Skip to main content

MINI REVIEW article

Front. Endocrinol., 25 February 2021
Sec. Obesity
This article is part of the Research Topic The Body and Mind: the Impact of Obesity in Severe Mental Illness. View all 9 articles

A Focused Review of the Metabolic Side-Effects of Clozapine

  • 1Department of Psychiatry, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
  • 2Department of Anesthesiology, Pharmacology and Therapeutics, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada

The second generation antipsychotic drug clozapine represents the most effective pharmacotherapy for treatment-resistant psychosis. It is also associated with low rates of extrapyramidal symptoms and hyperprolactinemia compared to other antipsychotic drugs. However, clozapine tends to be underutilized in clinical practice due to a number of disabling and serious side-effects. These are characterized by a constellation of metabolic side-effects which include dysregulation of glucose, insulin, plasma lipids and body fat. Many patients treated with clozapine go on to develop metabolic syndrome at a higher rate than the general population, which predisposes them for Type 2 diabetes mellitus and cardiovascular disease. Treatments for the metabolic side-effects of clozapine vary in their efficacy. There is also a lack of knowledge about the underlying physiology of how clozapine exerts its metabolic effects in humans. In the current review, we focus on key studies which describe how clozapine affects each of the main symptoms of the metabolic syndrome, and cover some of the treatment options. The clinical data are then discussed in the context of preclinical studies that have been conducted to identify the key biological substrates involved, in order to provide a better integrated overview. Suggestions are provided about key areas for future research to better understand how clozapine causes metabolic dysregulation.

Introduction

Antipsychotic drugs represent the primary pharmacological treatment for schizophrenia spectrum disorders, and are increasingly used to treat other psychiatric conditions (14). Commonly categorized into first, second and third-generation drugs (5), the second-generation antipsychotics (SGAs) significantly improved quality of life by decreasing the incidence of neurological side-effects, such as extrapyramidal symptoms (EPS), that occurred with first-generation antipsychotic (FGA) drugs. However, SGAs are associated with higher rates of metabolic side-effects, which vary considerably by drug (610).

The SGA clozapine is the preferred drug for treatment resistant psychosis (1115), producing therapeutic responses in approximately 30% of patients previously refractory to other antipsychotics (1618), possibly reflecting a unique mechanism of action based on its complex pharmacology (19). It also has relatively low risk for EPS and hyperprolactinemia (20). Clinically, clozapine reduces violent and aggressive behavior in patients with schizophrenia (21, 22), and has been associated with the lowest all-cause mortality rate among all antipsychotics (2325). Yet it is estimated that only 10–20% of eligible patients in the U.S. are prescribed clozapine, indicating that the drug is strongly underutilized (26).

This underutilization is due to a number of factors, including lack of prescribing experience by clinicians, institutional take-up of the drug, and concerns about blood monitoring for neutropenia/agranulocytosis, as well as other drug side-effects (27). With regards to the latter, clozapine is associated with a plethora of adverse effects (13, 26), which include a wide range of immune, metabolic, cardiovascular and psychiatric complications. Serious adverse effects include neutropenia/agranulocytosis (28), myocarditis/cardiomyopathy and tachycardia (2931), and obsessive–compulsive symptoms (32, 33). However, the most common issues associated with clozapine use are the metabolic side-effects (34, 35), which occur in a majority of patients. These span a range of metabolic substrates, including glucose, insulin, lipids and body fat, which are collectively referred to as the “metabolic syndrome” (36). Patients who use clozapine consistently have more severe metabolic side-effects than with any other antipsychotic drug (6, 37). For example, a recent observational study of clozapine-treated outpatients noted that 80% were overweight, and 58% met criteria for metabolic syndrome, with concurrent high rates of hypertension, hyperglycemia and hyperlipidemia (38). Similarly, a retrospective chart review of clozapine users at community mental health clinics noted that 45% met criteria for metabolic syndrome, but these physical symptoms were often undertreated, with only 31% receiving treatment for hyperglycemia and 16% for hypertension (39). As premature death in schizophrenia is primarily caused by cardiometabolic disorders (40, 41), and clozapine often remains the only option for treatment resistant schizophrenia, it is imperative to understand in more detail the metabolic side-effects of clozapine use. In the present review, we summarize the main metabolic side-effects of clozapine in clinical populations, and integrate findings from recent preclinical studies to help elucidate the physiological pathways involved (42).

Metabolic Syndrome

The clinical definition of metabolic syndrome has varied over past years depending on whether the emphasis was on insulin resistance, obesity or cardiovascular anomalies (36). In addition to Reaven’s initial description of hypertension, dysglycemia and dyslipidemia as factors that raise the risk for cardiovascular disease (CVD) and Type 2 diabetes mellitus (T2DM), the metabolic syndrome is also known to include abnormalities in coagulation and inflammation (43), and are frequently associated with obesity (44). Clinically, the metabolic risk factors themselves are not routinely measured for a diagnosis of metabolic syndrome. Instead, a diagnosis is made if three of the five following criteria are met: 1) waist circumference ≥ 102 cm in men and 88 cm in women (numbers change based on ethnicity), 2) triglyceride levels ≥ 150 mg/dl, 3) HDL cholesterol below 40 mg/dl in men and 50 mg/dl in women, 4) hypertension (blood pressure ≥ 130/85 mm Hg) and 5) glucose levels ≥ 100 mg/dl (45). Drugs used to treatment metabolic syndrome in clozapine users may target individual or multiple symptoms; for example, metformin is efficacious in concurrently ameliorating obesity, hyperglycemia and triglycerides (46).

Abdominal Obesity

Obesity and weight gain, commonly estimated by the body mass index (47), potentially contribute to increased risk of cardiometabolic disorders. Previous studies have identified excess abdominal fat as an independent risk factor for CVD, where abdominal fat distribution in particular is a better predictor of CVD than body mass index (48). Of note, obesity does not necessarily coincide with insulin resistance, diabetes mellitus nor risk for CVD, since weight gain can be similar between patients, but visceral fat distribution can vary, thus emphasizing the importance of abdominal adiposity as an independent risk factor for cardiometabolic disorders in schizophrenia patients (4951).

The propensity for clozapine to cause weight gain and obesity is well documented in patients with schizophrenia (52, 53). Compared to other antipsychotics, clozapine was associated with the largest amount of weight gain during the first 12 months of treatment and at up to 46 months, with 30.5% of patients subsequently developing T2DM (54). A follow-up study performed by the same authors revealed patients gained approximately 13.6 kg over a 10-year period of clozapine administration, with the risk of CVD and T2DM increasing over time in this cohort (41). Significant weight gain is a concern as an increase of >7% of desirable weight (the midpoint of a weight range for a specific height) can strongly predispose patients to risk for CVD (55, 56), which can only be partially reversed in clozapine treated patients by routine antidiabetic drugs, such as metformin (57). The specific risk factors associated with clozapine-induced weight gain, which varies considerably at the individual level, include variables such as sex, smoking status, and baseline levels of BMI, as well as interactions between these variables (58).

One of the key theories regarding the harms caused by fat accumulation, and visceral fat in particular, is the inflammatory hypothesis, in which clozapine-induced weight gain increases production of proinflammatory cytokines in insulin responsive cells, and monocyte infiltration and the inflammatory state contribute to insulin resistance (59). Cytokines and adipokines secreted by visceral white adipose tissue maintain metabolic and energy balance (60). Alterations in the levels of cytokines including interleukin 6 (IL-6) and tumor necrosis factor alpha (TNF-α), and adipokines such as adiponectin, leptin and resistin, have been associated with metabolic abnormalities (61, 62).

In particular, the extensively studied pro-inflammatory cytokine IL-6 is strongly correlated with all components of the metabolic syndrome in patients with schizophrenia (63). Increased plasma IL-6 levels are linked to obesity and reduced insulin sensitivity, via increased lipolysis, release of adiponectin and disruption of insulin signaling cascades (61, 64). TNF-α is also interconnected with obesity, T2DM and insulin resistance, where its expression is upregulated in obese individuals (65). TNF-α induces lipolysis through suppression of phosphodiesterase-3B and subsequently interfering with insulin’s antilipolytic effects (66). Alternatively, perilipin is subject to downregulation by TNF-α, resulting in increased access of hormone-sensitive lipase to triglycerides and increased lipolysis (67). Targeted mutations to abolish the function TNF-α receptors and expression of TNF-α improved insulin signaling in obese mice, suggesting TNF-α is critical for the development of insulin resistance (68, 69). Finally, inhibition of inositol receptor pathways by IL-6 and TNF-α leads to decreased glucose transport, thus resulting in insulin resistance (66).

Adipokines such as adiponectin, leptin and resistin originate from adipose tissue with important roles in metabolic homeostasis. Adiponectin has been extensively studied for its involvement in insulin sensitivity, obesity and T2DM (64). Circulating adiponectin levels are inversely related to risk for T2DM and metabolic syndrome and a promising therapeutic target (70). Adiponectin influences insulin sensitivity by activating 5’-AMP-activated protein kinase, leading to increased glucose uptake and free fatty acid oxidation (71). In obesity, the combined effects of reduced adiponectin and adiponectin receptor expression exacerbates insulin resistance and hyperinsulinemia (71, 72). Consequently, therapeutic strategies to manage metabolic syndrome and insulin resistance include raising adiponectin levels through increased release from adipocyte differentiation and gene transcription with the antidiabetic agent thiazolidinedione (TZD) (70, 73). Upregulation of adiponectin receptor expression is achievable by activation of peroxisome proliferator-activated receptor (PPAR)-α/γ (74, 75). Of note, PPAR-γ is also positively correlated with adiponectin levels in circulation, believed to be the result of directly affecting adiponectin production or its secretory mechanism (76). It was previously shown that the metabolic syndrome was more prevalent in patients treated with clozapine and this is associated with lower adiponectin levels (77).

Resistin is recognized as a connecting factor between insulin resistance, diabetes and obesity, albeit with debatable connections to the individual components of the metabolic syndrome (78, 79). Resistin is abundantly expressed in mononuclear cells and promotes inflammation, and raises the levels of cytokines such as TNF-α (78, 80). In contrast to its established pro-inflammatory effects, the evidence for resistin’s associations with obesity, insulin resistance and glucose regulation remains weak (78). Interest for resistin as a link between obesity and diabetes originated from the observations of increased insulin sensitivity in response to reducing resistin levels in obese mice (81). In humans, however, studies investigating resistin’s association with insulin resistance and T2DM are inconclusive and resistin has stronger implications in atherosclerosis and cardiovascular disease (80, 82, 83). While a causal role of resistin in metabolic anomalies remains to be determined, there is evidence of positive correlations between resistin and T2DM pathogenesis, which can largely be attributed to resistin’s pro-inflammatory properties (78, 84). Future studies focusing on neutralizing resistin in humans are warranted, especially when its native receptor remains to be identified in humans (84).

In addition, waist circumference is positively correlated with insulin resistance in nondiabetic patients with schizophrenia who received clozapine treatment (49). Of interest, increased waist circumference is the strongest predictor of insulin resistance among commonly used anthropometric measurements (e.g. body mass index, insulin sensitivity index, lipid levels) in clozapine-treated patients, but the same association was absent in patients treated with olanzapine (49).

Clozapine’s complex pharmacology and blockade of multiple receptors may contribute to this weight gain, where the histamine H1-receptor is believed to play a significant role (85). Weight gain occurs in H1-receptor “knock out” mice, in which high fat diets cause quicker body fat deposition compared to wild-type mice (86). Leptin potentially mediates the increase in adiposity, acting to disinhibit the negative feedback loop involving histamine neurons that normally suppress food intake (86) and increasing feeding. In support of this, animal experiments have shown clozapine reverses leptin’s effects via selective augmentation of hypothalamic adenosine monophosphate-activated protein kinase (AMPK) activity (87). As AMPK stimulation in the hypothalamus is known to increase food intake (88), clozapine potentially removes leptin’s anorexigenic effects by activating AMPK. In addition to suppressing appetite, decreased lipolysis resulting from inhibition of H1-receptors (89) may also contribute to clozapine-induced weight gain.

Clozapine also affects other hormones associated with obesity such as ghrelin and neuropeptide Y (NPY). Ghrelin serves to increase food intake by stimulation of ghrelin receptors located in brain centers involved in energy homeostasis, such as the hypothalamus and hindbrain (90). Chronically, ghrelin’s effects on adipogenesis, energy expenditure, lipolysis and dietary preference leads to the imbalance of energy intake and expenditure, eventually causing weight gain and potentially metabolic syndrome (90). Ghrelin is known to decrease as insulin levels rise in both rodents and humans, is correlated positively with insulin sensitivity, and has shown promise as a therapeutic target for diabetes (91, 92). Clozapine-treated patients have higher fasting serum ghrelin levels than control subjects, attributed to aberrant ghrelin secretion that is mediated through receptors such as the H1, serotonin (5-HT2A/2C) and dopamine (D2) receptors (93).

NPY also stimulates food intake and exogenous administration has demonstrated significant weight gain in rodents (94). NPY is elevated in obesity and promotes energy storage, and decreases in response to administration of leptin or insulin (94). Furthermore, it has been demonstrated that knockdown of the Y2 receptor for NPY can reduce abdominal fat and alleviate most symptoms of the metabolic syndrome (95). In rats, clozapine treatment elevates NPY levels in the arcuate nucleus of the hypothalamus involved in meal initiation (96). It has been shown polymorphisms in the NPY gene is associated with clozapine-induced weight gain and contributes to the development of obesity (97).

Clozapine-induced weight gain is potentially mediated through melanocortin receptor-4 (MC4R). MC4R is a downstream target of leptin signaling and mice lacking MC4R developed obesity (98). In humans, single nucleotide polymorphisms in the MC4R gene is associated with increased risk for weight gain in patients treated with clozapine and of European descent (99). Carriers of MC4R mutations display metabolic anomalies including increased energy intake, obesity and hyperinsulinemia, and the severity of these symptoms are correlated with the amount of functional MC4R (100).

Glucagon-like peptide-1 (GLP-1) is of worthy mention as a therapeutic intervention for obesity and T2DM resulting from clozapine treatment. GLP-1 acts to decrease food intake and glucagon secretion and increase insulin secretion, rendering it an attractive target for the management of metabolic disturbances (101). Importantly, rodent experimental data revealed clozapine decreased GLP-1 levels to raise glucose output and glucagon release and administration of GLP-1 agonists neutralizes these effects (101). A setback for the direct administration of GLP-1 is its rapid degradation, hence GLP-1 receptor agonists (GLP-1RAs) are preferred for prolonged glycemic and weight control (102). Preclinical studies using GLP-1RAs such as exendin-4 and Boc5 reversed glucose intolerance induced by clozapine treatment (103, 104). Human studies detailing the beneficial effects of GLP-1ARs in clozapine-treated patients are limited—a recent systematic review identified three studies demonstrating favorable outcomes on body weight, BMI, fasting glucose, waist circumference and BMI (105). Exenatide and liraglutide were used in these studies, with two studies showing weight loss following GLP-1AR administration (57, 106), and the other showing insignificant weight loss compared to controls (107).

While the above discussion is primarily centered around white adipose tissue, there is evidence that brown adipose tissue can mediate antipsychotic-induced weight gain, obesity and insulin resistance (60). Clozapine inhibits the differentiation of brown adipocytes and lipogenic genes (108), actions that are known to be associated with insulin resistance and energy balance (109). The downregulation of brown adipose marker uncoupling protein-1 (UCP-1) is of interest, as UCP-1 promotes leptin activity and possibly contributes to clozapine-induced weight gain through interference of insulin signaling (108).

Dyslipidemia

Excessive plasma triglycerides ≥ 1.7 mmol/L and/or HDL-cholesterol levels below 40 mg/dl in men (<50 mg/dl in women), are part of the diagnostic criteria for the metabolic syndrome (36, 45). Dyslipidemia has detrimental effects on endothelial function and significantly increases the risk of coronary artery disease, particularly in individuals with diabetes (110). Endothelial injury arises through the accumulation of excessive lipids which eventually leads to atherosclerosis. Specifically, the infiltration of monocytes and T helper type-1 cells between dysfunctional endothelial cells results in the proliferation of smooth muscle cells and lipid-filled macrophages to form fibrous plaques characteristic of atherosclerosis (111).

Numerous studies have reported clozapine significantly raises serum triglyceride levels in patients (34, 35, 41, 112, 113). Treatment with clozapine over a 1-year period was associated with notable increases in serum triglycerides, and was significantly correlated with weight gain (114). The increase in serum triglycerides and total cholesterol occurred as early as the first month of initiating clozapine treatment and persisted throughout the study (114). However, other studies have noted that dyslipidemia can also occur independent of weight gain (113, 115). The causal association between clozapine treatment and hyperlipidemia is further confirmed in discontinuation studies, when increased triglyceride levels in clozapine-treated patients resolve follow switching to another antipsychotic (116). Of interest, elevated triglycerides in clozapine-treated patients are associated with improved outcome in patients with schizophrenia, as measured by decreased Positive and Negative Syndrome Scale (PANSS) scores (113, 117) and this is independent of weight gain (34). This raises the possibility of serum lipids influencing the pharmacokinetics and efficacy of clozapine (118), warranting further research, although it is unlikely clozapine causes changes in brain lipid levels (119, 120) which have been associated with clinical improvement.

Treatment options for clozapine-induced dyslipidemia include the use of statins that effectively lower cholesterol and triglyceride levels (121, 122). Considered the standard treatment for lowering cholesterol, statins lower LDL and total cholesterol levels, with a lesser effect on triglycerides (123). While displaying promising results for treating SGA-associated dyslipidemia, it should be noted statins can have adverse side effects. Notably, statins can elevate the risk of developing diabetes (124). Therefore, clinical benefits of improving cardiovascular health should be weighed against the increase in the incidence of T2DM from statin use, especially in patients treated with clozapine. The underlying mechanism for clozapine-induced dyslipidemia other than increased food intake remains unknown, and no confirmed receptor targets have been reliably identified. Given its involvement in cardiovascular function and regulating metabolism, the autonomic nervous system and its individual branches are potential candidates for mediating the cardiometabolic side effects of clozapine. In particular, heightened activity of the sympathetic nervous system contributes to glucose dysregulation and cardiovascular anomalies (125127).

Alternatively, PPAR-α agonists are feasible candidates to manage clozapine-induced dyslipidemias. PPAR-α activation leads to improved HDL, LDL, triglyceride levels and has evident modulatory roles in energy homeostasis, as demonstrated in knockout mice that display hyperlipidemia and hypoglycemia (128, 129). Hypolipidemic fibrates are synthetic ligands for PPAR-α and are used to manage elevations in circulating triglycerides and decreased HDL cholesterol (129). In a case study, fenofibrate was administered to treat hyperlipidemia caused by clozapine treatment (130).

Hypertension

Elevated blood pressure is common in both diabetic and obese individuals, present in 85% of patients diagnosed with metabolic syndrome (131). It was suggested the hypertension observed in these individuals is a result of compensatory mechanisms to the lack of response to insulin at the cellular level (132). Furthermore, poor response to insulin in insulin-resistant individuals is also a contributing factor, where insulin normally induces production of nitric oxide for vascular relaxation (133). Decreased insulin sensitivity leads to hyperinsulinemia as a compensatory mechanism, ultimately causing hypertension via activation of the renin angiotensin aldosterone system (131). Finally, an overactive sympathetic nervous system can lead to hypertension, often present in individuals with obesity and insulin resistance (134). Elevated plasma catecholamine levels induced by hyperinsulinemia may possibly contribute to the rise in blood pressure (135).

In comparison to the incidence rates of T2DM, dyslipidemia and obesity, there are fewer reported cases of hypertension in patients treated with clozapine (136). A claims-based approach study found no significant difference in the incidence of hypertension in patients treated with clozapine as compared to patients receiving FGAs (137). However, another chart review comparing patients treated with FGAs, SGAs (other than clozapine) or clozapine had contradictory results, where hypertension was strongly associated with clozapine use (136). At the end of the 5-year follow-up period, the clozapine group had significantly elevated blood pressure, resulting in 22% of these patients requiring medication for hypertension, as compared to 4% of the FGA group and 9% of the SGA group (136). Blood pressure increased as early as six months after initiating clozapine treatment, signifying the need to routinely monitor blood pressure as a prevention for CVD (136).

Hyperglycemia

Hyperglycemia is the defining characteristic of metabolic dysfunction linked to T2DM (138). Individuals with fasting blood glucose levels between 5.6–6.9 mmol/l or 2-hour plasma glucose values of 7.8–11.0 mmol/l in the oral glucose tolerance test (OGTT) are considered to have impaired fasting glucose and impaired glucose tolerance, respectively (139). These individuals are considered to have elevated risk for developing T2DM, commonly known as the pre-diabetic stage. For diagnosis of T2DM, the standard biomarker for glycemic control, hemoglobin A1C, is commonly used. The A1C assay measures the indirect effects of plasma glucose levels over a span of 2–3 months and a value of ≥ 6.5% is used to diagnose T2DM (140). The A1C assay, coupled with a fasting plasma glucose of ≥ 7.0 mmol/l or a 2 h plasma glucose of ≥ 11.1 mmol/l in the OGTT, form the diagnostic criteria for diabetes (140).

Hyperglycemia causes cardiovascular damage by activating pathways that lead to excessive oxidative stress (141). Four mechanisms have been proposed to underlie glycemia-related vascular damage: decreased production of the antioxidant glutathione via activation of the polyol pathway, excessive generation of advanced glycation end products, activation of protein kinase C and increased activation of the hexosamine pathway (142). The common link between these four mechanisms is the inhibition of the glycolytic enzyme, glyceraldehyde-3 phosphate dehydrogenase (GADPH), by excessive superoxide production. Inhibition of GADPH results in increased upstream glycolytic intermediates and glucose, subsequently activating the aforementioned damaging pathways (142, 143). Several regions are susceptible to hyperglycemia-induced vascular damage, including the peripheral nerve, renal glomerulus and retina, as well as arteries in the brain, heart and lower limbs (141). Exposure to reactive oxygen species can adversely affect vascular contractile function, result in cardiomyopathy and atherosclerosis, and cause renal dysfunction (144), and clozapine was shown to cause oxidative stress in the liver in rats (145).

Clozapine has the highest propensity of all of the SGAs to induce hyperglycemia, which can usually can be resolved upon discontinuation (146148). Glucose intolerance associated with clozapine treatment contributes to the development of new onset T2DM and exacerbates pre-existing cases (146, 149), both of which can occur independently of weight gain (148, 150). Weight gain is not present in all patients receiving clozapine treatment, and is consequently considered a contributing factor rather than the sole mechanism underlying insulin resistance (149, 151, 152). As a causal relationship between clozapine use and the development of diabetes mellitus cannot simply be attributed to excessive adiposity (153), there has been increasing attention given to weight-independent mechanisms to explain glucose dysregulation. One area of focus is clozapine’s antagonistic properties at receptors mediating glucose homeostasis, namely muscarinic, serotonergic and dopaminergic receptors (154). Acute antagonism of M3 and 5-HT2A receptors, known to directly affect pancreatic β-cell function and insulin secretion (155), was found to decrease insulin secretion during the hyperglycemic clamp (which estimates peripheral insulin sensitivity and secretory capacity of β-cells following a glucose challenge) in animals whereas blockade of D2/D3 receptors had the opposite effect (154). In a follow up study, α1 antagonism with prazosin inhibited insulin secretion and glucose infusion rates after a glucose challenge, indicative of impaired β-cell function (156). As clozapine is known to rapidly reduce insulin sensitivity and alter hepatic glucose production (157), the role of antagonism of the above receptors as responsible for clozapine-induced impairment of β-cell function remains to be determined.

Clozapine and Elevation of Plasma Catecholamines

The adrenoceptors and their endogenous ligands norepinephrine and epinephrine play a critical role in glucose homeostasis (127, 156, 158). Sympathetic activation rapidly raises blood glucose levels by suppressing insulin release, promoting glucagon secretion and hepatic gluconeogenesis and glycogenolysis via binding to G protein-coupled receptors (158). It is now well-established that clozapine treatment in both humans and animals causes a large increase in plasma levels of the these catecholamines. We recently reported the effects of multiple doses of the four different antipsychotic drugs haloperidol, risperidone, olanzapine, and clozapine on peripheral levels of the catecholamines dopamine, norepinephrine, and epinephrine in adult rats (159). While all drugs increased catecholamine levels, this effect was significantly larger in clozapine treated animals, and occurred with doses of the drug that we had previously shown to exert acute hyperglycemic effects (160162).

Clinically, an earlier study noted that treatment with clozapine at 175–600 mg/day for 30 days resulted in a significant elevation of plasma norepinephrine levels, as well as heart rate, in patients with psychosis compared to healthy controls (163). Subsequent studies in patients with schizophrenia produced similar results (164167). It was initially thought the increase in plasma norepinephrine was due to inhibition of α-adrenoceptors and the norepinephrine transporter, as the levels of the intraneuronal metabolite 3,4-dihydroxyphenylglycol (DHPG) remained unchanged (165). A follow up study refuted this hypothesis, because radiolabeled DHPG concentrations remained unchanged in plasma, and thus indicated normal reuptake and metabolism of norepinephrine (166). The authors suggested that increased norepinephrine vesicular fusion with the sympathetic nerve membrane accounts for the unchanged plasma DHPG levels and increased plasma norepinephrine. The mechanism through which clozapine elevates plasma norepinephrine, and whether this is associated with improved psychotic symptoms, remains moot (167). A possible reason for the discrepancy is the small sample numbers (n < 14) and short duration of available trials measuring plasma norepinephrine, with the longest published trial lasting 6 weeks (165, 167). However, the large increases in norepinephrine observed in these studies have potentially important implications not only for the metabolic side-effects of the drug, but also for the cardiovascular side-effects too, which we have described in detail previously (29).

Preclinical Studies

In patients treated with antipsychotics, the causes of metabolic dysregulation are multifactorial, and include poor diet, lack of exercise, unhealthy habits (such as smoking/drinking) and direct effects of the antipsychotic itself (168). Teasing apart these individual contributions is challenging, and thus animal models of antipsychotic-induced metabolic dysregulation have provided key mechanistic insights (42), where the drug-specific effects can be studied separately. Preclinical studies with rodents have strong predictive validity, as the antipsychotics with the greatest metabolic liability in humans show similar effects in animals (162, 169171). We would estimate that the most commonly studied antipsychotic is olanzapine (172182), due to its potent metabolic effects and widespread use in patients. However, a number of preclinical studies have focused specifically on clozapine. These studies have demonstrated conclusively that treatment with clozapine can cause glucose intolerance, measured using the glucose tolerance test, and these effects are dose-dependent (104, 160162, 183, 184). Importantly, these studies demonstrate that hyperglycemia occurs acutely, and is independent of weight gain (185). In a similar manner, a number of reports have examined the acute metabolic effects of clozapine using the hyperinsulinemic-euglycemic clamp, which is the “gold-standard” technique for measuring whole-body insulin resistance. Converging findings from different groups reliably show that clozapine causes profound insulin resistance (157, 186), and furthermore, the primary metabolite of the drug—norclozapine—also induces whole-body insulin resistance (160). The clamp studies implicate a direct effect of clozapine on increased hepatic glucose production and impaired beta cell function in the pancreas. The physiological mechanisms underlying these metabolic effects remain an ongoing area of study, but it has been suggested that clozapine’s effects on the autonomic nervous system may play a key role (187).

Summary

It is now well established that treatment with clozapine is commonly associated with pronounced metabolic side-effects in many patients, typically greater than for all other antipsychotic drugs. These metabolic changes span a range of diverse metabolic substrates, leading to the development of metabolic syndrome and ultimately T2DM and CVD in many patients. These sequelae contribute to underutilization of the drug, which represents a serious concern, as clozapine is uniquely efficacious in managing treatment resistant psychosis. Treatment of these metabolic changes is only partly effective in most cases, and so a better understanding of the physiology may be required to develop more effective interventions. Animal models of clozapine’s metabolic side-effects have already provided important insights into how the drug directly affects metabolic physiology, and may be used to help identify novel pharmacotherapies when working in parallel with clinical studies.

Author Contributions

All authors contributed to the article and approved the submitted version.

Funding

This work was supported by a Natural Sciences and Engineering Research Council of Canada grant to AB, and a British Columbia Provincial Health Services Authority grant to AB and RP. WH was supported by the Jack Bell Chair in Schizophrenia.

Conflict of Interest

WP reports personal fees from Abbatis Bioceuticals, Medipure Pharmaceuticals and is owner of Translational Life Sciences. RP reports personal fees from Janssen, Lundbeck and Otsuka. WH reports personal fees from Canadian Agency for Drugs and Technology in Health, AlphaSights, Guidepoint, Translational Life Sciences, Otsuka, Lundbeck, and Newron, grants from Canadian Institutes of Health Research, BC Mental Health and Addictions Services, and has been a consultant (non-paid) for In Silico. AB has been a scientific advisor to Emerald Health Therapeutics, Cannevert Therapeutics, Global Cannabis Applications Corp, Medipure Pharmaceuticals, Vitality Biopharma and Oakum Cannabis Corp.

The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

References

1. Vasudev A, Chaudhari S, Sethi R, Fu R, Sandieson RM, Forester BP. A Review of the Pharmacological and Clinical Profile of Newer Atypical Antipsychotics as Treatments for Bipolar Disorder: Considerations for Use in Older Patients. Drugs Aging (2018) 35(10):887–95. doi: 10.1007/s40266-018-0579-6

PubMed Abstract | CrossRef Full Text | Google Scholar

2. Hershenberg R, Gros DF, Brawman-Mintzer O. Role of atypical antipsychotics in the treatment of generalized anxiety disorder. CNS Drugs (2014) 28(6):519–33. doi: 10.1007/s40263-014-0162-6

PubMed Abstract | CrossRef Full Text | Google Scholar

3. Linton D, Barr AM, Honer WG, Procyshyn RM. Antipsychotic and psychostimulant drug combination therapy in attention deficit/hyperactivity and disruptive behavior disorders: a systematic review of efficacy and tolerability. Curr Psychiatry Rep (2013) 15(5):355. doi: 10.1007/s11920-013-0355-6

PubMed Abstract | CrossRef Full Text | Google Scholar

4. Mela M, Hanlon-Dearman A, Ahmed AG, Rich SD, Densmore R, Reid D, et al. Treatment algorithm for the use of psychopharmacological agents in individuals prenatally exposed to alcohol and/or with diagnosis of fetal alcohol spectrum disorder (FASD). J Population Ther Clin Pharmacol J La Therapeut Des Populations La Pharmacol Clin (2020) 27(3):e1–e13. doi: 10.15586/jptcp.v27i3.681

CrossRef Full Text | Google Scholar

5. Mailman RB, Murthy V. Third generation antipsychotic drugs: partial agonism or receptor functional selectivity? Curr Pharm Des (2010) 16(5):488–501. doi: 10.2174/138161210790361461

PubMed Abstract | CrossRef Full Text | Google Scholar

6. De Hert M, Detraux J, van Winkel R, Yu W, Correll CU. Metabolic and cardiovascular adverse effects associated with antipsychotic drugs. Nat Rev Endocrinol (2012) 8(2):114–26. doi: 10.1038/nrendo.2011.156

CrossRef Full Text | Google Scholar

7. Henderson DC. Managing weight gain and metabolic issues in patients treated with atypical antipsychotics. J Clin Psychiatry (2008) 69(2):e04. doi: 10.4088/jcp.0208e04

PubMed Abstract | CrossRef Full Text | Google Scholar

8. Rummel-Kluge C, Komossa K, Schwarz S, Hunger H, Schmid F, Lobos CA, et al. Head-to-head comparisons of metabolic side effects of second generation antipsychotics in the treatment of schizophrenia: A systematic review and meta-analysis. Schizophr Res (2010) 123(2–3):225–33. doi: 10.1016/j.schres.2010.07.012

PubMed Abstract | CrossRef Full Text | Google Scholar

9. Lang DJ, Barr AM, Procyshyn RM. Management of Medication-Related Cardiometabolic Risk in Patients with Severe Mental Illness. Curr Cardiovasc Risk Rep (2013) 7(4):283–7. doi: 10.1007/s12170-013-0321-1

PubMed Abstract | CrossRef Full Text | Google Scholar

10. Deng C. Effects of antipsychotic medications on appetite, weight, and insulin resistance. Endocrinol Metab Clinics North Am (2013) 42(3):545–63. doi: 10.1016/j.ecl.2013.05.006

CrossRef Full Text | Google Scholar

11. Swartz MS, Stroup TS, McEvoy JP, Davis SM, Rosenheck RA, Keefe RS, et al. What CATIE found: results from the schizophrenia trial. Psychiatr Serv (2008) 59(5):500–6. doi: 10.1176/ps.2008.59.5.500

PubMed Abstract | CrossRef Full Text | Google Scholar

12. Remington G, Addington D, Honer W, Ismail Z, Raedler T, Teehan M. Guidelines for the Pharmacotherapy of Schizophrenia in Adults. Can J Psychiatry (2017) 62(9):604–16. doi: 10.1177/0706743717720448

PubMed Abstract | CrossRef Full Text | Google Scholar

13. Remington G, Lee J, Agid O, Takeuchi H, Foussias G, Hahn M, et al. Clozapine’s critical role in treatment resistant schizophrenia: ensuring both safety and use. Expert Opin Drug Saf (2016) 15(9):1193–203. doi: 10.1080/14740338.2016.1191468

PubMed Abstract | CrossRef Full Text | Google Scholar

14. Lee LHN, Procyshyn RM, White RF, Woodward TS, Honer WG, Barr AM. Antipsychotic prescribing patterns on admission to and at discharge from a tertiary care program for treatment-resistant psychosis. PLoS One (2018) 13(8):e0199758. doi: 10.1371/journal.pone.0199758

PubMed Abstract | CrossRef Full Text | Google Scholar

15. Honer WG, Procyshyn RM, Chen EY, MacEwan GW, Barr AM. A translational research approach to poor treatment response in patients with schizophrenia: clozapine-antipsychotic polypharmacy. J Psychiatry Neurosci (2009) 34(6):433–42.

PubMed Abstract | Google Scholar

16. Meltzer HY. Treatment-resistant schizophrenia–the role of clozapine. Curr Med Res Opin (1997) 14(1):1–20. doi: 10.1185/03007999709113338

PubMed Abstract | CrossRef Full Text | Google Scholar

17. Honer WG, Jones AA, Thornton AE, Barr AM, Procyshyn RM, Vila-Rodriguez F. Response trajectories to clozapine in a secondary analysis of pivotal trials support using treatment response to subtype schizophrenia. Can J Psychiatry Rev Can Psychiatr (2015) 60(3 Suppl 2):S19.

Google Scholar

18. Yin J, Barr AM, Ramos-Miguel A, Procyshyn RM. Antipsychotic Induced Dopamine Supersensitivity Psychosis: A Comprehensive Review. Curr Neuropharmacol (2017) 15(1):174–83. doi: 10.2174/1570159x14666160606093602

PubMed Abstract | CrossRef Full Text | Google Scholar

19. Kim DD, Barr AM, Honer WG, Procyshyn RM. Reversal of Dopamine Supersensitivity as a Mechanism of Action of Clozapine. Psychother Psychosomat (2018) 87(5):306–7. doi: 10.1159/000491700

CrossRef Full Text | Google Scholar

20. Crilly J. The history of clozapine and its emergence in the US market: a review and analysis. Hist Psychiatry (2007) 18(1):39–60. doi: 10.1177/0957154X07070335

PubMed Abstract | CrossRef Full Text | Google Scholar

21. Frogley C, Taylor D, Dickens G, Picchioni M. A systematic review of the evidence of clozapine’s anti-aggressive effects. Int J Neuropsychopharmacol (2012) 15(9):1351–71. doi: 10.1017/S146114571100201X

PubMed Abstract | CrossRef Full Text | Google Scholar

22. Glazer WM, Dickson RA. Clozapine reduces violence and persistent aggression in schizophrenia. J Clin Psychiatry (1998) 59 Suppl 3:8–14.

PubMed Abstract | Google Scholar

23. Vera I, Rezende L, Molina V, Sanz-Fuentenebro J. Clozapine as treatment of first choice in first psychotic episodes. What do we know? Actas Esp Psiquiatr (2012) 40(5):281–9.

PubMed Abstract | Google Scholar

24. Tiihonen J, Lonnqvist J, Wahlbeck K, Klaukka T, Niskanen L, Tanskanen A, et al. 11-year follow-up of mortality in patients with schizophrenia: a population-based cohort study (FIN11 study). Lancet (2009) 374(9690):620–7. doi: 10.1016/S0140-6736(09)60742-X

PubMed Abstract | CrossRef Full Text | Google Scholar

25. Taipale H, Tanskanen A, Mehtälä J, Vattulainen P, Correll CU, Tiihonen J. 20-year follow-up study of physical morbidity and mortality in relationship to antipsychotic treatment in a nationwide cohort of 62,250 patients with schizophrenia (FIN20). World Psychiatry (2020) 19(1):61–8. doi: 10.1002/wps.20699

PubMed Abstract | CrossRef Full Text | Google Scholar

26. Meltzer HY. Clozapine: balancing safety with superior antipsychotic efficacy. Clin Schizophr Relat Psychoses (2012) 6(3):134–44. doi: 10.3371/CSRP.6.3.5

PubMed Abstract | CrossRef Full Text | Google Scholar

27. Verdoux H, Quiles C, Bachmann CJ, Siskind D. Prescriber and institutional barriers and facilitators of clozapine use: A systematic review. Schizophr Res (2018) 201:10–9. doi: 10.1016/j.schres.2018.05.046

PubMed Abstract | CrossRef Full Text | Google Scholar

28. Mijovic A, MacCabe JH. Clozapine-induced agranulocytosis. Ann Hematol (2020) 99(11):2477–82. doi: 10.1007/s00277-020-04215-y

CrossRef Full Text | Google Scholar

29. Yuen JWY, Kim DD, Procyshyn RM, White RF, Honer WG, Barr AM. Clozapine-Induced Cardiovascular Side Effects and Autonomic Dysfunction: A Systematic Review. Front Neurosci (2018) 12:203. doi: 10.3389/fnins.2018.00203

PubMed Abstract | CrossRef Full Text | Google Scholar

30. Kim DD, White RF, Barr AM, Honer WG, Procyshyn RM. Clozapine, elevated heart rate and QTc prolongation. J Psychiatry Neurosci (2018) 43(1):71–2. doi: 10.1503/jpn.170135

PubMed Abstract | CrossRef Full Text | Google Scholar

31. Kim DD, Lang DJ, Warburton DER, Woodward ML, White RF, Barr AM, et al. Heart-rate response to alpha2-adrenergic receptor antagonism by antipsychotics. Clin Autonomic Res (2017) 27(6):407–10. doi: 10.1007/s10286-017-0444-4

CrossRef Full Text | Google Scholar

32. Kim DD, Barr AM, Lu C, Stewart SE, White RF, Honer WG, et al. Clozapine-Associated Obsessive-Compulsive Symptoms and Their Management: A Systematic Review and Analysis of 107 Reported Cases. Psychother Psychosomat (2020) 89(3):151–60. doi: 10.1159/000505876

CrossRef Full Text | Google Scholar

33. Kim DD, Barr AM, White RF, Honer WG, Procyshyn RM. Clozapine-induced obsessive–compulsive symptoms: mechanisms and treatment. J Psychiatry Neurosci (2019) 44(1):71–2. doi: 10.1503/jpn.180087

PubMed Abstract | CrossRef Full Text | Google Scholar

34. Kim DD, Barr AM, Fredrikson DH, Honer WG, Procyshyn RM. Association between Serum Lipids and Antipsychotic Response in Schizophrenia. Curr Neuropharmacol (2019) 17(9):852–60. doi: 10.2174/1570159x17666190228113348

PubMed Abstract | CrossRef Full Text | Google Scholar

35. Whitney Z, Procyshyn RM, Fredrikson DH, Barr AM. Treatment of clozapine-associated weight gain: a systematic review. Eur J Clin Pharmacol (2015) 71(4):389–401. doi: 10.1007/s00228-015-1807-1

PubMed Abstract | CrossRef Full Text | Google Scholar

36. Alberti KG, Zimmet P, Shaw J. The metabolic syndrome–a new worldwide definition. Lancet (2005) 366(9491):1059–62. doi: 10.1016/s0140-6736(05)67402-8

PubMed Abstract | CrossRef Full Text | Google Scholar

37. Henderson DC. Weight gain with atypical antipsychotics: evidence and insights. J Clin Psychiatry (2007) 68 Suppl 12:18–26.

PubMed Abstract | Google Scholar

38. Lappin JM, Wijaya M, Watkins A, Morell R, Teasdale S, Lederman O, et al. Cardio-metabolic risk and its management in a cohort of clozapine-treated outpatients. Schizophr Res (2018) 199:367–73. doi: 10.1016/j.schres.2018.02.035

PubMed Abstract | CrossRef Full Text | Google Scholar

39. Tso G, Kumar P, Jayasooriya T, Kisely S, Siskind D. Metabolic monitoring and management among clozapine users. Australas Psychiatry (2017) 25(1):48–52. doi: 10.1177/1039856216665282

PubMed Abstract | CrossRef Full Text | Google Scholar

40. Lahti M, Tiihonen J, Wildgust H, Beary M, Hodgson R, Kajantie E, et al. Cardiovascular morbidity, mortality and pharmacotherapy in patients with schizophrenia. Psychol Med (2012) 42(11):2275–85. doi: 10.1017/S0033291712000396

PubMed Abstract | CrossRef Full Text | Google Scholar

41. Henderson DC, Nguyen DD, Copeland PM, Hayden DL, Borba CP, Louie PM, et al. Clozapine, diabetes mellitus, hyperlipidemia, and cardiovascular risks and mortality: results of a 10-year naturalistic study. J Clin Psychiatry (2005) 66(9):1116–21. doi: 10.4088/jcp.v66n0905

PubMed Abstract | CrossRef Full Text | Google Scholar

42. Boyda HN, Tse L, Procyshyn RM, Honer WG, Barr AM. Preclinical models of antipsychotic drug-induced metabolic side effects. Trends Pharmacol Sci (2010) 31(10):484–97. doi: 10.1016/j.tips.2010.07.002

PubMed Abstract | CrossRef Full Text | Google Scholar

43. Gallagher EJ, LeRoith D, Karnieli E. The metabolic syndrome–from insulin resistance to obesity and diabetes. Endocrinol Metab Clin North Am (2008) 37(3):559–79. doi: 10.1016/j.ecl.2008.05.002

PubMed Abstract | CrossRef Full Text | Google Scholar

44. Grundy SM. Metabolic syndrome update. Trends Cardiovasc Med (2016) 26(4):364–73. doi: 10.1016/j.tcm.2015.10.004

PubMed Abstract | CrossRef Full Text | Google Scholar

45. Grundy SM. Obesity, metabolic syndrome, and cardiovascular disease. J Clin Endocrinol Metab (2004) 89(6):2595–600. doi: 10.1210/jc.2004-0372

PubMed Abstract | CrossRef Full Text | Google Scholar

46. Siskind DJ, Leung J, Russell AW, Wysoczanski D, Kisely S. Metformin for Clozapine Associated Obesity: A Systematic Review and Meta-Analysis. PLoS One (2016) 11(6):e0156208. doi: 10.1371/journal.pone.0156208

PubMed Abstract | CrossRef Full Text | Google Scholar

47. Newcomer JW, Haupt DW. The metabolic effects of antipsychotic medications. Can J Psychiatry (2006) 51(8):480–91. doi: 10.1177/070674370605100803

PubMed Abstract | CrossRef Full Text | Google Scholar

48. Dagenais GR, Yi Q, Mann JF, Bosch J, Pogue J, Yusuf S. Prognostic impact of body weight and abdominal obesity in women and men with cardiovascular disease. Am Heart J (2005) 149(1):54–60. doi: 10.1016/j.ahj.2004.07.009

PubMed Abstract | CrossRef Full Text | Google Scholar

49. Henderson DC, Fan X, Sharma B, Copeland PM, Borba CP, Freudenreich O, et al. Waist circumference is the best anthropometric predictor for insulin resistance in nondiabetic patients with schizophrenia treated with clozapine but not olanzapine. J Psychiatr Pract (2009) 15(4):251–61. doi: 10.1097/01.pra.0000358312.99233.ef

PubMed Abstract | CrossRef Full Text | Google Scholar

50. Van Gaal LF. Long-term health considerations in schizophrenia: metabolic effects and the role of abdominal adiposity. Eur Neuropsychopharmacol (2006) 16 Suppl 3:S142–8. doi: 10.1016/j.euroneuro.2006.06.005

PubMed Abstract | CrossRef Full Text | Google Scholar

51. Fredrikson DH, Boyda HN, Tse L, Whitney Z, Pattison MA, Ott FJ, et al. Improving metabolic and cardiovascular health at an early psychosis intervention program in Vancouver, Canada. Front Psychiatry (2014) 5:105. doi: 10.3389/fpsyt.2014.00105

PubMed Abstract | CrossRef Full Text | Google Scholar

52. Lamberti JS, Olson D, Crilly JF, Olivares T, Williams GC, Tu X, et al. Prevalence of the metabolic syndrome among patients receiving clozapine. Am J Psychiatry (2006) 163(7):1273–6. doi: 10.1176/appi.ajp.163.7.1273

PubMed Abstract | CrossRef Full Text | Google Scholar

53. Allison DB, Casey DE. Antipsychotic-induced weight gain: a review of the literature. J Clin Psychiatry (2001) 62 Suppl 7:22–31.

PubMed Abstract | Google Scholar

54. Henderson DC, Cagliero E, Gray C, Nasrallah RA, Hayden DL, Schoenfeld DA, et al. Clozapine, diabetes mellitus, weight gain, and lipid abnormalities: A five-year naturalistic study. Am J Psychiatry (2000) 157(6):975–81. doi: 10.1176/appi.ajp.157.6.975

PubMed Abstract | CrossRef Full Text | Google Scholar

55. Nasrallah H. A review of the effect of atypical antipsychotics on weight. Psychoneuroendocrinology (2003) 28 Suppl 1:83–96. doi: 10.1016/s0306-4530(02)00114-2

PubMed Abstract | CrossRef Full Text | Google Scholar

56. Hubert HB, Feinleib M, McNamara PM, Castelli WP. Obesity as an independent risk factor for cardiovascular disease: a 26-year follow-up of participants in the Framingham Heart Study. Circulation (1983) 67(5):968–77. doi: 10.1161/01.cir.67.5.968

PubMed Abstract | CrossRef Full Text | Google Scholar

57. Siskind DJ, Russell AW, Gamble C, Winckel K, Mayfield K, Hollingworth S, et al. Treatment of clozapine-associated obesity and diabetes with exenatide in adults with schizophrenia: A randomized controlled trial (CODEX). Diabetes Obes Metab (2018) 20(4):1050–5. doi: 10.1111/dom.13167

PubMed Abstract | CrossRef Full Text | Google Scholar

58. Lau SL, Muir C, Assur Y, Beach R, Tran B, Bartrop R, et al. Predicting Weight Gain in Patients Treated With Clozapine: The Role of Sex, Body Mass Index, and Smoking. J Clin Psychopharmacol (2016) 36(2):120–4. doi: 10.1097/jcp.0000000000000476

PubMed Abstract | CrossRef Full Text | Google Scholar

59. Contreras-Shannon V, Heart DL, Paredes RM, Navaira E, Catano G, Maffi SK, et al. Clozapine-induced mitochondria alterations and inflammation in brain and insulin-responsive cells. PLoS One (2013) 8(3):e59012. doi: 10.1371/journal.pone.0059012

PubMed Abstract | CrossRef Full Text | Google Scholar

60. Ferreira V, Grajales D, Valverde AM. Adipose tissue as a target for second-generation (atypical) antipsychotics: A molecular view. Biochim Biophys Acta Mol Cell Biol Lipids (2020) 1865(2):158534. doi: 10.1016/j.bbalip.2019.158534

PubMed Abstract | CrossRef Full Text | Google Scholar

61. Klemettila JP, Kampman O, Seppala N, Viikki M, Hamalainen M, Moilanen E, et al. Cytokine and adipokine alterations in patients with schizophrenia treated with clozapine. Psychiatry Res (2014) 218(3):277–83. doi: 10.1016/j.psychres.2014.04.049

PubMed Abstract | CrossRef Full Text | Google Scholar

62. Klemettila JP, Kampman O, Seppala N, Viikki M, Hamalainen M, Moilanen E, et al. Resistin as an inflammatory marker in patients with schizophrenia treated with clozapine. Nord J Psychiatry (2017) 71(2):89–95. doi: 10.1080/08039488.2016.1230649

PubMed Abstract | CrossRef Full Text | Google Scholar

63. Mori N, McEvoy JP, Miller BJ. Total and differential white blood cell counts, inflammatory markers, adipokines, and the metabolic syndrome in phase 1 of the clinical antipsychotic trials of intervention effectiveness study. Schizophr Res (2015) 169(1-3):30–5. doi: 10.1016/j.schres.2015.10.001

PubMed Abstract | CrossRef Full Text | Google Scholar

64. Harwood HJ Jr. The adipocyte as an endocrine organ in the regulation of metabolic homeostasis. Neuropharmacology (2012) 63(1):57–75. doi: 10.1016/j.neuropharm.2011.12.010

PubMed Abstract | CrossRef Full Text | Google Scholar

65. Hotamisligil GS, Arner P, Caro JF, Atkinson RL, Spiegelman BM. Increased adipose tissue expression of tumor necrosis factor-alpha in human obesity and insulin resistance. J Clin Invest (1995) 95(5):2409–15. doi: 10.1172/JCI117936

PubMed Abstract | CrossRef Full Text | Google Scholar

66. Leonard BE, Schwarz M, Myint AM. The metabolic syndrome in schizophrenia: is inflammation a contributing cause? J Psychopharmacol (2012) 26(5 Suppl):33–41. doi: 10.1177/0269881111431622

PubMed Abstract | CrossRef Full Text | Google Scholar

67. Laurencikiene J, van Harmelen V, Arvidsson Nordstrom E, Dicker A, Blomqvist L, Naslund E, et al. NF-kappaB is important for TNF-alpha-induced lipolysis in human adipocytes. J Lipid Res (2007) 48(5):1069–77. doi: 10.1194/jlr.M600471-JLR200

PubMed Abstract | CrossRef Full Text | Google Scholar

68. Nieto-Vazquez I, Fernandez-Veledo S, Kramer DK, Vila-Bedmar R, Garcia-Guerra L, Lorenzo M. Insulin resistance associated to obesity: the link TNF-alpha. Arch Physiol Biochem (2008) 114(3):183–94. doi: 10.1080/13813450802181047

PubMed Abstract | CrossRef Full Text | Google Scholar

69. Uysal KT, Wiesbrock SM, Marino MW, Hotamisligil GS. Protection from obesity-induced insulin resistance in mice lacking TNF-alpha function. Nature (1997) 389(6651):610–4. doi: 10.1038/39335

PubMed Abstract | CrossRef Full Text | Google Scholar

70. Kadowaki T, Yamauchi T, Kubota N, Hara K, Ueki K, Tobe K. Adiponectin and adiponectin receptors in insulin resistance, diabetes, and the metabolic syndrome. J Clin Invest (2006) 116(7):1784–92. doi: 10.1172/JCI29126

PubMed Abstract | CrossRef Full Text | Google Scholar

71. Yamauchi T, Kamon J, Minokoshi Y, Ito Y, Waki H, Uchida S, et al. Adiponectin stimulates glucose utilization and fatty-acid oxidation by activating AMP-activated protein kinase. Nat Med (2002) 8(11):1288–95. doi: 10.1038/nm788

PubMed Abstract | CrossRef Full Text | Google Scholar

72. Tsuchida A, Yamauchi T, Ito Y, Hada Y, Maki T, Takekawa S, et al. Insulin/Foxo1 pathway regulates expression levels of adiponectin receptors and adiponectin sensitivity. J Biol Chem (2004) 279(29):30817–22. doi: 10.1074/jbc.M402367200

PubMed Abstract | CrossRef Full Text | Google Scholar

73. Phillips SA, Ciaraldi TP, Kong AP, Bandukwala R, Aroda V, Carter L, et al. Modulation of circulating and adipose tissue adiponectin levels by antidiabetic therapy. Diabetes (2003) 52(3):667–74. doi: 10.2337/diabetes.52.3.667

PubMed Abstract | CrossRef Full Text | Google Scholar

74. Tsuchida A, Yamauchi T, Takekawa S, Hada Y, Ito Y, Maki T, et al. Peroxisome proliferator-activated receptor (PPAR)alpha activation increases adiponectin receptors and reduces obesity-related inflammation in adipose tissue: comparison of activation of PPARalpha, PPARgamma, and their combination. Diabetes (2005) 54(12):3358–70. doi: 10.2337/diabetes.54.12.3358

PubMed Abstract | CrossRef Full Text | Google Scholar

75. Yamauchi T, Iwabu M, Okada-Iwabu M, Kadowaki T. Adiponectin receptors: a review of their structure, function and how they work. Best Pract Res Clin Endocrinol Metab (2014) 28(1):15–23. doi: 10.1016/j.beem.2013.09.003

PubMed Abstract | CrossRef Full Text | Google Scholar

76. Astapova O, Leff T. Adiponectin and PPARgamma: cooperative and interdependent actions of two key regulators of metabolism. Vitam Horm (2012) 90:143–62. doi: 10.1016/B978-0-12-398313-8.00006-3

PubMed Abstract | CrossRef Full Text | Google Scholar

77. Bai YM, Chen TT, Yang WS, Chi YC, Lin CC, Liou YJ, et al. Association of adiponectin and metabolic syndrome among patients taking atypical antipsychotics for schizophrenia: a cohort study. Schizophr Res (2009) 111(1-3):1–8. doi: 10.1016/j.schres.2009.03.014

PubMed Abstract | CrossRef Full Text | Google Scholar

78. Abate N, Sallam HS, Rizzo M, Nikolic D, Obradovic M, Bjelogrlic P, et al. Resistin: an inflammatory cytokine. Role in cardiovascular diseases, diabetes and the metabolic syndrome. Curr Pharm Des (2014) 20(31):4961–9. doi: 10.2174/1381612819666131206103102

PubMed Abstract | CrossRef Full Text | Google Scholar

79. Jamaluddin MS, Weakley SM, Yao Q, Chen C. Resistin: functional roles and therapeutic considerations for cardiovascular disease. Br J Pharmacol (2012) 165(3):622–32. doi: 10.1111/j.1476-5381.2011.01369.x

PubMed Abstract | CrossRef Full Text | Google Scholar

80. Park HK, Kwak MK, Kim HJ, Ahima RS. Linking resistin, inflammation, and cardiometabolic diseases. Korean J Intern Med (2017) 32(2):239–47. doi: 10.3904/kjim.2016.229

PubMed Abstract | CrossRef Full Text | Google Scholar

81. Steppan CM, Bailey ST, Bhat S, Brown EJ, Banerjee RR, Wright CM, et al. The hormone resistin links obesity to diabetes. Nature (2001) 409(6818):307–12. doi: 10.1038/35053000

PubMed Abstract | CrossRef Full Text | Google Scholar

82. Huang X, Yang Z. Resistin’s, obesity and insulin resistance: the continuing disconnect between rodents and humans. J Endocrinol Invest (2016) 39(6):607–15. doi: 10.1007/s40618-015-0408-2

PubMed Abstract | CrossRef Full Text | Google Scholar

83. Park HK, Ahima RS. Resistin in rodents and humans. Diabetes Metab J (2013) 37(6):404–14. doi: 10.4093/dmj.2013.37.6.404

PubMed Abstract | CrossRef Full Text | Google Scholar

84. Schwartz DR, Lazar MA. Human resistin: found in translation from mouse to man. Trends Endocrinol Metab (2011) 22(7):259–65. doi: 10.1016/j.tem.2011.03.005

PubMed Abstract | CrossRef Full Text | Google Scholar

85. Kroeze WK, Hufeisen SJ, Popadak BA, Renock SM, Steinberg S, Ernsberger P, et al. H1-histamine receptor affinity predicts short-term weight gain for typical and atypical antipsychotic drugs. Neuropsychopharmacol (2003) 28(3):519–26. doi: 10.1038/sj.npp.1300027

CrossRef Full Text | Google Scholar

86. Masaki T, Yoshimatsu H, Chiba S, Watanabe T, Sakata T. Targeted disruption of histamine H1-receptor attenuates regulatory effects of leptin on feeding, adiposity, and UCP family in mice. Diabetes (2001) 50(2):385–91. doi: 10.2337/diabetes.50.2.385

PubMed Abstract | CrossRef Full Text | Google Scholar

87. Kim SF, Huang AS, Snowman AM, Teuscher C, Snyder SH. From the Cover: Antipsychotic drug-induced weight gain mediated by histamine H1 receptor-linked activation of hypothalamic AMP-kinase. Proc Natl Acad Sci U S A (2007) 104(9):3456–9. doi: 10.1073/pnas.0611417104

PubMed Abstract | CrossRef Full Text | Google Scholar

88. Minokoshi Y, Alquier T, Furukawa N, Kim YB, Lee A, Xue B, et al. AMP-kinase regulates food intake by responding to hormonal and nutrient signals in the hypothalamus. Nature (2004) 428(6982):569–74. doi: 10.1038/nature02440

PubMed Abstract | CrossRef Full Text | Google Scholar

89. Masaki T, Yoshimatsu H, Chiba S, Watanabe T, Sakata T. Central infusion of histamine reduces fat accumulation and upregulates UCP family in leptin-resistant obese mice. Diabetes (2001) 50(2):376–84. doi: 10.2337/diabetes.50.2.376

PubMed Abstract | CrossRef Full Text | Google Scholar

90. Cummings DE. Ghrelin and the short- and long-term regulation of appetite and body weight. Physiol Behav (2006) 89(1):71–84. doi: 10.1016/j.physbeh.2006.05.022

PubMed Abstract | CrossRef Full Text | Google Scholar

91. Wiedmer P, Nogueiras R, Broglio F, D’Alessio D, Tschop MH. Ghrelin, obesity and diabetes. Nat Clin Pract Endocrinol Metab (2007) 3(10):705–12. doi: 10.1038/ncpendmet0625

PubMed Abstract | CrossRef Full Text | Google Scholar

92. Purnell JQ, Weigle DS, Breen P, Cummings DE. Ghrelin levels correlate with insulin levels, insulin resistance, and high-density lipoprotein cholesterol, but not with gender, menopausal status, or cortisol levels in humans. J Clin Endocrinol Metab (2003) 88(12):5747–52. doi: 10.1210/jc.2003-030513

PubMed Abstract | CrossRef Full Text | Google Scholar

93. Esen-Danaci A, Sarandol A, Taneli F, Yurtsever F, Ozlen N. Effects of second generation antipsychotics on leptin and ghrelin. Prog Neuropsychopharmacol Biol Psychiatry (2008) 32(6):1434–8. doi: 10.1016/j.pnpbp.2008.03.015

PubMed Abstract | CrossRef Full Text | Google Scholar

94. Gehlert DR. Role of hypothalamic neuropeptide Y in feeding and obesity. Neuropeptides (1999) 33(5):329–38. doi: 10.1054/npep.1999.0057

PubMed Abstract | CrossRef Full Text | Google Scholar

95. Kuo LE, Kitlinska JB, Tilan JU, Li L, Baker SB, Johnson MD, et al. Neuropeptide Y acts directly in the periphery on fat tissue and mediates stress-induced obesity and metabolic syndrome. Nat Med (2007) 13(7):803–11. doi: 10.1038/nm1611

PubMed Abstract | CrossRef Full Text | Google Scholar

96. Kirk SL, Cahir M, Reynolds GP. Clozapine, but not haloperidol, increases neuropeptide Y neuronal expression in the rat hypothalamus. J Psychopharmacol (2006) 20(4):577–9. doi: 10.1177/0269881106061199

PubMed Abstract | CrossRef Full Text | Google Scholar

97. Tiwari AK, Brandl EJ, Weber C, Likhodi O, Zai CC, Hahn MK, et al. Association of a functional polymorphism in neuropeptide Y with antipsychotic-induced weight gain in schizophrenia patients. J Clin Psychopharmacol (2013) 33(1):11–7. doi: 10.1097/JCP.0b013e31827d145a

PubMed Abstract | CrossRef Full Text | Google Scholar

98. Ste Marie L, Miura GI, Marsh DJ, Yagaloff K, Palmiter RD. A metabolic defect promotes obesity in mice lacking melanocortin-4 receptors. Proc Natl Acad Sci U S A (2000) 97(22):12339–44. doi: 10.1073/pnas.220409497

PubMed Abstract | CrossRef Full Text | Google Scholar

99. Chowdhury NI, Tiwari AK, Souza RP, Zai CC, Shaikh SA, Chen S, et al. Genetic association study between antipsychotic-induced weight gain and the melanocortin-4 receptor gene. Pharmacogenom J (2013) 13(3):272–9. doi: 10.1038/tpj.2011.66

CrossRef Full Text | Google Scholar

100. Farooqi IS, Keogh JM, Yeo GS, Lank EJ, Cheetham T, O’Rahilly S. Clinical spectrum of obesity and mutations in the melanocortin 4 receptor gene. N Engl J Med (2003) 348(12):1085–95. doi: 10.1056/NEJMoa022050

PubMed Abstract | CrossRef Full Text | Google Scholar

101. Mayfield K, Siskind D, Winckel K, Hollingworth S, Kisely S, Russell AW. Treatment of clozapine-associated obesity and diabetes with exenatide (CODEX) in adults with schizophrenia: study protocol for a pilot randomised controlled trial. BJPsych Open (2015) 1(1):67–73. doi: 10.1192/bjpo.bp.115.001073

PubMed Abstract | CrossRef Full Text | Google Scholar

102. Mayfield K, Siskind D, Winckel K, Russell AW, Kisely S, Smith G, et al. Glucagon-like peptide-1 agonists combating clozapine-associated obesity and diabetes. J Psychopharmacol (2016) 30(3):227–36. doi: 10.1177/0269881115625496

PubMed Abstract | CrossRef Full Text | Google Scholar

103. Smith GC, Zhang ZY, Mulvey T, Petersen N, Lach S, Xiu P, et al. Clozapine directly increases insulin and glucagon secretion from islets: implications for impairment of glucose tolerance. Schizophr Res (2014) 157(1–3):128–33. doi: 10.1016/j.schres.2014.05.003

PubMed Abstract | CrossRef Full Text | Google Scholar

104. Smith GC, Vickers MH, Cognard E, Shepherd PR. Clozapine and quetiapine acutely reduce glucagon-like peptide-1 production and increase glucagon release in obese rats: implications for glucose metabolism and food choice behaviour. Schizophr Res (2009) 115(1):30–40. doi: 10.1016/j.schres.2009.07.011

PubMed Abstract | CrossRef Full Text | Google Scholar

105. Siskind D, Hahn M, Correll CU, Fink-Jensen A, Russell AW, Bak N, et al. Glucagon-like peptide-1 receptor agonists for antipsychotic-associated cardio-metabolic risk factors: A systematic review and individual participant data meta-analysis. Diabetes Obes Metab (2019) 21(2):293–302. doi: 10.1111/dom.13522

PubMed Abstract | CrossRef Full Text | Google Scholar

106. Larsen JR, Vedtofte L, Jakobsen MSL, Jespersen HR, Jakobsen MI, Svensson CK, et al. Effect of Liraglutide Treatment on Prediabetes and Overweight or Obesity in Clozapine- or Olanzapine-Treated Patients With Schizophrenia Spectrum Disorder: A Randomized Clinical Trial. JAMA Psychiatry (2017) 74(7):719–28. doi: 10.1001/jamapsychiatry.2017.1220

PubMed Abstract | CrossRef Full Text | Google Scholar

107. Ishoy PL, Knop FK, Broberg BV, Bak N, Andersen UB, Jorgensen NR, et al. Effect of GLP-1 receptor agonist treatment on body weight in obese antipsychotic-treated patients with schizophrenia: a randomized, placebo-controlled trial. Diabetes Obes Metab (2017) 19(2):162–71. doi: 10.1111/dom.12795

PubMed Abstract | CrossRef Full Text | Google Scholar

108. Oh JE, Cho YM, Kwak SN, Kim JH, Lee KW, Jung H, et al. Inhibition of mouse brown adipocyte differentiation by second-generation antipsychotics. Exp Mol Med (2012) 44(9):545–53. doi: 10.3858/emm.2012.44.9.062

PubMed Abstract | CrossRef Full Text | Google Scholar

109. Yang X, Enerback S, Smith U. Reduced expression of FOXC2 and brown adipogenic genes in human subjects with insulin resistance. Obes Res (2003) 11(10):1182–91. doi: 10.1038/oby.2003.163

PubMed Abstract | CrossRef Full Text | Google Scholar

110. Stamler J, Vaccaro O, Neaton JD, Wentworth D. Diabetes, other risk factors, and 12-yr cardiovascular mortality for men screened in the Multiple Risk Factor Intervention Trial. Diabetes Care (1993) 16(2):434–44. doi: 10.2337/diacare.16.2.434

PubMed Abstract | CrossRef Full Text | Google Scholar

111. Ross R. The pathogenesis of atherosclerosis: a perspective for the 1990s. Nature (1993) 362(6423):801–9. doi: 10.1038/362801a0

PubMed Abstract | CrossRef Full Text | Google Scholar

112. Gaulin BD, Markowitz JS, Caley CF, Nesbitt LA, Dufresne RL. Clozapine-associated elevation in serum triglycerides. Am J Psychiatry (1999) 156(8):1270–2. doi: 10.1176/ajp.156.8.1270

PubMed Abstract | CrossRef Full Text | Google Scholar

113. Procyshyn RM, Wasan KM, Thornton AE, Barr AM, Chen EY, Pomarol-Clotet E, et al. Changes in serum lipids, independent of weight, are associated with changes in symptoms during long-term clozapine treatment. J Psychiatry Neurosci (2007) 32(5):331–8.

PubMed Abstract | Google Scholar

114. Baymiller SP, Ball P, McMahon RP, Buchanan RW. Serum glucose and lipid changes during the course of clozapine treatment: the effect of concurrent beta-adrenergic antagonist treatment. Schizophr Res (2003) 59(1):49–57. doi: 10.1016/s0920-9964(02)00158-5

PubMed Abstract | CrossRef Full Text | Google Scholar

115. Newcomer JW. Antipsychotic medications: metabolic and cardiovascular risk. J Clin Psychiatry (2007) 68 Suppl 4:8–13.

PubMed Abstract | Google Scholar

116. Ghaeli P, Dufresne RL. Elevated serum triglycerides with clozapine resolved with risperidone in four patients. Pharmacotherapy (1999) 19(9):1099–101. doi: 10.1592/phco.19.13.1099.31586

PubMed Abstract | CrossRef Full Text | Google Scholar

117. Kim DD, Barr AM, Thornton AE, Honer WG, Procyshyn RM. Statin add-on therapy for schizophrenia: Is the effect the same for clozapine? Psychiatry Res (2018) 263:289–90. doi: 10.1016/j.psychres.2018.02.039

PubMed Abstract | CrossRef Full Text | Google Scholar

118. Gershkovich P, Sivak O, Sharma A, Barr AM, Procyshyn R, Wasan KM. Effect of hypertriglyceridemia on the pharmacokinetics and blood-brain barrier penetration of clozapine and norclozapine following administration to rats. Eur Neuropsychopharmacol (2010) 20(8):545–52. doi: 10.1016/j.euroneuro.2010.01.002

PubMed Abstract | CrossRef Full Text | Google Scholar

119. Barakauskas VE, Ypsilanti AR, Barr AM, Innis SM, Honer WG, Beasley CL. Effects of sub-chronic clozapine and haloperidol administration on brain lipid levels. Prog Neuropsychopharmacol Biol Psychiatry (2010) 34(4):669–73. doi: 10.1016/j.pnpbp.2010.03.010

PubMed Abstract | CrossRef Full Text | Google Scholar

120. Feresten AH, Barakauskas V, Ypsilanti A, Barr AM, Beasley CL. Increased expression of glial fibrillary acidic protein in prefrontal cortex in psychotic illness. Schizophr Res (2013) 150(1):252–7. doi: 10.1016/j.schres.2013.07.024

PubMed Abstract | CrossRef Full Text | Google Scholar

121. Tse L, Procyshyn RM, Fredrikson DH, Boyda HN, Honer WG, Barr AM. Pharmacological treatment of antipsychotic-induced dyslipidemia and hypertension. Int Clin Psychopharmacol (2014) 29(3):125–37. doi: 10.1097/YIC.0000000000000014

PubMed Abstract | CrossRef Full Text | Google Scholar

122. Landry P, Dimitri E, Tessier S, Legare N. Efficacy of lipid-lowering medications in patients treated with clozapine: a naturalistic study. J Clin Psychopharmacol (2008) 28(3):348–9. doi: 10.1097/JCP.0b013e3181727592

PubMed Abstract | CrossRef Full Text | Google Scholar

123. Ojala K, Repo-Tiihonen E, Tiihonen J, Niskanen L. Statins are effective in treating dyslipidemia among psychiatric patients using second-generation antipsychotic agents. J Psychopharmacol (2008) 22(1):33–8. doi: 10.1177/0269881107077815

PubMed Abstract | CrossRef Full Text | Google Scholar

124. Sattar N, Preiss D, Murray HM, Welsh P, Buckley BM, de Craen AJ, et al. Statins and risk of incident diabetes: a collaborative meta-analysis of randomised statin trials. Lancet (2010) 375(9716):735–42. doi: 10.1016/S0140-6736(09)61965-6

PubMed Abstract | CrossRef Full Text | Google Scholar

125. Bravo EL. Metabolic factors and the sympathetic nervous system. Am J Hypertens (1989) 2(12 Pt 2):339S–44S. doi: 10.1093/ajh/2.12.339s

PubMed Abstract | CrossRef Full Text | Google Scholar

126. Tentolouris N, Argyrakopoulou G, Katsilambros N. Perturbed autonomic nervous system function in metabolic syndrome. Neuromol Med (2008) 10(3):169–78. doi: 10.1007/s12017-008-8022-5

CrossRef Full Text | Google Scholar

127. Boyda HN, Procyshyn RM, Pang CC, Barr AM. Peripheral adrenoceptors: the impetus behind glucose dysregulation and insulin resistance. J Neuroendocrinol (2013) 25(3):217–28. doi: 10.1111/jne.12002

PubMed Abstract | CrossRef Full Text | Google Scholar

128. Nikolic D, Castellino G, Banach M, Toth PP, Ivanova E, Orekhov AN, et al. PPAR Agonists, Atherogenic Dyslipidemia and Cardiovascular Risk. Curr Pharm Des (2017) 23(6):894–902. doi: 10.2174/1381612822666161006151134

PubMed Abstract | CrossRef Full Text | Google Scholar

129. Berger JP, Akiyama TE, Meinke PT. PPARs: therapeutic targets for metabolic disease. Trends Pharmacol Sci (2005) 26(5):244–51. doi: 10.1016/j.tips.2005.03.003

PubMed Abstract | CrossRef Full Text | Google Scholar

130. Ahmed M, Griffin D, O’Toole R, McDonald C. Clozapine-induced severe mixed hyperlipidemia: a case report. Gen Hosp Psychiatry (2009) 31(1):93–6. doi: 10.1016/j.genhosppsych.2008.07.003

PubMed Abstract | CrossRef Full Text | Google Scholar

131. O’Neill S, O’Driscoll L. Metabolic syndrome: a closer look at the growing epidemic and its associated pathologies. Obes Rev (2015) 16(1):1–12. doi: 10.1111/obr.12229

PubMed Abstract | CrossRef Full Text | Google Scholar

132. Sowers JR. Insulin resistance and hypertension. Mol Cell Endocrinol (1990) 74(2):C87–9. doi: 10.1016/0303-7207(90)90110-t

PubMed Abstract | CrossRef Full Text | Google Scholar

133. Sowers JR. Insulin resistance and hypertension. Am J Physiol Heart Circ Physiol (2004) 286(5):H1597–602. doi: 10.1152/ajpheart.00026.2004

PubMed Abstract | CrossRef Full Text | Google Scholar

134. Ginsberg HN. Insulin resistance and cardiovascular disease. J Clin Invest (2000) 106(4):453–8. doi: 10.1172/JCI10762

PubMed Abstract | CrossRef Full Text | Google Scholar

135. Reaven GM. Banting lecture 1988. Role of insulin resistance in human disease. Diabetes (1988) 37(12):1595–607. doi: 10.2337/diab.37.12.1595

PubMed Abstract | CrossRef Full Text | Google Scholar

136. Henderson DC, Daley TB, Kunkel L, Rodrigues-Scott M, Koul P, Hayden D. Clozapine and hypertension: a chart review of 82 patients. J Clin Psychiatry (2004) 65(5):686–9. doi: 10.4088/jcp.v65n0514

PubMed Abstract | CrossRef Full Text | Google Scholar

137. Lund BC, Perry PJ, Brooks JM, Arndt S. Clozapine use in patients with schizophrenia and the risk of diabetes, hyperlipidemia, and hypertension: a claims-based approach. Arch Gen Psychiatry (2001) 58(12):1172–6. doi: 10.1001/archpsyc.58.12.1172

PubMed Abstract | CrossRef Full Text | Google Scholar

138. Nathan DM, Buse JB, Davidson MB, Ferrannini E, Holman RR, Sherwin R, et al. Medical management of hyperglycemia in type 2 diabetes: a consensus algorithm for the initiation and adjustment of therapy: a consensus statement of the American Diabetes Association and the European Association for the Study of Diabetes. Diabetes Care (2009) 32(1):193–203. doi: 10.2337/dc08-9025

PubMed Abstract | CrossRef Full Text | Google Scholar

139. Association AD. Diagnosis and classification of diabetes mellitus. Diabetes Care (2010) 33 Suppl 1:S62–9. doi: 10.2337/dc10-S062

PubMed Abstract | CrossRef Full Text | Google Scholar

140. American Diabetes Association. Diagnosis and classification of diabetes mellitus. Diabetes Care (2010) 33 Suppl 1(Suppl 1):S62–9. doi: 10.2337/dc10-S062

PubMed Abstract | CrossRef Full Text | Google Scholar

141. Brownlee M. Biochemistry and molecular cell biology of diabetic complications. Nature (2001) 414(6865):813–20. doi: 10.1038/414813a

PubMed Abstract | CrossRef Full Text | Google Scholar

142. Brownlee M. The pathobiology of diabetic complications: a unifying mechanism. Diabetes (2005) 54(6):1615–25. doi: 10.2337/diabetes.54.6.1615

PubMed Abstract | CrossRef Full Text | Google Scholar

143. Du X, Matsumura T, Edelstein D, Rossetti L, Zsengeller Z, Szabo C, et al. Inhibition of GAPDH activity by poly(ADP-ribose) polymerase activates three major pathways of hyperglycemic damage in endothelial cells. J Clin Invest (2003) 112(7):1049–57. doi: 10.1172/JCI18127

PubMed Abstract | CrossRef Full Text | Google Scholar

144. Fiorentino TV, Prioletta A, Zuo P, Folli F. Hyperglycemia-induced oxidative stress and its role in diabetes mellitus related cardiovascular diseases. Curr Pharm Des (2013) 19(32):5695–703. doi: 10.2174/1381612811319320005

PubMed Abstract | CrossRef Full Text | Google Scholar

145. Andreazza AC, Barakauskas VE, Fazeli S, Feresten A, Shao L, Wei V, et al. Effects of haloperidol and clozapine administration on oxidative stress in rat brain, liver and serum. Neurosci Lett (2015) 591:36–40. doi: 10.1016/j.neulet.2015.02.028

PubMed Abstract | CrossRef Full Text | Google Scholar

146. Kamran A, Doraiswamy PM, Jane JL, Hammett EB, Dunn L. Severe hyperglycemia associated with high doses of clozapine. Am J Psychiatry (1994) 151(9):1395. doi: 10.1176/ajp.151.9.1395a

PubMed Abstract | CrossRef Full Text | Google Scholar

147. Lindenmayer JP, Nathan AM, Smith RC. Hyperglycemia associated with the use of atypical antipsychotics. J Clin Psychiatry (2001) 62 Suppl 23:30–8.

PubMed Abstract | Google Scholar

148. Yazici KM, Erbas T, Yazici AH. The effect of clozapine on glucose metabolism. Exp Clin Endocrinol Diabetes (1998) 106(6):475–7. doi: 10.1055/s-0029-1212019

PubMed Abstract | CrossRef Full Text | Google Scholar

149. Wirshing DA, Spellberg BJ, Erhart SM, Marder SR, Wirshing WC. Novel antipsychotics and new onset diabetes. Biol Psychiatry (1998) 44(8):778–83. doi: 10.1016/s0006-3223(98)00100-0

PubMed Abstract | CrossRef Full Text | Google Scholar

150. Popli AP, Konicki PE, Jurjus GJ, Fuller MA, Jaskiw GE. Clozapine and associated diabetes mellitus. J Clin Psychiatry (1997) 58(3):108–11. doi: 10.4088/jcp.v58n0304

PubMed Abstract | CrossRef Full Text | Google Scholar

151. McIntyre RS, McCann SM, Kennedy SH. Antipsychotic metabolic effects: weight gain, diabetes mellitus, and lipid abnormalities. Can J Psychiatry (2001) 46(3):273–81. doi: 10.1177/070674370104600308

PubMed Abstract | CrossRef Full Text | Google Scholar

152. Newcomer JW, Haupt DW, Fucetola R, Melson AK, Schweiger JA, Cooper BP, et al. Abnormalities in glucose regulation during antipsychotic treatment of schizophrenia. Arch Gen Psychiatry (2002) 59(4):337–45. doi: 10.1001/archpsyc.59.4.337

PubMed Abstract | CrossRef Full Text | Google Scholar

153. Sernyak MJ, Leslie DL, Alarcon RD, Losonczy MF, Rosenheck R. Association of diabetes mellitus with use of atypical neuroleptics in the treatment of schizophrenia. Am J Psychiatry (2002) 159(4):561–6. doi: 10.1176/appi.ajp.159.4.561

PubMed Abstract | CrossRef Full Text | Google Scholar

154. Hahn M, Chintoh A, Giacca A, Xu L, Lam L, Mann S, et al. Atypical antipsychotics and effects of muscarinic, serotonergic, dopaminergic and histaminergic receptor binding on insulin secretion in vivo: an animal model. Schizophr Res (2011) 131(1-3):90–5. doi: 10.1016/j.schres.2011.06.004

PubMed Abstract | CrossRef Full Text | Google Scholar

155. Gautam D, Han SJ, Hamdan FF, Jeon J, Li B, Li JH, et al. A critical role for beta cell M3 muscarinic acetylcholine receptors in regulating insulin release and blood glucose homeostasis in vivo. Cell Metab (2006) 3(6):449–61. doi: 10.1016/j.cmet.2006.04.009

PubMed Abstract | CrossRef Full Text | Google Scholar

156. Guenette MD, Giacca A, Hahn M, Teo C, Lam L, Chintoh A, et al. Atypical antipsychotics and effects of adrenergic and serotonergic receptor binding on insulin secretion in-vivo: an animal model. Schizophr Res (2013) 146(1-3):162–9. doi: 10.1016/j.schres.2013.02.023

PubMed Abstract | CrossRef Full Text | Google Scholar

157. Chintoh AF, Mann SW, Lam L, Giacca A, Fletcher P, Nobrega J, et al. Insulin resistance and secretion in vivo: Effects of different antipsychotics in an animal model. Schizophr Res (2009) 108(1-3):127–33. doi: 10.1016/j.schres.2008.12.012

PubMed Abstract | CrossRef Full Text | Google Scholar

158. Barth E, Albuszies G, Baumgart K, Matejovic M, Wachter U, Vogt J, et al. Glucose metabolism and catecholamines. Crit Care Med (2007) 35(9 Suppl):S508–18. doi: 10.1097/01.ccm.0000278047.06965.20

PubMed Abstract | CrossRef Full Text | Google Scholar

159. Boyda HN, Ho AA, Tse L, Procyshyn RM, Yuen JWY, Kim DD, et al. Differential Effects of Acute Treatment With Antipsychotic Drugs on Peripheral Catecholamines. Front Psychiatry (2020) 11:617428. doi: 10.3389/fpsyt.2020.617428

PubMed Abstract | CrossRef Full Text | Google Scholar

160. Yuen JWY, Wu C, Wang CK, Kim DD, Procyshyn RM, Honer WG, et al. A comparison of the effects of clozapine and its metabolite norclozapine on metabolic dysregulation in rodent models. Neuropharmacology (2019) 175:107717. doi: 10.1016/j.neuropharm.2019.107717

PubMed Abstract | CrossRef Full Text | Google Scholar

161. Boyda HN, Procyshyn RM, Tse L, Xu J, Jin CH, Wong D, et al. Antipsychotic polypharmacy increases metabolic dysregulation in female rats. Exp Clin Psychopharmacol (2013) 21(2):164–71. doi: 10.1037/a0031228

PubMed Abstract | CrossRef Full Text | Google Scholar

162. Boyda HN, Tse L, Procyshyn RM, Wong D, Wu TK, Pang CC, et al. A parametric study of the acute effects of antipsychotic drugs on glucose sensitivity in an animal model. Prog Neuropsychopharmacol Biol Psychiatry (2010) 34(6):945–54. doi: 10.1016/j.pnpbp.2010.04.024

PubMed Abstract | CrossRef Full Text | Google Scholar

163. Sarafoff M, Davis L, Ruther E. Clozapine induced increase of human plasma norepinephrine. J Neural Transm (1979) 46(2):175–80. doi: 10.1007/BF01250337

PubMed Abstract | CrossRef Full Text | Google Scholar

164. Pickar D, Owen RR, Litman RE, Konicki E, Gutierrez R, Rapaport MH. Clinical and biologic response to clozapine in patients with schizophrenia. Crossover comparison with fluphenazine. Arch Gen Psychiatry (1992) 49(5):345–53. doi: 10.1001/archpsyc.1992.01820050009001

PubMed Abstract | CrossRef Full Text | Google Scholar

165. Breier A, Buchanan RW, Waltrip RW 2nd, Listwak S, Holmes C, Goldstein DS. The effect of clozapine on plasma norepinephrine: relationship to clinical efficacy. Neuropsychopharmacol (1994) 10(1):1–7. doi: 10.1038/npp.1994.1

CrossRef Full Text | Google Scholar

166. Elman I, Goldstein DS, Eisenhofer G, Folio J, Malhotra AK, Adler CM, et al. Mechanism of peripheral noradrenergic stimulation by clozapine. Neuropsychopharmacol (1999) 20(1):29–34. doi: 10.1016/S0893-133X(98)00047-5

CrossRef Full Text | Google Scholar

167. Brown AS, Gewirtz G, Harkavy-Friedman J, Cooper T, Brebion G, Amador XF, et al. Effects of clozapine on plasma catecholamines and relation to treatment response in schizophrenia: a within-subject comparison with haloperidol. Neuropsychopharmacol (1997) 17(5):317–25. doi: 10.1016/S0893-133X(97)00073-0

CrossRef Full Text | Google Scholar

168. Amiel JM, Mangurian CV, Ganguli R, Newcomer JW. Addressing cardiometabolic risk during treatment with antipsychotic medications. Curr Opin Psychiatry (2008) 21(6):613–8. doi: 10.1097/YCO.0b013e328314b74b

PubMed Abstract | CrossRef Full Text | Google Scholar

169. Wu C, Yuen J, Boyda HN, Procyshyn RM, Wang CK, Asiri YI, et al. An evaluation of the effects of the novel antipsychotic drug lurasidone on glucose tolerance and insulin resistance: a comparison with olanzapine. PLoS One (2014) 9(9):e107116. doi: 10.1371/journal.pone.0107116

PubMed Abstract | CrossRef Full Text | Google Scholar

170. Boyda HN, Procyshyn RM, Pang CC, Hawkes E, Wong D, Jin CH, et al. Metabolic side-effects of the novel second-generation antipsychotic drugs asenapine and iloperidone: a comparison with olanzapine. PLoS One (2013) 8(1):e53459. doi: 10.1371/journal.pone.0053459

PubMed Abstract | CrossRef Full Text | Google Scholar

171. Barr AM, Powell SB, Markou A, Geyer MA. Iloperidone reduces sensorimotor gating deficits in pharmacological models, but not a developmental model, of disrupted prepulse inhibition in rats. Neuropharmacology (2006) 51(3):457–65. doi: 10.1016/j.neuropharm.2006.04.004

PubMed Abstract | CrossRef Full Text | Google Scholar

172. Boyda HN, Ramos-Miguel A, Procyshyn RM, Topfer E, Lant N, Choy HH, et al. Routine exercise ameliorates the metabolic side-effects of treatment with the atypical antipsychotic drug olanzapine in rats. Int J Neuropsychopharmacol (2014) 17(1):77–90. doi: 10.1017/S1461145713000795

PubMed Abstract | CrossRef Full Text | Google Scholar

173. Boyda HN, Procyshyn RM, Tse L, Wong D, Pang CC, Honer WG, et al. Intermittent treatment with olanzapine causes sensitization of the metabolic side-effects in rats. Neuropharmacology (2012) 62(3):1391–400. doi: 10.1016/j.neuropharm.2011.02.019

PubMed Abstract | CrossRef Full Text | Google Scholar

174. Boyda HN, Procyshyn RM, Tse L, Hawkes E, Jin CH, Pang CC, et al. Differential effects of 3 classes of antidiabetic drugs on olanzapine-induced glucose dysregulation and insulin resistance in female rats. J Psychiatry Neurosci (2012) 37(6):407–15. doi: 10.1503/jpn.110140

PubMed Abstract | CrossRef Full Text | Google Scholar

175. Kowalchuk C, Teo C, Wilson V, Chintoh A, Lam L, Agarwal SM, et al. In male rats, the ability of central insulin to suppress glucose production is impaired by olanzapine, whereas glucose uptake is left intact. J Psychiatry Neurosci (2017) 42(6):424–31. doi: 10.1503/jpn.170092

PubMed Abstract | CrossRef Full Text | Google Scholar

176. Mann S, Chintoh A, Giacca A, Fletcher P, Nobrega J, Hahn M, et al. Chronic olanzapine administration in rats: Effect of route of administration on weight, food intake and body composition. Pharmacol Biochem Behav (2013) 103(4):717–22. doi: 10.1016/j.pbb.2012.12.002

PubMed Abstract | CrossRef Full Text | Google Scholar

177. Bush ND, Townsend LK, Wright DC. AICAR Prevents Acute Olanzapine-Induced Disturbances in Glucose Homeostasis. J Pharmacol Exp Ther (2018) 365(3):526–35. doi: 10.1124/jpet.118.248393

PubMed Abstract | CrossRef Full Text | Google Scholar

178. Weston-Green K, Huang XF, Deng C. Alterations to melanocortinergic, GABAergic and cannabinoid neurotransmission associated with olanzapine-induced weight gain. PLoS One (2012) 7(3):e33548. doi: 10.1371/journal.pone.0033548

PubMed Abstract | CrossRef Full Text | Google Scholar

179. Skrede S, Ferno J, Vazquez MJ, Fjaer S, Pavlin T, Lunder N, et al. Olanzapine, but not aripiprazole, weight-independently elevates serum triglycerides and activates lipogenic gene expression in female rats. Int J Neuropsychopharmacol (2012) 15(2):163–79. doi: 10.1017/S1461145711001271

PubMed Abstract | CrossRef Full Text | Google Scholar

180. Albaugh VL, Singareddy R, Mauger D, Lynch CJ. A double blind, placebo-controlled, randomized crossover study of the acute metabolic effects of olanzapine in healthy volunteers. PLoS One (2011) 6(8):e22662. doi: 10.1371/journal.pone.0022662

PubMed Abstract | CrossRef Full Text | Google Scholar

181. Martins PJ, Haas M, Obici S. Central nervous system delivery of the antipsychotic olanzapine induces hepatic insulin resistance. Diabetes (2011) 59(10):2418–25. doi: 10.2337/db10-0449

CrossRef Full Text | Google Scholar

182. Girault EM, Alkemade A, Foppen E, Ackermans MT, Fliers E, Kalsbeek A. Acute peripheral but not central administration of olanzapine induces hyperglycemia associated with hepatic and extra-hepatic insulin resistance. PLoS One (2012) 7(8):e43244. doi: 10.1371/journal.pone.0043244

PubMed Abstract | CrossRef Full Text | Google Scholar

183. Tulipano G, Rizzetti C, Bianchi I, Fanzani A, Spano P, Cocchi D. Clozapine-induced alteration of glucose homeostasis in the rat: the contribution of hypothalamic-pituitary-adrenal axis activation. Neuroendocrinology (2007) 85(2):61–70. doi: 10.1159/000100981

PubMed Abstract | CrossRef Full Text | Google Scholar

184. Babic I, Gorak A, Engel M, Sellers D, Else P, Osborne AL, et al. Liraglutide prevents metabolic side-effects and improves recognition and working memory during antipsychotic treatment in rats. J Psychopharmacol (2018) 32(5):578–90. doi: 10.1177/0269881118756061

PubMed Abstract | CrossRef Full Text | Google Scholar

185. Grajales D, Ferreira V, Valverde ÁM. Second-Generation Antipsychotics and Dysregulation of Glucose Metabolism: Beyond Weight Gain. Cells (2019) 8(11):1–27. doi: 10.3390/cells8111336

CrossRef Full Text | Google Scholar

186. Houseknecht KL, Robertson AS, Zavadoski W, Gibbs EM, Johnson DE, Rollema H. Acute effects of atypical antipsychotics on whole-body insulin resistance in rats: implications for adverse metabolic effects. Neuropsychopharmacology (2007) 32(2):289–97. doi: 10.1038/sj.npp.1301209

PubMed Abstract | CrossRef Full Text | Google Scholar

187. Kowalchuk C, Kanagasundaram P, McIntyre WB, Belsham DD, Hahn MK. Direct effects of antipsychotic drugs on insulin, energy sensing and inflammatory pathways in hypothalamic mouse neurons. Psychoneuroendocrinology (2019) 109:104400. doi: 10.1016/j.psyneuen.2019.104400

PubMed Abstract | CrossRef Full Text | Google Scholar

Keywords: antipsychotic, clozapine, cardiovascular disease, diabetes, metabolic syndrome, preclinical, side-effects

Citation: Yuen JWY, Kim DD, Procyshyn RM, Panenka WJ, Honer WG and Barr AM (2021) A Focused Review of the Metabolic Side-Effects of Clozapine. Front. Endocrinol. 12:609240. doi: 10.3389/fendo.2021.609240

Received: 22 September 2020; Accepted: 15 January 2021;
Published: 25 February 2021.

Edited by:

Katherine Samaras, St Vincent’s Hospital Sydney, Australia

Reviewed by:

Julia M. Lappin, University of New South Wales, Australia
Anthony Russell, Princess Alexandra Hospital, Australia
Dan Siskind, Metro South Addiction and Mental Health Services (MSAMHS), Australia

Copyright © 2021 Yuen, Kim, Procyshyn, Panenka, Honer and Barr. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Alasdair M. Barr, al.barr@ubc.ca

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.