NoGroup I:
CLOZ ≥300
Group II:
CLOZ <300
≥5 yearsCognitive performanceNo relationship between clozapine plasma levels and cognitive performance. Tendency to significance regarding the executive test (31% of variability of number of attempts in the WCST was explained by clozapine plasma levels)0
[43]Open-label13.3 ± 2.7 (range 9–16)NoCrude (ng/mL):
289 ± 116, normalized (ng/mL-mg-Kg):
99 ± 37.3
6 weeksSedationNo0
[59]Retrospective analysis of clinically collected cross-sectional data.41.6 ± 12.0Not reportedAll subjects (n = 73): 458.5 ± 248.8 Low cognitive impairment (n = 57): 437.1 ± 249.6 High cognitive impairment (n = 16): 534.7 ± 237.7≥3 monthsCognitive impairmentSixteen subjects (21.9%) had high cognitive impairment and the rest had low cognitive impairment. Age and clozapine levels were associated with high cognitive impairment, as well as clozapine/desmethylclozapine raitio (OR: 7.3). Yes0
[56]Cross-sectional39.3 ± 8.8Mood stabilizer (31%), Anticholinergic (18%), antidepressant (16%) other antipsychotic (5%), anxiolytic or hypnotic (5%)CLOZ: 530 ± 370,
NCLOZ: 310 ± 190
Median (range): 30 (3–156) monthsMemory and concentration problems, night-time sleep problems,
Parkinsonism, akathisia, tardive dyskinesia
No correlation1
[62]Double-blind, prospective.
Randomized to clozapine doses
Not reportedHaloperidolEnd of first trial (16 weeks): 335 ± 340First trial: 16 weeks, second trial:16 weeksDrowsiness, sedationClozapine levels were very good predictors of serum antimuscarinic activity in doses of 300 mg/d or higher. Sedation showed no significant association with serum antimuscarinic activity.2
[60]Retrospective, naturalistic, 1-year study/cross-sectional37.9 ± 9.3Fluvoxamine (n = 8), fluoxetine (n = 2), sertraline (n = 2), paroxetine (n = 1), valproate (n = 15)Patients with OCS: 595.1 ± 364.9 (range 84–1491) Patients without OCS: 433.5 ± 252.8
(range 82–1273)
Patients with OCS: 81.8 ± 32.2 months, Patients without OCS: 56.1 ± 40.6 monthsOCSPlasma concentration of clozapine was significantly higher in patients with OCS than in those without (p = 0.001)0
CLOZ: clozapine, NCLOZ: norclozapine, OCS: obsessive-compulsive symptoms.
Table 4. Cardiovascular adverse effects in relation to CLOZAPINE blood levels.
Table 4. Cardiovascular adverse effects in relation to CLOZAPINE blood levels.
ReferenceType of StudyAge (Mean)ComedicationAveraged Serum Levels of Clozapine (ng/mL)DurationReported Side EffectsCorrelation to Clozapine Plasma LevelJadad Score
[35]Cross-sectional37.6 ± 1.67Levomepromazine or chlorprothixene. Other medication as usualMedian S-CLOZ: 351 (231–615) (range: 64–1824)2.5 (1.0–9.0) yearsIncreased pulse rate (>80 bpm),
tachycardia (>100 bpm),
orthostatic hypotension
No0
[39]Prospective, randomized, double-blind38 (range 21–56)Haloperidol, phluphenazine, valproateLow: 91 ± 15 (50–150), medium: 251 ± 13 (200–300), high: 396 ± 16 (350–450)12 weeksTachycardia: Orthostaic hypotension:No2
[40]Prospective, longitudinal, observational, partly double-blind (n = 22) partly open-label (n = 32)Range: 8–18NoWeek 6: CLOZ 455 ± 285.1, NCLOZ 302.4 ± 142.26 week treatment at first and then 2–6 years follow-upHypertension (>140/90 mmHg), orthostatic hypotension,
tachycardia (>120 bpm)
No0
[42]Prospective, non-randomized, double-blind, observational37.61 ± 8.68 (range 21–63)Chloral hydrate.
Lorazepam.
Valproate
Week 6:
Clozapine: 470.20 ± 234.23, range 100–1220
Norclozapine: 233.06 ± 105.56, range 70–670, Week 12: clozapine 681 ± 390.71 ng/mL (range 220–1920 ng/mL), NCLOZ 297.8 ± 146.49 ng/mL (range 8–720 ng/mL)
12 weeksQTc alterationsNo0
[44]Prospective, open-labelNot reportedNot reported200–300 ng/mL (divided into three groups I: <30 ng/mL, II: 30–100 ng/mL, III: >100 ng/mL for paranoid-hallucinatory schizophrenic patients and manic syndrome)30 daysOrthostatic hypontension, temperatureYes, with severity of orthostatic dysregulation0
[43]Open-label13.3 ± 2.7 (range 9–16)NoCrude (ng/mL): 289 ± 116, Normalized (ng/mL-mg-Kg): 99 ± 37.36 weeksTachycardiaNo0
[64]Prospective longitudinalNot reportedLorazepam allowedMean serum CLOZ level by week (1–8): 63.5 ± 46.0 199.3 ± 149.1 251.9 ± 178.0 300.7 ± 200.2 316.2 ± 189.4 364.7 ± 195.6 351.9 ± 176.2 379.5 ± 156.5First 8 weeks of administrationHypotension, temperature, pulseThe blood pressure and pulse did not change significantly from baseline to week 8.
Temperature was inversely related to clozapine dose (p < 0.003)
Higher norclozapine to clozapine ratios were associated with higher BP measures (p = 0.002).
The magnitude of these relationships is weak (r < 0.30).
There is a tendency to autonomic dysregulation during clozapine use
0
[80]Cohort, prospective study, open44 ± 12Not reported475 ± 236Mean follow-up of 9 ± 6 yearsMyocarditis, sudden deathNo0
[79]Retrospective, review of case records31.21 ± 9.59None: 52.44%
Antidepressants: 19.51%
Mood stabilizers:31.71%
Stimulants: 1.22%
CLOZ: 305.56 ± 299.64, nor CLZ: 160.23 ± 105.1218 weeks-No1
[76]Preliminary prospective study35.5 ± 11.0One other antipsychotic, one mood-stabilizing drug, or a benzodiazepineCLOZ: 124 ± 70.8 nor CLOZ: 52.3 ± 35.74 weeksIncreased Heart Rate,
Myocardial Performance Index (MPI) > 0.44.
Reduced LV functioning
No0
[72]Cross-sectionalPatients: 40.7 (range: 21–68)
Controls: 41.2
(range: 20–64)
No290>8 weeksHRVYes, negative (inverse) correlation0
[73]Retrospective42.1 (range: 19–73)Haloperidol, benzodiazepines, SSRIs, Carbamazepine331 ± 294 (range: 65–1475)>1 weekHRVYes, negative (inverse) correlation0
[48]Cross-sectional33.5 (range: 26–41)Not reported451 (range: 337–569) (n = 29)7 (range: 3–13)Persistent tachycardiaNo0
HRV: Heart Rate Variability.
Table 5. Metabolic and endocrine adverse effects in relation to clozapine plasma levels.
Table 5. Metabolic and endocrine adverse effects in relation to clozapine plasma levels.
ReferenceType of StudyAge (Mean) (Years)ComedicationAveraged Serum Levels of Clozapine (ng/mL)DurationReported Side
Effects
Correlation to Clozapine Plasma LevelsJadad Score
[63]Prospective, double-blind, randomized44.8 ± 9.6NoCLOZ: >350 in responders29.7 ± 13.2 weeks (16 weeks, 32 weeks, 48 weeks, based on response status)Weight gainIn nonsmokers (n = 8) r = 0.89 (p = 0.046)2
[81]Prospective, observational, open-label37.4 ± 9.3, (range 22–57)Not reported500 ± 280 (range 70–1360)32.6 ± 6.6 weeks (23–62 weeks)Weight gain, waist circumferenceNo
(r = −0.04, p = 0.77) at follow-up
0
[65]Cross-sectional38.2 ± 11.3 (range 22–74); Caucasians: 40.2 ± 8.6, Asians: 36.3 ± 13.4Not reportedCaucasians: 415.3 ± 185.8
Asians: 417.1 ± 290.8
≥6 monthsLipid profiles, fasting glucose levelsNo0
[70]Cross-sectional46 (29–63)Not reportedMedian: 359.1 (range 60.5–810.46)At least 6 months
Median: 5.3 years (range 0.5–16.3 years)
Fasting insulin, C-peptide, insulin-like growth factor I, insulin-like growth factor binding protein-1, leptin, glucose and lipidsCLOZ vs. insulin: r = 0.51, p = 0.03
CLOZ vs. C-peptide: r = 0.48, p = 0.04
CLOZ vs. triglycerides: r = 0.50, p = 0.03)
0
[49]Cross-sectional35 (26–47)No28.8–7212.7 (range 0.5–7.3 years)Fasting glucose, insulin, growth hormone (GH)-dependent insulin-like growth factor I (IGF-I), and insulin-dependent insulin-like growth factor binding protein-1CLOZ vs. insulin levels0
[50]Open, prospectiveMales: 34.7 ± 8.1
Females: 36.2 ± 11.6
Benztropine (n = 6), diphenylhydantoin (n = 2), fluoxetine (n = 1), divalproate (n = 1)6 weeks: 388 ± 242
6 months: 444 ± 355
6 monthsWeight gainNo0
[51]Cross-sectional36.5 ± 11.3Antipsychotics (61.9%), antidepressants (14.3%), mood stabilizers (21.4%)CLOZ: 1613.57 ± 976.05
NCLOZ: 964.60 ± 976.05
Stable clozapine therapy for at least 6 monthsMetabolic syndrome
BMI
CLOZ vs. metabolic syndrome0
[74]RetrospectiveMales: 36.9 (95% CI: 33.9–39.8)
Females: 39 (95% CI: 35.4–42.7)
Aripiprazole (n = 6)
Amisulpride (n = 4)
Haloperidol (n = 2)
For the females: estrogen-containing contraceptive pill (n = 5), estrogen-containing hormone replacement treatment
CLOZ:
Males: 440 (10th–90th percentile: 260–700)
Females: 490 (10th–90th percentile: 270–790)
NCLOZ:
Males: 310 (10th–90th percentile: 260–350)
Females: 310 (10th–90th percentile: 270–340
Males: 4.4 (95% CI: 1.2–10.3) years
Females: 5.1 (95% CI: 2.3–7.9) years
BMI
Fasting blood glucose
HDL
CLOZ vs. BMI
CLOZ vs. fasting blood glucose
0
[45]Cross-sectionalMedian: 41 (range: 29–36)Benzodiazepines (n = 4), and/or levomepromazine (n = 3) and/or lithium (n = 1)CLOZ: 392(69–918)
NCLOZ: 288 (88–641)
6.9 years (range: 0.7–16.3 years)Elevated blood glucose, elevated levels of insulin, elevated levels of C-peptide, elevated triglycerides, cholesterol, HOMA-IRCLOZ vs. insulin: r = 0.53, p = 0.03,
CLOZ vs. C-peptide r = 0.51, p = 0.04
CLOZ vs. triglyceride levels: r = 0.46, p = 0.06
0
[71]Prospective, randomizedCoadministration group: 32.9 ± 8.5
Monotherapy group: 35.1 ± 9.4
NoCoadministration group:
CLOZ: 509.8 ± 281.1
NCLOZ: 179.0 ± 95.8
Monotherapy group:
CLOZ: 502.0 ± 220.6
NCLOZ: 242.8 ± 100.3
12 weeksSerum glucose, cholesterol, and TRG levels,
weight gain
NCLOZ vs. weight gain: r = 0.27, p = 0.026
NCLOZ vs. blood sugar: r = 0.34, p = 0.005
NCLOZ vs. Triglycerides: r = 0.27, p = 0.028
2
[77]Cross-sectional, controlledPatients: 40.94 ± 10.15
Controls: 40.09 ± 1.67
Not reportedCLOZ: 594.90 ± 492.90
NCLOZ: 220.33 ± 182.55
At least 4 monthsBlood measuresCLOZ vs. total cholesterol: r = 0.34, p = 0.040
[41]Cross-sectional36.6 ± 9.1 (range 20–54)Benzodiazepines, lithium, antidepressants, other medically indicated agentsCLOZ: mean = 297 (median: 291), among 68 samples.
Subsample not exposed to fluoxetine or valproate (n = 27): 239 ± 159
2.15 ± 2.30 yearsExcess weightNo0
[66]Double-blind dose–response49 females (32–60 years old), 42 males (31–58 years old)No400–160016-weekHyperprolactinemiaFor every 100 ng/mL increase in plasma clozapine levels, average increments in prolactin levels of 0.45 ng/mL in females and 0.15 ng/mL in males were recorded2
[75]Cross-sectional42.5 (20–65)CLOZ
Monotherapy: 65.4%,
CLOZ + atypical: 22.5%,
CLOZ + typical: 9.7%,
CLOZ +typical
+ atypical:
1.7%
Men (722 ± 366) and women (886 ± 480)3–12 months (1.7%), 1–5 years (32.5%).
5 years (57.8%), unspecified (8%)
Menstrual problemsNo correlation with CLOZ + NCLOZ concentration1
[77]Cross-sectional, controlledPatients: 40.94 ± 10.15
Controls: 40.09 ± 1.67
Not reported594.90 ± 492.90
220.33 ± 182.55
At least 4 monthsBlood measuresNo correlation with TSH, FT4, PRL.
CLOZ vs. FT3:
r = −0.373, p = 0.021
0
Table 6. Gastrointestinal adverse effects in relation to CLOZAPINE blood levels.
Table 6. Gastrointestinal adverse effects in relation to CLOZAPINE blood levels.
ReferenceType of StudyAge
(Mean)
(Years)
ComedicationAveraged Serum
Levels of Clozapine (ng/mL)
DurationReported Side
Effects
Correlation to Clozapine Plasma LevelsJadad Score
[41]Cross-sectional36.6 ± 9.1 (range 20–54)Benzodiazepines, lithium,
antidepressants, other
medically
indicated agents
Median 291, range: 15–7262.15 ± 2.30 yearsNocturnal sialorrhea, drooling, parotid swelling, constipationNo0
[46]Prospective, observational follow-up19–65BenzodiazepinesCLOZ: 385 ± 183 (range 147–974)
NCLOZ: 174 ± 84 (range: 43–445)
12 weeksHypersalivation, constipationNo1
[52]ProspectiveCLOZAPINE subsample: 31.37 ± 11.8Not reported165.4 ± 163.418 weeksPathologic liver function tests (LFTs): SGOT, SGPT, GGT, ALP, bilirubinYes, for SGPT only0
[35]Cross-sectional, naturalisticRange 22–55Nortriptyline, levomepromazine, clonazepam, hyoscyamine, oxazepam, chlorprothixene, phenobarbital, nitrazepam, biperiden, orphenadrine, benztropine, diazepam, piroxicam, disulfiramMedian: 1076 (range 706–1882)
NCLOZ/CLOZ ratio: 0.77 ± 0.17
Median: 2.5 (range 1.0–9.0) yearsIncreased liver enzyme activity (increased GGT, ALP, AST, ALT)No0
[77]Cross-sectional, controlledPatients: 40.94 ± 10.15
Controls: 40.09 ± 1.67
Not reported594.90 ± 492.90
220.33 ± 182.55
At least 4 monthsAST, ALTNo
[65]Cross-sectional38.2 ± 11.3 (range 22–74); Caucasians: 40.2 ± 8.6, Asians: 36.3 ± 13.4Not reportedCaucasians: 415.3 ± 185.8, Asians: 417.1 ± 290.8≥6 monthsElevated levels of alanine (ALT) and aspartate (AST) transferasesNo0
[43]Open-label trial13.3 ± 2.7 (range 9–16)No289 ± 1166 weeksIncreased hepatic transaminaseNo0
[40]Data from double-blind and open-label clozapine trials13.5 ± 2.5 (range 7.0–19.1)Not reported455.6 ± 285.1 (n = 46)6 weeksElevated liver enzymes (AST, ALP, ALT)No0
[53]Cross-sectional39.3 ± 9.8 (range 20–61)
(CLOZAPINE group: 37 ± 8.2, NON
CLOZAPINE group:
Laxatives (laxsol, polyethylene glycol, lactulose), antipsychotics (risperidone, aripiprazole, haloperidol, amisulpride, aripiprazole + quetiapine) omeprazole, metformin, cholecalciferol
489 ± 137 (range 284–885)At least 3 monthsColonic hypomotilityPositive correlation
Clear colonic hypomotility in 80% of CLOZAPINE patients, with Colonic Transit Time (CCT) four times longer than population morms (p < 0.0001) and NON-CLOZAPINE patients (p < 0.0001)
0
[54]Retrospective, collection of recordsMales: 43.5 ± 10.1
Females: 47.2 ± 11.2
Anticholinergic agentsCLOZ:
Laxative users: 533 ± 0.29
Non-laxative users: 486 ± 30
NCLOZ:
Laxative users: 337 ± 0.19
Non-laxative users: 269 ± 0.18
>3 monthsConstipationNCLOZ vs. laxative use, p = 0.046)0
Table 7. Studies reporting hematological, genitourinary, and other adverse effects in relation to CLOZAPINE blood levels.
Table 7. Studies reporting hematological, genitourinary, and other adverse effects in relation to CLOZAPINE blood levels.
ReferenceType of StudyAge
(Mean)
(Years)
ComedicationAveraged Serum
Levels of Clozapine (ng/mL)
DurationReported Side EffectsCorrelation to Clozapine Plasma LevelsJadad Score
Hematological System
[41]Cross-sectional35.6 ± 9.3 (range 17–54)Not reportedCLOZ: 304 ± 174
NCLOZ: 216 ± 133
2.16 ± 0.35 yearsWBC
Neutrophil Count
No0
[35]Cross-sectional37.6 ± 1.67Levomepromazine or chlorprothixeneMedian 430.4 (282.4–752.8)At least 3 months (range: 1–17 years)Mild leukocytosis and increased erythrocyte folateNo0
[37]Prospective longitudinal35.2 ± 10.2 (range: 18–57)Lorazepam379.5 ± 1564 to 8 weeksHematological parameters (WBC, red blood count, neutrophils, platelets, and lymphocytes counts or hemoglobin and hematocrit)CLOZ vs. platelets count r = 0.32,
p = 0.042
0
[40]Prospective, double-blind (n = 22), open-label (n = 32)13.5 ± 2.5 (range 7.0–19.1)Mood stabilizers and antidepressantsCLOZ: 455 ± 285.1
NCLOZ: 302.4 ± 142.2
6 week treatment (2–6 years follow-up)NeutropeniaNo0
[43]Prospective, open-label/double-blind13.3 ± 2.7 (range 9–16)NoCLOZ: 289 ± 116
NCLOZ: 410 ± 190
6 weeksModerate neutropeniaNo0
[36]Retrospective chart review36.4 ± 10.4Not reportedCLOZ: 389 ± 386
NCLOZ: 199 ± 216
Mean duration not reported; samples for measuring drug levels
drawn
at least once during the first 3 months and then randomly after
WBC or granulocyte countsNo0
[78]Retrospective, observational34 (median)
20–84 (range)
Not reportedCLOZ: 1068 (26–3955)
NCLOZ: 712 (31–5813)
Not reportedAbsolute neutrophil count (ANC)NCLOZ vs. ANC p = 0.002)0
[77]Cross-sectional, controlledPatients: 40.94 ± 10.15
Controls: 40.09 ± 1.67
Not reported594.90 ± 492.90
220.33 ± 182.55
At least 4 monthsHematological parametersNCLOZ vs. PLT:
r = −0.362, p = 0.025
NCLOZ vs. HGB:
r = −0.342, p = 0.025,
NCLOZ vs. NEU:
r = −0.385, p = 0.017
0
[38]Prospective, open-label34.62 ± 7.56 (range: 25–48)NoCLOZ: 266.27 ± 197.44 (25–1270)
NCLOZ: 169.0 ± 127.94 (25–1280)
9 weeksLeucocyte count
Neutrophil count
CLOZ vs. NEU: r = 026, p = 0.001
NCLOZ vs. NEU
r = 0.20, p = 0.01
.NCLOZ/CLOZ ratio vs. NEU:
r = −0.26, p = 0.002
0
[69]Prospective, openMales: 28.9 ± 9.7
Females: 34.2 ± 10.7
Not reported145.8 ± 160.1 (3.1–1571.0)16.7 ± 24.7 weeksWBC disorders:
Transient neutropenia, eosinophilia, and leukocytosis.
Progressive neutropenia,
chronic leukocytosis
No0
[67]Cross-sectionalMales: 34.68 ± 1.58
Females: 31.58 ± 1.29
Clonazepam, Fluoxetine,
Paroxetine,
Lithium
Mirtazapine,
Metformin, Sulpiride, valproate, venlafaxine,
Duloxetine, escitalopram, imipramine, losartan, omeprazole, pregabalin
Males: 290.11 ± 51.56
Females: 336.36 ± 29.16
Males: 10.05 ± 1.74 months
Females: 6.63 ± 1.15 months
Leukocyte count
Neutrophil count
CLOZ vs. NEU: r: 0.631, p = 0.004
CLOZ vs. leukocyte count
r: −0.725, p = 0.001
0
Geniturinary Side Effects
[62]Prospective, double-blindNot reportedNot reportedCLOZ: 325 ± 199 NCLOZ: 576 ± 326 (dose-dependent)16 weeksUrinary disturbancesNo2
[40]Prospective, double-blind (n = 22), open-label (n = 32)13.5 ± 2.5 (range 7.0–19.1)Mood stabilizers and antidepressantsCLOZ: 455 ± 285.1
NCLOZ: 302.4 ± 142.2
6 week treatment (2–6 years follow-up)EnuresisNo0
[43]Prospective, open-label/double-blind13.3 ± 2.7 (range 9–16)NoCLOZ: 289 ± 116
NCLOZ: 410 ± 190
6 weeksEnuresisNo0
[41]Cross-sectional35.6 ± 9.3 (range 17–54)Not reportedCLOZ: 304 ± 174
NCLOZ: 216 ± 133
2.16 ± 0.35 yearsNocturnal enuresisNo0
[57]Prospective28.6 ± 9.5Benzodiazepines, anticholinergic drugs, b-blockers, antidepressants, anticonvulsants183.3 ± 150.218 weeksSexual disturbancesCLOZ vs. sexual desire (p = 0.02)
CLOZ and sexual functional disturbances (p = 0.008)
0
Other
[55]Clinical studyGroup I: 38.8 (23–58), group II: 41.5 (21–58), group III: 43.9 (22–60)Not mentioned221.4 ± 109.638.3 ± 6.3 days (CLOZ group)Serum anticardiolipin antibodies (aCL) IgM, IgGCLOZ vs. aCL IgM: r = 0.461, p = 0.0011
[77]Cross-sectional, controlledPatients: 40.94 ± 10.15
Controls: 40.09 ± 1.67
Not reportedCLOZ: 594.90 ± 4 NCLOZ: 92.90
220.33 ± 182.55
At least 4 monthsB12, Na, K, ure, creNo0
Table 8. Classification of side effects according to their relationship to plasma clozapine or norclozapine levels, based on current evidence.
Table 8. Classification of side effects according to their relationship to plasma clozapine or norclozapine levels, based on current evidence.
SystemSide Effect
Positive Correlation with Plasma Clozapine LevelsNo Significant Correlation with Plasma Clozapine LevelsControversial
Nervous systemΕlectroencephalographic abnormalities, EEG slowing Seizures, sedation
Impaired cognitive performance (incl. memory problems and lack of concentration)
Extrapyramidal symptoms and tardive dyskinesia
PsychiatricObsessive-compulsive symptoms
Depression/anxiety factor score (incl. difficulty in concentrating, tension, difficulty remembering things, depression, restlessness, difficulty getting to sleep)
CardiovascularHeart rate variabilityQTc prolongationMyocarditis, pericarditis
Μyocardial function
Sudden death
Autonomic DysregulationBody temperature dysregulationHypertensionOrthostatic hypotension
Τachycardia
MetabolicHyperinsulinemia, C-peptideWeight gain
HyperlipidemiaHyperglycemia
EndocrineInverse correlation with fT3Hyperprolactinemia
GastrointestinalParalytic ileus, constipation, colonic hypomotilityElevation of serum liver enzymesSialorrhea, drooling
Geniturinary EnuresisSexual disturbances
OtherElevation of IgM anticardiolipin antibodies
Table 9. The Jadad scoring system and the interpretation of each score.
Table 9. The Jadad scoring system and the interpretation of each score.
QuestionYes/No
Was the study described as random?1/0
Was the randomization scheme described and appropriate?1/0
Was the study described as double-blind?1/0
Was the method of double-blinding appropriate?1/0
Was there a description of dropouts and withdrawals?1/0
Quality Assessment Based on Total Score
Jadad ScoreQuality of evidence
0–2Low
3–5High
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Skokou, M.; Karavia, E.A.; Drakou, Z.; Konstantinopoulou, V.; Kavakioti, C.-A.; Gourzis, P.; Kypreos, K.E.; Andreopoulou, O. Adverse Drug Reactions in Relation to Clozapine Plasma Levels: A Systematic Review. Pharmaceuticals 2022, 15, 817. https://doi.org/10.3390/ph15070817

AMA Style

Skokou M, Karavia EA, Drakou Z, Konstantinopoulou V, Kavakioti C-A, Gourzis P, Kypreos KE, Andreopoulou O. Adverse Drug Reactions in Relation to Clozapine Plasma Levels: A Systematic Review. Pharmaceuticals. 2022; 15(7):817. https://doi.org/10.3390/ph15070817

Chicago/Turabian Style

Skokou, Maria, Eleni A. Karavia, Zoi Drakou, Vassiliki Konstantinopoulou, Christina-Anna Kavakioti, Philippos Gourzis, Kyriakos E. Kypreos, and Ourania Andreopoulou. 2022. "Adverse Drug Reactions in Relation to Clozapine Plasma Levels: A Systematic Review" Pharmaceuticals 15, no. 7: 817. https://doi.org/10.3390/ph15070817

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop