You are currently viewing a new version of our website. To view the old version click .
Journal of Clinical Medicine
  • Review
  • Open Access

9 November 2025

Metabolic and Endocrine ADRs of Atypical Antipsychotics (AAPs) in Paediatric Patients with Autism Spectrum Disorder (ASD): A Review of Prevalence, Risk Factors, and Implications for Clinical Monitoring

,
,
and
1
School of Pharmacy, University of Birmingham, Birmingham B15 2TT, UK
2
Psychiatry Services, Brooklands Hospital, Coventry and Warwickshire Partnership NHS Trust, Birmingham B37 7HH, UK
*
Author to whom correspondence should be addressed.
J. Clin. Med.2025, 14(22), 7942;https://doi.org/10.3390/jcm14227942 
(registering DOI)
This article belongs to the Special Issue New Update on Personalized Treatment of Autism Spectrum Disorder and Related Disorders

Abstract

Atypical antipsychotics (AAPs) remain the most effective treatment to control irritability associated with autism spectrum disorder (ASD). Although there is no pharmaceutical treatment to target the core symptoms of ASD, AAPs reduce their severity. However, AAPs have been reported to be associated with severe adverse drug reactions (ADRs) that may lead to long-term conditions such as diabetes mellitus and heart disease. Their prevalence varies depending on the type of AAP prescribed, age, ethnicity, gender, healthcare systems, and the severity of the ASD. Current ADR monitoring guidelines exist, but they are broad in scope and do not fully account for these factors. Therefore, the need to develop ADR monitoring guidelines considering these factors has increased with the expanded use of AAPs in paediatrics with ASD. This gap in knowledge and clinical practice highlights the ongoing need for research to explore these factors and how they can inform the creation of tailored guidelines for monitoring ADRs in this population.

1. Introduction

Autism spectrum disorder (ASD) is a neurological condition characterised by challenges in social interaction and repetitive interests []. Recently, its prevalence has been reported to be 1% of the population in the UK, 2.5% in Saudi Arabia, and 2.8% in the USA [,,]. The UK and the USA are presented as well-studied populations, whereas Saudi Arabia as an underrepresented population []. To date, there is no pharmacological treatment targeting core symptoms (difficulties in social interaction and restricted interests) []. However, atypical antipsychotics (AAPs) are more effective at reducing core symptoms than other treatment options, such as antidepressants, stimulants, and mood stabilisers []. Additionally, AAPs have been shown to be most effective in managing irritability and comorbidities associated with ASD [,]. Furthermore, two AAPs, risperidone and aripiprazole, have been approved by the Food and Drug Administration (FDA) and the European Medicines Agency (EMA) for children aged 5 and older with ASD [,]. Risperidone and aripiprazole are therefore the first-line treatments for irritability in children and adolescents with ASD []. While other AAPs, including olanzapine, quetiapine, lurasidone, and ziprasidone, are still prescribed for off-label use. These differences in FDA approval status may explain why risperidone and aripiprazole are more commonly prescribed and prioritised in clinical guidelines compared to other AAPs []. However, administration of AAPs is associated with neurological, cardiac, metabolic, and endocrine adverse drug reactions (ADRs). Recent studies have focused on metabolic and endocrine ADRs than other ADRs for several reasons. Firstly, they may contribute to the development of long-term conditions, such as diabetes and heart disease []. Secondly, they are associated with the risk of osteoporosis and reproductive system issues (e.g., hypogonadism and menstrual irregularities), particularly in children and adolescents []. Thirdly, they are easily detected and managed in the early stages, whereas preventing or slowing the progression of their complications can be challenging []. These ADRs and their clinical complications are influenced by various risk factors and also impose a burden on healthcare systems []. Therefore, investigating the factors affecting ADR prevalence is essential for developing monitoring guidelines for paediatric patients with ASD.
The aim of this review is to explore the factors related to medications, patients, healthcare systems, and disease that influence the prevalence of ADRs, specifically metabolic and endocrine ADRs, and their clinical implications.

2. Adverse Drug Reactions (ADRs)

The World Health Organization (WHO) plays a crucial role in ensuring safety for those who are treated with medications and other therapeutic agents by developing clinical guidelines and initiatives. Health organisations worldwide have recently renewed their focus on pharmacovigilance due to the complexity of drug evaluation and the rapid evolution of medications and diagnostic tools [,]. WHO defines pharmacovigilance as “The science and activities related to the detection, assessment, understanding, and prevention of adverse drug effects or any other possible drug-related problems” [,].
The drug event terminology includes ADRs and medication errors (MEs) []. According to WHO, MEs is “any preventable event that may cause or lead to inappropriate medication use or patient harm while the medication is in the control of the healthcare professional, patient, or consumer” []. By contrast, WHO defines ADR as “Any response to a drug which is noxious and unintended which occurs at doses normally used in man for prophylaxis, diagnosis, or therapy of disease, or for the modifications of physiological function” [,]. In parallel, EMA and the Medicines and Healthcare Products Regulatory Agency (MHRA) in the UK, define ADR as “a response to a medicinal product which is noxious and unintended” []. From these definitions, the key difference lies in preventability. To illustrate this explicitly, MEs can be prevented through correct use, while ADRs may occur even when medication is used properly [].
National and international health organisations encourage health professionals and patients to report any ADRs through national spontaneous reporting systems such as the FDA, the MHRA and the Saudi Food and Drug Authority (SFDA), which in turn analyse the data and report to the Uppsala Monitoring Centre (UMC) []. In the UK, ADRs comprised 16% of all hospital admissions, with an average length of stay of approximately one week. The direct cost of ADR admissions for one month was around GBP 468,993 (USD 634,409) in the UK []. Similarly, ADRs account for 11.7% of all hospital admissions in the USA, with the average length of stay reaching 11 days. Conversely, in Saudi Arabia, ADRs account for 1.6% of hospital admissions with no prolongation of hospital stay [].
ADRs are an avoidable strain on the finite resources of healthcare systems. Thus, investigation of these ADRs is essential, particularly in medications potentially associated with severe ADRs or in vulnerable populations such as children or patients with mental health disorders []. Based on the classification of Rawlins and Thompson, ADRs can be categorised into six types: A, B, C, D, E, and F [,]. Type A (augmented reaction) is dose-dependent and results from the pharmacological activity of the drug, while Type B (bizarre reaction) is idiosyncratic and unrelated to the pharmacological activity of the drug. Type C (chronic reaction) is a time-dependent reaction related to the accumulation of the dose over time, whereas Type D (delayed reaction) appears long after drug use. Type E (end-of-use) occurs upon withdrawal of the drug, and Type F (failure of treatment) refers to unpredictable treatment failure [,].
Another scale for ADR classification is the Hartwig Scale (Table 1). It classifies ADRs into seven levels, ranging from mild (levels 1–2) and moderate (levels 3–4) to severe (levels 5–7) []. This scale is preferred over other scales for several reasons. Firstly, the Hartwig scale better guides clinical decision-making and patient management by prioritising ADR severity. Secondly, it is more applicable to pharmacovigilance and hospital settings []. Thirdly, it is directly relevant to patient safety because it highlights ADRs requiring intervention []. This review emphasises levels 3–6. However, level 7 is associated with clozapine administration, which is not used in ASD management [].
Table 1. The classification of atypical antipsychotics (AAPs) ADRs based on the Hartwig scale.

2.1. ADRs Associated with AAPs

2.1.1. Metabolic ADRs

Metabolic ADRs encompass metabolic abnormalities that include hyperglycaemia (fasting blood glucose ≥ 110 mg/dL) and hyperlipidaemia (triglycerides ≥ 100 mg/dL or total cholesterol ≥ 170 mg/dL), which contribute to type 2 diabetes and cardiovascular diseases []. Children and adolescents receiving AAPs seem to be more prone to metabolic ADRs compared to adults []. In particular, children with ASD are more likely to develop metabolic ADR than adults or children with other mental disorders [,]. For example, the prevalence of metabolic ADRs reaches up to 55% with AAPs in ASD patients [].
A recent umbrella review was conducted by Carnovale et al. (2024) [], evaluating 23 studies (six systematic reviews, 13 meta-analysis, and four networking meta-analysis) to compare the prevalence of metabolic ADRs among AAPs. It found that no AAP showed a statistically significant risk of hyperglycaemia compared to the placebo, as all 95% confidence intervals (CI) crossed zero, indicating a lack of statical significance However, among AAPs, olanzapine (median difference (medianD) = 4.51, 95% CI = −1.70, 10.72) and aripiprazole (medianD = 3.53, 95% CI = −1.66, 8.84) showed comparatively higher effect size on glucose levels, while ziprasidone (medianD = −5.93, 95% CI = −13.23, 1.37) and lurasidone (medianD = 1.67, 95% CI = −3.50, 6.88) showed comparatively smaller effect [,]. However, there was no statistically significant difference among these AAPs. Regarding hyperlipidaemia, olanzapine (standardised mean difference (SMD) = 0.40, 95% CI = −0.01, 0.80) showed the highest increase in triglyceride levels, although this was not statistically significant, followed by quetiapine (SMD = 0.37, 95% CI = 0.06, 0.68), which was statistically significant. In contrast, ziprasidone, followed by aripiprazole and risperidone, showed comparatively smaller effect size for hypertriglyceridemia (SMD = 0.09, 95% CI = −0.25, 0.43), (SMD = 0.06, 95% CI = −0.29, 0.42), (SMD = 0.12, 95% CI = −0.07, 0.32), respectively. However, none were statistically significant, as all 95% CIs crossed zero. With respect to weight gain, olanzapine (odds ratio (OR) = 17.34, 95% CI = 3.97, 75.65) posed the highest estimated risk among AAPs, followed by risperidone (OR = 9.01, 95% CI = 2.10, 38.69) and quetiapine (OR = 8.4, 95% CI = 1.58, 44.82), while ziprasidone (OR = 0.45, 95% CI = 0.04, 5.49), followed by aripiprazole (OR = 2.34, 95% CI = 0.47, 11.64), were associated with the lowest estimated risk [,]. However, the wide 95% CIs indicate considerable uncertainty that may be due to a small sample size. A study by DelBello et al. (2022) indicated that olanzapine may be associated with an increased risk of weight gain (OR = 44.81, 95% CI = (11.19, 147.70)), while lurasidone appeared to have a lower risk of weight gain (OR = 0.82, 95% CI = 0.22, 2.13) [,]. However, the wide 95% CIs indicate considerable uncertainty. The umbrella review demonstrated a wide range and lack of precision in 95% CI findings, which may be due to heterogeneity resulting from the inclusion of studies that use different effect sizes, such as SMD and OR, making direct comparisons and evidence synthesis more challenging. Additionally, the review does not focus specifically on ASD populations, which contributes to inconsistencies in results. Ultimately, the lack of consideration for key demographic factors such as age, gender, and ethnicity limits the generalisability and applicability of the findings.

2.1.2. Endocrine ADRs

Endocrine ADRs are characterised as unintended and harmful effects on the endocrine system that result in abnormalities in hormone secretion, such as hyperprolactinaemia (for <12 years: >25.4 μg/L; for ≥12 years: >18.4 μg/L for males and >24.1 μg/L for females) and thyroid dysfunction (for 2 to ≤17 years; thyroid-stimulating hormone (TSH) is ≥10 mIU/L and thyroxine (T4) is <0.80–1.8 ng/dL) [,].
Hyperprolactinaemia ADRs
Hyperprolactinaemia is studied more extensively compared to thyroid dysfunction. Although children and adolescents have a higher risk for hyperprolactinaemia, its contributions are primarily examined in adults [,]. Hyperprolactinaemia may cause serious complications such as galactorrhoea, menstrual irregularities, infertility, and changes in bone density []. The prevalence of hyperprolactinaemia ADR with AAP use was 38.5% in children and adolescents with neurological disorders, including those with ASD [].
AAPs were linked to significantly increased prolactin levels in children and adolescents compared to placebo (mean difference (MD) = 10.10 μg/L, 95%CI  =  6.97,13.23) []. AAPs differ in their risk of inducing hyperprolactinaemia. For example, the prolactin level increases by 28.24 μg/L (p < 0.05) and 11.34 μg/L (p < 0.01) in risperidone and olanzapine, respectively []. By contrast, aripiprazole significantly decreased prolactin to 4.91 μg/L (p < 0.01). Other AAPs were not associated with significant elevation, included quetiapine (p < 0.06) and lurasidone (p = 0.22) []. Although ziprasidone is associated with hyperprolactinaemia, the limitations of the studies make it challenging to provide robust conclusions []. The main limitations include a small sample for ziprasidone (16 patients), a predominantly male adolescent population (up to 66%), and although the risk is low and unclear, bias could remain, which may limit generalizability.
Thyroid Dysfunction ADRs
Quetiapine is the most extensively studied AAP and has been confirmed to cause significant hypothyroidism (p < 0.01) with low prevalence [,]. Other AAPs lack a significant impact on thyroid function, such as risperidone (p > 0.05) and olanzapine (p > 0.05) []. Due to a lack of controlled evidence supporting the positive correlation between AAP and hypothyroidism, several studies did not demonstrate the need for monitoring serum thyroid hormones in patients with normal thyroid levels hormones [,,]. However, the Canadian Alliance for Monitoring Effectiveness and Safety of Antipsychotics (CAMESA) recommended monitoring thyroid hormones for children and adolescents receiving quetiapine [,]. For example, patients with normal TSH levels should undergo repeated testing, while those with abnormal TSH levels require ongoing evaluation monitoring []. Additionally, the Maudsley prescribing guidelines recommend annual monitoring of thyroid function in patients taking quetiapine, even though the incidence of thyroid dysfunction is low []. Contradictory results have been reported regarding the administration of quetiapine and thyroid dysfunction []. For instance, studies by Alvarez-Herrera et al. (2020) [] and Khoodoruth, Abdo, and Ouanes, (2022) [] reported elevated TSH levels and a decline in T4 with quetiapine. Meanwhile, the study by Findling et al. (2014) [] observed no change in T4 and elevation in TSH with quetiapine compared to placebo (p = 0.25).

3. Risk Factors Influencing the Prevalence of Metabolic and Endocrine ADRs

3.1. Medication-Related Factors

3.1.1. Mechanism of Metabolic ADRs

AAPs induce metabolic ADRs by affecting neurotransmitter signalling (H1, D2, 5-HT2A) or by impairing the function of molecules that regulate appetite, glucose, and lipid metabolism in the central nervous system (CNS) and peripheral organs, including the liver, pancreas, and adipose tissues (Figure 1) [,].
Figure 1. Mechanistic overview of atypical antipsychotics (AAPs)–induced metabolic dysregulation across multiple organ systems. Abbreviation: D2: Dopamine; 5-HT2A: 5-Hydroxytryptamine (Serotonin) 2A subtype; 5-HT2C: 5-Hydroxytryptamine (Serotonin) 2C subtype, H1: Histamine, α2: Alpha-2 adrenergic; M1: Muscarinic acetylcholine (M1 subtype); POMC: Pro-opiomelanocortin; α-MSH: Alpha-melanocyte-stimulating hormone; NPY: Neuropeptide Y; AgRP: Agouti-related peptide; AMPK: AMP-activated protein kinase; GLUT4: Glucose transporter 4, PI3K: Phosphoinositide 3-kinase; IRS: Insulin receptor substrate; ERK: Extracellular signal-regulated kinase; FFA: Free fatty acids; SLD: steatotic liver disease; GCGR: Glucagon receptor; PEPCK: Phosphoenolpyruvate carboxykinase; G6Pase: Glucose-6-phosphatase; SREBP1/2: Sterol regulatory element-binding protein ½; mTOR: Mammalian target of rapamycin; PPARγ: Peroxisome proliferator-activated receptor γ; IL-2: Interleukin-2; IL-6: Interleukin-6; TNF: Tumour necrosis factor; ↑: increase; ↓: decrease. Created in BioRender. Aljead, M. (2025) https://app.biorender.com/illustrations/6807a71b3c36b68f8a93e223.
Weight Gain and Hyperglycaemia
Dopamine receptor D2:
The blockage of D2 receptors may indirectly induce metabolic ADRs. For instance, D2 inhibits orexin, a neuropeptide that regulates appetite and energy homeostasis []. When AAPs block the D2 receptor, it causes stimulation of orexin expression, resulting in increased hyperphagia and reduced physical activity []. Additionally, the prolactin hormone is regulated by D2 in the hypothalamus, which affects biological pathways in pancreatic β-cells and adipose tissues []. Thus, blocking the D2 receptor leads to elevated prolactin levels, decreased insulin sensitivity, and increased food consumption by dysregulating glucose metabolism []. Taken together, these mechanisms contribute to weight gain and hyperglycaemia [,].
5-HT2C/2A receptors
In the hypothalamus, 5-HT2C activates Pro-opiomelanocortin (POMC) neurons to express α-melanocyte stimulating hormone (α-MSH), which is a key peptide in the regulation of appetite as well as glucose and lipid metabolism. AAPs block 5-HT2C, causing inhibition of the POMC/α-MSH (anorexigenic) pathway, leading to hyperphagia and decreased fatty acid oxidation and glucose uptake [,]. In contrast, AAPs stimulate the neuropeptide Y (NPY)/Agouti-related protein (AgRP) (orexigenic) pathway to induce hyperphagia and weight gain []. Additionally, AAPs block 5-HT2C in pancreatic β-cells, resulting in a decrease in insulin secretion and hyperglycaemia []. Moreover, the blockade of 5-HT2C activity disrupts leptin regulation and increases food intake, which leads to weight gain [,].
In contrast, AAPs block 5-HT2A in skeletal muscles and indirectly impair glucose transporter type 4 (GLUT4), leading to a reduction in glucose uptake by cells [,]. This mechanism causes an increase in insulin resistance and hyperglycaemia [].
H1 receptors
One proposed mechanism is that AAPs induce metabolic ADRs through H1 antagonism, which indirectly interferes with the AMP-activated protein kinase (AMPK) pathway, which regulates appetite in the hypothalamus. AAPs block H1 receptors in the hypothalamus, leading to the activation of AMPK activity, which increases hyperphagia and weight gain [,]. In skeletal muscles, AAPs block H1 receptors, inducing insulin resistance and impairing insulin signalling by inhibiting phosphoinositide 3-kinase (PI3K), insulin receptor substrate (IRS) proteins, and extracellular signal-regulated kinase (ERK) cascades [,]. Furthermore, the activation of the H1 receptors in pancreatic β-cells increases insulin secretion []. Hence, interfering with insulin signalling or secretion by AAPs contributes to hyperglycaemia and elevation of risk of type 2 diabetes [].
α2-adrenergic and M1 acetylcholine receptors
AAPs modulate α2-adrenergic receptors activity, subsequently altering sympathetic activity. This contributes to increased glucose production and lipolysis, resulting in short-term hyperglycaemia [,]. They also act as M1 antagonists in the hypothalamus and impair acetylcholine activity in glucose metabolism by inhibiting insulin signalling [,].
Hyperlipidaemia
All the underlying pharmacological mechanisms of AAPs induce hyperlipidaemia through an indirect effect via the inhibition of neurotransmitter receptors, leading to disruption of glucose and lipid metabolism and insulin resistance [,]. However, AAPs also directly affect biological molecules that regulate lipid metabolism []. For example, AAPs directly inhibit the AMPK pathway in the liver or do so indirectly through adiponectin signalling in adipose tissue []. This leads to the stimulation of lipogenesis by reducing fatty acid oxidation, resulting in the accumulation of lipids in hepatic tissues and weight gain [,]. In contrast, AAPs activate the glucagon receptor (GCGR), which stimulates gluconeogenesis by increasing the expression of phosphoenolpyruvate carboxykinase (PEPCK) and G6Pase (glucose-6-phosphat(ase)) while reducing glucose uptake. The activation of GCGR contributes to hyperglycaemia and insulin resistance []. Moreover, in adipose tissue, AAPs stimulate the mammalian target of rapamycin (mTOR), which activates lipid metabolism by enhancing the expression of peroxisome proliferator-activated receptor-γ (PPARγ), leading to increased lipid storage []. Additionally, AAPs upregulate sterol regulatory element-binding protein 1/2 (SREBP1/2) in the liver, resulting in increased lipogenesis []. AAPs also enhance the expression of IL-2, IL-6, and TNF-α in adipose tissue that activates lipolysis and free fatty acids and causes hyperlipidaemia [,].
Olanzapine carries the highest risk of metabolic ADRs, followed by risperidone and quetiapine, and then aripiprazole, ziprasidone, and lurasidone [,]. Olanzapine carries the highest risk of metabolic ADRs compared to other AAPs, possibly due to its greatest affinity for H1 and M1 receptors. In contrast, lurasidone presents the lowest risk of metabolic ADRs because it lacks activity on the H1 receptor. Consequently, other biological pathways regulated by H1, which impact body weight and glucose and lipid metabolism, such as AMPK, PI3K, IRS proteins, and ERK, are not dysregulated by lurasidone [].

3.1.2. Mechanism of Endocrine ADRs

Hyperprolactinaemia
Hyperprolactinaemia induces menstrual disorders and sexual dysfunction and inhibits bone formation in paediatric patients []. This occurs through blockade of D2 receptors, leading to elevated prolactin levels. Elevated prolactin levels directly inhibit gonadotropin-releasing hormone (GnRH), which in turn decreases the secretion of follicle-stimulating hormone (FSH) and luteinizing hormone (LH) (Figure 2) []. Both FSH and LH play crucial roles in menstrual regulation and fertility. Thus, a decline in these hormones contributes to infertility and sexual dysfunction []. Additionally, reduced levels of sex hormones (estradiol and testosterone) interfere with bone mineralization and decrease bone density, particularly in adolescents during peak bone development []. The risk of hyperprolactinaemia tends to be increased with risperidone due to its highest affinity for D2 (Ki = 2.29 nM) [].
Figure 2. Potential mechanism of AAPs inducing hyperprolactinaemia and their effects on sex organs and bone functions. Abbreviations: D2R: Dopamine D2 Receptor; AAPs: Atypical Antipsychotics; PRH: Prolactin-Releasing Hormone; GnRH: Gonadotropin-Releasing Hormone; FSH: Follicle-Stimulating Hormone; LH: Luteinizing Hormone; PRL: Prolactin; E2: Estradiol, T: Testosterone, BMD: Bone Mineral Density. : inhibition; : stimulation; ↑↑: increase; ↓: decrease; Created in BioRender. Aljead, M. (2025) https://app.biorender.com/illustrations/6808f6c4346bb57af605562f.
Thyroid Dysfunction
The underlying mechanism of quetiapine induced thyroid dysfunction remains unclear. However, multiple hypothesised mechanisms have been proposed (Figure 3) []. The blockade of D2 receptors by AAPs may reduce the release of thyrotropin-releasing hormone (TRH) from the hypothalamus, resulting in decreased TSH secretion from the anterior pituitary gland. This leads to decreased triiodothyronine (T3) and T4, causing hypothyroidism [,,]. A major criticism of this hypothesised mechanism, linking D2 receptor blockade to reduced TRH and subsequent hypothyroidism, is that quetiapine has a low affinity for D2 receptors (Ki = 567 nM) compared to other AAPs [,]. If this mechanism were the primary driver of thyroid dysfunction, one would expect higher frequency for this ADR with risperidone, which has the highest D2 receptor affinity (Ki = 2 nM) among AAPs [,].
Figure 3. Hypothesised mechanisms related to D2 and H1 Blockers, including thyroid dysfunction caused by Quetiapine. Abbreviation: QTP: Quetiapine, D2R: Dopamine D2 Receptor, H1R: Histamine H1 Receptor, TRH: Thyrotropin-Releasing Hormone, TSH: Thyroid-Stimulating Hormone, T3: Triiodothyronine, T4: Thyroxine; ⊥: inhibition; ↓↓: decrease Created in BioRender. Aljead, M. (2025) https://app.biorender.com/illustrations/680a5309a561a9a36a47aa85.
Quetiapine has a high affinity for the H1 receptor, which could disrupt the hypothalamic-pituitary-thyroid (HPT) axis through alternative neuroendocrine pathways, such as the leptin pathway []. However, this proposed mechanism has limitations, olanzapine exhibits higher H1 affinity (2.3 nM) than quetiapine (8.7 nM), which would suggest a higher risk of hypothyroidism. Yet, clinical outcomes differ that may be due to pharmacokinetic factors (e.g., higher cmax for quetiapine [1291.4 nM] and differences in tissue distribution compared to olanzapine) and downstream signalling effects rather than receptor affinity alone. Another mechanism associated with quetiapine that induces thyroid dysfunction is the competition between thyroid hormone and quetiapine, and quetiapine being metabolised by uridine diphosphate glucuronosyltransferase (UGT), leading to reduced thyroid hormone levels []. This mechanism aligns more closely with olanzapine, for which UGT is one of the primary enzymes involved in metabolism, whereas it plays a secondary role in quetiapine. One further hypothesis considered is that quetiapine stimulates autoimmune thyroiditis, although this has been reported in an inconsistent manner in adults [,].

3.1.3. Pharmacokinetic Factors

Risperidone and olanzapine pharmacokinetics (PK) have been the most extensively researched AAP’s in children and adolescents (Table 2) [].
Table 2. Summary of pharmacokinetics of AAPs in paediatric populations. Abbreviation: cmax: maximum plasma concentration, F: female, ↑: increase, ↓: decrease, X: no effect, Vd; volume distribution, PPB: plasma protein binding, t1/2: half-life, UGT: uridine diphosphate glucuronosyltransferase, a: not effected by low-fat meal, b: no active metabolites, PM: Poor metabolizer, EM: extensive metabolizers.
Risperidone
A single oral dose (2 mg) of risperidone is rapidly absorbed and exhibits 70% bioavailability (%F), regardless of food intake. Risperidone reaches maximum plasma concentration (cmax = 36.5 nM) within one hour and three hours for the active metabolite (9-OH-risperidone) [,,]. It is highly protein bound (90%), with a volume distribution (Vd) ranging between 1 and 2 L/kg. It exhibits linear pharmacokinetics within its therapeutic dose []. Risperidone is primarily metabolised by CYP2D6, converting it to its active metabolite (9-OH-risperidone). The half-life (t1/2) for risperidone and 9-OH-risperidone is 20 h. The kidneys primarily excrete risperidone, and its clearance significantly declines in patients with renal impairment, causing drug accumulation and an increased risk of ADRs [].
Generally, in children aged 4 to 15 years receiving risperidone, the pharmacokinetics are similar to those observed in adults []. However, the concentration-to-dosage (C/D) ratio of risperidone and 9-OH-risperidone in children is lower than in adolescents and adults []. Compared to adults, protein binding is 16% lower in children and adolescents. As a result, the risk of ADRs may be higher in the paediatric population due to an increased free fraction (26%) of risperidone [].
The pharmacokinetics of risperidone are influenced by several factors, including concomitant medications and CYP2D6 phenotype. The co-administration of risperidone with CYP2D6 inhibitors (e.g., fluoxetine, valproate) reduces the metabolism of risperidone and increases the level of exposure by up to 55%, resulting in an increased risk of ADRs []. Moreover, the metabolism of risperidone is influenced by the CYP2D6 phenotype, which may lead to variability in the prevalence of ADRs due to differences in drug metabolism activity across populations and ethnicities [,,]. In the study by Biswas, Vanwong, and Sukasem (2022) [], weight gain and hyperglycaemia ADRs are reported among poor metabolizers (PMs). Therefore, dose adjustment of risperidone should be considered for patients who are PMs or those who concomitantly take CYP2D6 inhibitors [].
Aripiprazole
Aripiprazole is well absorbed with or without food, showing an 87% bioavailability []. It exhibits linear pharmacokinetics at therapeutic doses, reaching cmax, which is approximately 240.8 nM, within 3–5 h after 2–15 mg of oral aripiprazole [,]. It has a Vd of around 5 L/kg with 99% plasma protein binding, predominantly to albumin []. It is mainly metabolised by CYP2D6, producing the active metabolite (dehydroaripiprazole) and inactive metabolite (aripiprazole metabolites-1451) [,]. The aripiprazole t1/2 is 75 h and becomes longer by 20% in dehydroaripiprazole. Aripiprazole is excreted mainly via faeces (55%) and urine (30%) [].
Aripiprazole pharmacokinetics are comparable between children aged 10–17 years old and adults [,]. Nonetheless, factors such as gender and enzyme phenotype play a crucial role in the variability of aripiprazole pharmacokinetics [,]. Regarding gender differences, at an equivalent therapeutic dose of aripiprazole, the cmax was 11% higher in females than in males (p < 0.05) [].
There is no significant correlation between the concentration of aripiprazole in plasma and the co-administration of CYP2D6 inhibitors. However, this concentration is influenced by the CYP2D6 genotype [,]. For example, the t1/2 is doubled in PM, reaching 150 h compared to extensive metabolizers (EMs) [,].
Olanzapine
Olanzapine is well absorbed after oral administration, and its absorption is not affected by food. It exhibits linear pharmacokinetics, with a bioavailability of ≥65% [,,]. Its cmax is 48 nM after oral administration []. It binds approximately 93% of albumin and α1−acid glycoprotein and has a Vd of 21 L/kg []. Regarding its metabolism, CYP2D6, CYP1A2, and UGT1 convert olanzapine into three inactive metabolites, including 2-hydroxymethylolanzapine, N-desmethylolanzapine, and 10′/4′-N-glucuronides, respectively [,,]. The t1/2 of olanzapine ranges from 30 to 52 h, with an average clearance rate of 25 L/h. Up to 57% of olanzapine is renally excreted, while the remaining amount is excreted through faeces [,].
Factors such as age, gender, and concomitant medications that may affect the pharmacokinetics of olanzapine have shown conflicting results. Studies indicated that the plasma concentrations of olanzapine differ between adolescents and adults [,,,]. In contrast, several studies reported that a negative association was identified between age and plasma concentration of olanzapine [,]. These divergent outcomes may result from overlooking the factors influencing the pharmacokinetic profile. For instance, the t1/2 and cmax in adults are comparable to those observed in children aged 10–18 years old []. By contrast, clearance is reduced by 48% in younger children (those under 12 years old), leading to a 43% increase in olanzapine exposure compared to adults [,]. Additionally, females tend to have lower CYP1A2 activity than males, contributing to a 30% reduction in clearance. Consequently, both children and females may require a lower dose of olanzapine than adult males [,]. On the other hand, the co-administration of CYP1A2 inducers, such as cigarette smoking, increases olanzapine clearance by 40% in patients who smoke tobacco or e-cigarette (vaping) [,].
Quetiapine
Oral quetiapine is rapidly absorbed and is not affected by a low-fat meal, showing approximately 10% bioavailability and demonstrating linear pharmacokinetics [,]. Following 300 mg of quetiapine, the cmax was observed to be 1291.4 nM []. Its Vd varies from 6 to 14 L/kg, and albumin binding reaches 83% [,]. It is extensively metabolised by CYP3A4, which converts it to the active metabolite. N-desalkyl quetiapine []. The t1/2 of quetiapine is approximately 6 h, with an elevation of 12 h for N-desalkyl quetiapine []. Quetiapine is excreted through urine and faeces, accounting for 73% and 20%, respectively [].
The influence of age on quetiapine pharmacokinetics has been reported in several studies [,,]. For example, the plasma concentration of quetiapine is 41% higher in adults compared to children aged 10–17 years old []. In contrast, in the study by McConville et al. (2000) [], the t1/2 and cmax in adults are similar to those observed in children aged 12–17 years. Specifically, this discrepancy between these findings may be due to variations in study design, sample size, and age range of participants. The strength of this study is due to the systematic, population-based pharmacokinetics data across a wide paediatric age range. Meanwhile, the open-label trial conducted by McConville et al. (2000) [] had smaller sample sizes that followed DSM-IV criteria and provided limited pharmacokinetic detail, making them less suitable for modelling or dose optimisation.
The pharmacokinetics of quetiapine are influenced by concurrent medications, CYP3A4-dependent factors, and hepatic impairment [,,]. The administration of 400 mg/day of quetiapine with CYP3A4 inhibitors (e.g., erythromycin) increases its t1/2 by 92%. Therefore, a six-fold dose reduction is essential for the co-administration of quetiapine with CYP3A4 inhibitors [,,]. The quetiapine dose is reduced by 10% to 50% due to decreased clearance in patients with hepatic impairment []. There is a lack of evidence to support a gender difference with quetiapine [].
Lurasidone
The absorption of lurasidone increases by 2 to 3 times when taken with food []. Its cmax is 60.9 nM after a 20 mg dose with bioavailability varying between 9 and 19% due to first-pass metabolism [,,]. Lurasidone binds approximately 99% to plasma proteins, with Vd varying between 2.4 and 20 L/kg []. It is primarily metabolised by CYP3A4, producing two active metabolites, namely exo-hydroxy metabolite ID-14283 and endo-hydroxy metabolite ID-14326 []. The average t1/2 of lurasidone is 29 h as the parent drug and becomes 2.5-fold shorter in ID-14283. Approximately 80% of lurasidone is excreted in faeces, while 9% is excreted in urine [].
Numerous studies show that pharmacokinetics (e.g., plasma concentration) negatively correlates with age [,,]. However, both gender and concurrent medication influence its pharmacokinetics. In the study by Yang et al. (2022b) [], it was reported that the area under the curve (AUC) of lurasidone is approximately 15% higher in females than in males. In relation to concurrent medications, prescribing lurasidone (40 mg/day) with CYP3A4 inhibitors (e.g., ketoconazole 400 mg/day) increases AUC by approximately ten times and Cmax by seven times compared to lurasidone alone [,]. Consequently, it is advisable to avoid co-administration of ketoconazole with lurasidone and switch to an alternative antifungal [].
Ziprasidone
Ziprasidone is well absorbed, and its cmax reaches 121 nM within 6 h after a 20 mg dose [,]. The bioavailability of ziprasidone is approximately 60% and increases to 100% when consumed with food []. It is 99% bound to plasma albumin and α1 acid glycoprotein. It has a Vd of ~1.5 L/kg. The majority of ziprasidone’s metabolism is mediated by glutathione and aldehyde oxidase, while CYP3A4 plays a lesser role [,]. Ziprasidone’s t1/2 is approximately 7 h, with the main route of elimination being faecal (66%), while the remainder is excreted in the urine [].
The pharmacokinetics of ziprasidone in children are comparable to those in adults [,]. Several factors, including gender and polypharmacy, play a critical role in ziprasidone pharmacokinetics. The cmax of ziprasidone is approximately 25% higher in females than in males, which is statistically significant (p  <  0.05) []. Despite CYP3A4 having a minor effect on ziprasidone metabolism, the co-administration of 400 mg/day of ketoconazole significantly increases the AUC and cmax by approximately 34% for a 40 mg/day dose of ziprasidone (p < 0.05) [,].

3.2. Patient-Specific Factors

3.2.1. Age

Children and adolescents, particularly those with ASD who are administered AAPs, have a greater prevalence of metabolic and endocrine ADRs compared to adults [,]. There are multiple reasons that may explain why paediatric populations are more likely to develop these ADRs. As noted, their physiological systems for metabolising and eliminating medications are not as fully developed as those in adults [,].
In children, gastric pH is initially higher and gradually decreases to reach adult levels in the second year of childhood due to an immature digestive system. This can affect the absorption of AAPs []. For distribution, body composition changes by decreasing extracellular water and increasing fat storage throughout childhood. This causes a continuous change in Vd of drugs and t1/2, necessitating adjustments in dosage for AAPs [,]. In addition, paediatric populations have lower plasma protein levels compared to adults. For example, AAPs are highly protein-bound, leading to increased free AAP concentrations and a higher risk of ADRs in children [].
In addition, historically, children have been insufficiently included in clinical trials, which results in limited paediatric studies and creates challenges in determining safe and effective doses for children [,]. Furthermore, during puberty, these differences become more pronounced as hormonal fluctuations influence the concentration and clearance of the drug [,].

3.2.2. Gender

With respect to gender variation, female adolescents who are receiving risperidone or aripiprazole are more prone to metabolic ADRs, particularly weight gain, than males []. Furthermore, a statistical difference in the prevalence of weight gain (i.e., the increase in body mass index (BMI) was 1.3–1.5 kg/m2) was observed between males and females receiving AAPs (i.e., aripiprazole, olanzapine) (p < 0.001) [,]
Generally, the prevalence of endocrine ADRs, particularly hyperprolactinaemia, is 20% higher in females than in males who are receiving AAPs (i.e., risperidone, olanzapine, quetiapine, ziprasidone, and lurasidone), which may be due to the estrogenic effect on prolactin levels []. Another study showed that females receiving AAPs (i.e., aripiprazole, olanzapine, and quetiapine) experienced a higher rate of thyroid dysfunction ADRs compared to males, although the difference was not statistically significant (p  = 0.134) [].
There are several hypotheses for the variation in the prevalence of ADRs between males and females []. In the past, females were not included in clinical trials due to concerns about the teratogenic risk to those who could become pregnant []. Additionally, females typically have lower body mass and a higher proportion of adipose tissue, which influences the absorption and distribution of medications []. Furthermore, females exhibit a lower glomerular filtration rate (GFR) and P-glycoprotein (P-gp) renal efflux of medication compared to males, resulting in a prolonged t1/2 [,]. Moreover, females report ADRs more often than males []. Therefore, gender differences should be considered in guidelines to reduce the risk of ADRs.

3.2.3. Ethnicity

Ethnicity also plays a crucial role in determining the prevalence of metabolic and endocrine ADRs among AAPs. The Caucasian group is overrepresented compared to other ethnicities, such as Middle Eastern populations, which limits the generalizability of these study findings [].
This lack of diversity is particularly significant when considering pharmacogenetic factors that influence drug metabolism. AAPs are primarily metabolised by three key enzymes: CYP2D6, CYP3A4, and CYP1A2 []. The activity of these enzymes varies across ethnic groups due to differences in the frequency of genetic variants, which can, in turn, affect the efficacy and safety profiles of AAPs [].
Regarding CYP2D6, the metabolites OH-risperidone and dehydroaripiprazole decrease approximately two times in PMs and one and a half times in intermediate metabolizers [,]. A cohort study showed that CYP2D6 PMs of risperidone are most prevalent in Oceania (21%). In contrast, in the Middle East, CYP2D6 PMs rate is around 12.7%, followed by Europe (9.3%) and America (8.4%). While CYP2D6 PMs of aripiprazole are most prevalent among Europeans at 6.1%, the rate in Americans is 3.7%, and in Middle Easterns, it is 1.2% []. This indicates that the risk of risperidone ADRs is most frequently observed in Oceania, followed by the Middle East, Europe, and America. In contrast, the risk of ADRs associated with aripiprazole appears to be higher in Europe than in the Middle East populations. According to the Dutch Pharmacogenetics Working Group (DPWG) guideline, in CYP2D6 PMs, the dose of risperidone and aripiprazole should be prescribed at 50–67% of the therapeutic dose to reduce the risk of ADRs. In contrast, in ultrarapid metabolizers, risperidone should be switched to other medications to prevent treatment failure []. Interestingly, the rate of CYP2D6 PMs in the East Asian population is between 0.9% and 1.2% for aripiprazole and risperidone, respectively. This may explain why the therapeutic dose of risperidone in East Asians is reduced by 26% compared to Europeans (around 50%) [,,].
With reference to CYP3A4 PM, quetiapine plasma concentrations were observed to be more than three times higher. Therefore, they need a dose reduction to less than a third or a switch to an alternative medication with lower CYP3A4 dependence based on clinical use [,]. N-Desalkyl quetiapine exhibits antidepressant effects and is present in reduced concentrations in PM. Therefore, switching to an alternative drug is highly recommended for treating depression in PMs [].
In terms of CYP1A2, there is no significant correlation between olanzapine exposure and its phenotype. Numerous clinical studies did not observe any difference in the prevalence of ADRs across different populations [,,]. Therefore, the DPWG recommended that no further action be taken [].

3.3. Healthcare System Factors

The differences in strategies and policies for reporting and managing ADRs are among the key factors that contribute to variations in ADR prevalence between countries []. For instance, the MHRA partnered with patient organisations to develop pharmacovigilance guidelines and enhance patient awareness []. Similarly, the SFDA collaborated with the WHO to enhance ADR reporting among patients []. In contrast, in some countries, the absence of guidance for patients on reporting ADRs has led to a decline in the number of reported cases []. Moreover, some countries, such as the UK, standardise ADR reporting for both medications and herbal medicines, while Taiwan uses different forms that may delay responses from healthcare professionals (HCPs) []. Furthermore, the use of information technology in ADR reporting has increased to improve the validity and reliability of ADR data and to speed up response times for curating, managing, and analysing ADRs []. However, this facility remains limited in low-income countries, resulting in a misrepresentation of ADR prevalence globally [].

3.4. Disease-Related Factors

As described, ASD is a heterogeneous disorder that produces varying severity of symptoms and sensitivity to medications, resulting in increased susceptibility to ADRs [,]. In addition, the increased risk of ADRs in ASD patients may occur due to several reasons. Children and adolescents with ASD are typically prescribed more than one medication to manage ASD along with comorbidities, which increases the risk of drug–drug interactions (DDIs) and the likelihood of ADRs []. For example, in Ritter, Hewitt and McMorris (2021) [] study indicated that the rate of concurrent medications in this population reached up to 87%. Another reason is due to gut microbiota imbalance in ASD patients, which affects the absorption of medications and alters their pharmacokinetics []. Additionally, most ASD patients follow restricted diets (e.g., gluten-free), which contributes to a deficiency in co-factors (e.g., zinc, selenium) that negatively affects the metabolism of medications and alters their safety profiles []. Notably, ASD is linked to thyroid and adrenal dysfunction, which may potentiate endocrine-related ADR []. Similarly, a relationship exists between ASD and developed metabolic ADRs []. HCPs and caregivers also face difficulties differentiating between ADRs and ASD symptoms, such as weight gain due to physical inactivity [,,]. However, there is currently no standardised framework for assessing and controlling this factor, which may contribute to variability and inconsistent ADR reporting across studies. This lack of standardisation highlights the need for an ASD-specific approach to evaluate ADR susceptibility rather than relying solely on data from the general population of mental disorders and overlooking the potential impact of ASD on ADR prevalence [].

4. Research Gap and Future Directions

With the increasing global prevalence of heterogeneous diseases such as ASD and the use of AAPs as first-line treatments, the need for guidelines that consider population variation has become essential []. Until now, the DPWG, Clinical Pharmacogenetics Implementation Consortium (CPIC), and CAMESA have been the only organisations developing monitoring guidelines that considered individual variation within the population [,,].
Over the last five years, only four studies Al-Huseini et al. (2022), Alsabhan et al. (2024), Shilbayeh and Adeen (2023), and Makary et al. (2023)—[,,,]—from the Middle Eastern regions have examined the prevalence of metabolic or endocrine ADRs associated with AAPs. Furthermore, thyroid dysfunction remains unexplored in the Middle East and is rarely investigated in other regions []. However, current evidence remains limited, particularly regarding population-specific ADR prevalence in paediatric patients with ASD, especially within the Middle Eastern population. This underscores the urgent need for further research and the development of tailored monitoring guidelines [].

5. Clinical Implications

The study of the prevalence of metabolic and endocrine ADRs associated with AAPs in paediatric populations with ASD across different countries improves medication safety. Risk assessment helps identify high-risk populations and highlights variability in ADRs due to ethnicity, age, gender, disease, and healthcare-related factors [,]. As a result, this study encourages personalised treatment approaches and supports the development of tailored monitoring guidelines based on regional and individual risk profiles, including age and specific disease []. Additionally, it helps HCPs select safer AAPs based on the prevalence of ADRs in susceptible individuals based on their unique characteristics, thereby improving prescribing practices []. This study also contributes to the early detection and optimised monitoring of ADRs to prevent long-term complications, such as type 2 diabetes and cardiovascular diseases []. Moreover, it underscores the impact of healthcare infrastructure (e.g., spontaneous reporting systems) on the reporting and management of ADRs [].

6. Conclusions

AAPs have remained the first line of treatment for paediatric patients’ irritability with ASD due to their efficacy. However, AAPs use in ASD is associated with metabolic and endocrine ADRs that contribute to public health concerns. Although data on the prevalence of these ADRs inform current monitoring guidelines, such guidelines often rely on findings from specific populations (e.g., UK, USA) without accounting for inter-individual diversity factors such as ethnicity and age.
In the future, tailored monitoring guidelines for paediatric patients with heterogeneous disorders like ASD, particularly in underrepresented populations such as those in the Middle East, should be considered in clinical practice and research. This would support developing more personalised therapies, aiming to avoid both over-monitoring of rare or non-serious ADRs and under-monitoring serious ADRs not currently included in guidelines. Taken together this would promote the safer use of AAPs in paediatric populations with ASD.

Author Contributions

Conceptualization, M.A., A.M.J. and Z.J.; methodology, M.A.; validation, A.M.J. and Z.J.; investigation, M.A.; resources, M.A.; writing—original draft preparation, M.A.; writing—review and editing, A.M.J., Z.J. and A.Q.; visualisation, M.A.; supervision, A.M.J. and Z.J.; project administration, M.A. All authors have read and agreed to the published version of the manuscript.

Funding

This research was supported by the University of Birmingham Open Access Fund and the Saudi Cultural Bureau (SCB), as this work is part of a PhD scholarship funded by SCB.

Institutional Review Board Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

The authors thank the researchers prior published studies that have aided the development of this review.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

The following abbreviations are used in this manuscript:
AAPsAtypical antipsychotics
ADRsAdverse drug reactions
AgRPAgouti-related protein
AMPKAMP-activated protein kinase
ASDAutism spectrum disorder
AUCArea under the curve
BMIBody mass index
CAMESACanadian Alliance for Monitoring Effectiveness and Safety of Antipsychotics
CmaxMaximum plasma concentration
CNSCentral nervous system
CIConfidence interval
CPICClinical Pharmacogenetics Implementation Consortium
C/DConcentration-to-dosage
D2Dopamine D2
D2RDopamine receptor D2
DDIsDrug–drug interactions
DPWGDutch Pharmacogenetics Working Group
DSM-IVDiagnostic and Statistical Manual of Mental Disorders—Fourth Edition
EMAEuropean Medicines Agency and the Medicines
EMsExtensive metabolizers
ERKExtracellular signal-regulated kinase
FDAFood and Drug Administration
FSHFollicle-stimulating hormone
G6PaseGlucose-6-Phosphatase
GCGRGlucagon receptor
GFRGlomerular filtration rate
GLUT4Glucose transporter type 4
GnRHGonadotrophin-releasing hormone
H1, H1RHistamine 1 Receptor
HCPsHealthcare professionals
HPTHypothalamic-pituitary-thyroid
ICD-10International Classification of Diseases, 10th edition
IL-6/2Interleukin-6/2
IRSInsulin receptor substrate
LHLuteinizing hormone
M1Muscarinic acetylcholine
MDMean difference
medianDMedian difference
MHRAHealthcare Products Regulatory Agency
MoAMechanism of Action
mTORMammalian target of rapamycin
NPYNeuropeptide Y
OROdds ratio
P-gpP-glycoprotein
PEPCKPhosphoenolpyruvate carboxykinase
PI3KPhosphoinositide 3-kinase proteins
PMsPoor metabolizers
POMCPro-opiomelanocortin
PPARγPeroxisome proliferator-activated receptor-γ
PPBPlasma protein binding
SFDASaudi Food and Drug Authority
SMDStandardised mean difference
SREBP1/2Sterol regulatory element-binding protein 1/2
T3Triiodothyronine
T4Thyroxine
t1/2Half-life
TNF-αTumour necrosis factor-alpha
TRHThyrotropin-releasing hormone
TSHThyroid-stimulating hormone
UGTUridine diphosphate glucuronosyltransferase
VdVolume distribution
WHOWorld Health Organization
α-MSHα-melanocyte stimulating hormone
α1/α2alpha-1/2 adrenergic
5-HT1A/2A5-Hydroxytryptamine (Serotonin) 1A/2A subtype
5-HT2C5-Hydroxytryptamine (Serotonin) 2C subtype

References

  1. Hodges, H.; Fealko, C.; Soares, N. Autism spectrum disorder: Definition, epidemiology, causes, and clinical evaluation. Transl. Pediatr. 2020, 9 (Suppl. 1), S55. [Google Scholar] [CrossRef]
  2. O’Nions, E.; Petersen, I.; Buckman, J.E.; Charlton, R.; Cooper, C.; Corbett, A.; Happé, F.; Manthorpe, J.; Richards, M.; Saunders, R. Autism in England: Assessing underdiagnosis in a population-based cohort study of prospectively collected primary care data. Lancet Reg. Health Eur. 2023, 29, 100626. [Google Scholar] [CrossRef]
  3. AlBatti, T.H.; Alsaghan, L.B.; Alsharif, M.F.; Alharbi, J.S.; BinOmair, A.I.; Alghurair, H.A.; Aleissa, G.A.; Bashiri, F.A. Prevalence of autism spectrum disorder among Saudi children between 2 and 4 years old in Riyadh. Asian J. Psychiatry 2022, 71, 103054. [Google Scholar] [CrossRef]
  4. Maenner, M.J. Prevalence and characteristics of autism spectrum disorder among children aged 8 years—Autism and developmental disabilities monitoring network, 11 sites, United States, 2020. MMWR Surveill. Summ. 2023, 72, 1–14. [Google Scholar] [CrossRef]
  5. Hassan, A. Arab views on autism. In Encyclopedia of Autism Spectrum Disorders; Springer: Berlin/Heidelberg, Germany, 2021; pp. 302–305. [Google Scholar]
  6. Davico, C.; Secci, I.; Vendrametto, V.; Vitiello, B. Pharmacological treatments in autism spectrum disorder: A narrative review. J. Psychopathol. 2023, 29, 38–52. [Google Scholar]
  7. Zhou, M.S.; Nasir, M.; Farhat, L.C.; Kook, M.; Artukoglu, B.B.; Bloch, M.H. Meta-analysis: Pharmacologic treatment of restricted and repetitive behaviors in autism spectrum disorders. J. Am. Acad. Child Adolesc. Psychiatry 2021, 60, 35–45. [Google Scholar] [CrossRef]
  8. de Pablo, G.S.; Jorda, C.P.; Vaquerizo-Serrano, J.; Moreno, C.; Cabras, A.; Arango, C.; Hernández, P.; Veenstra-VanderWeele, J.; Simonoff, E.; Fusar-Poli, P. Systematic review and meta-analysis: Efficacy of pharmacological interventions for irritability and emotional dysregulation in autism spectrum disorder and predictors of response. J. Am. Acad. Child Adolesc. Psychiatry 2023, 62, 151–168. [Google Scholar] [CrossRef]
  9. Vita, G.; Nöhles, V.B.; Ostuzzi, G.; Barbui, C.; Tedeschi, F.; Heuer, F.H.; Keller, A.; DelBello, M.P.; Welge, J.A.; Blom, T.J.; et al. Systematic Review and Network Meta-Analysis: Efficacy and Safety of Antipsychotics vs. Antiepileptics or Lithium for Acute Mania in Children and Adolescents. J. Am. Acad. Child Adolesc. Psychiatry 2025, 64, 143–157. [Google Scholar] [CrossRef] [PubMed]
  10. Persico, A.M.; Ricciardello, A.; Lamberti, M.; Turriziani, L.; Cucinotta, F.; Brogna, C.; Vitiello, B.; Arango, C. The pediatric psychopharmacology of autism spectrum disorder: A systematic review-Part I: The past and the present. Prog. Neuropsychopharmacol. Biol. Psychiatry 2021, 110, 110326. [Google Scholar] [CrossRef]
  11. Alsayouf, H.A. Growing evidence of pharmacotherapy effectiveness in managing attention-deficit/hyperactivity disorder in young children with or without autism spectrum disorder: A minireview. Front. Psychiatry 2024, 15, 1408876. [Google Scholar] [CrossRef]
  12. D’Alò, G.L.; De Crescenzo, F.; Amato, L.; Cruciani, F.; Davoli, M.; Fulceri, F.; Minozzi, S.; Mitrova, Z.; Morgano, G.P.; Nardocci, F. Impact of antipsychotics in children and adolescents with autism spectrum disorder: A systematic review and meta-analysis. Health Qual. Life Outcomes. 2021, 19, 33. [Google Scholar] [CrossRef]
  13. Fonseca, M.; Carmo, F.; Martel, F. Metabolic effects of atypical antipsychotics: Molecular targets. J. Neuroendocrinol. 2023, 35, e13347. [Google Scholar] [CrossRef] [PubMed]
  14. Jensen, K.G. Severe hyperprolactinemia during lurasidone treatment in a 16-year old girl with schizophrenia–a case report. Scand. J. Child Adolesc. Psychiatry Psychol. 2022, 10, 87. [Google Scholar]
  15. Carli, M.; Kolachalam, S.; Longoni, B.; Pintaudi, A.; Baldini, M.; Aringhieri, S.; Fasciani, I.; Annibale, P.; Maggio, R.; Scarselli, M. Atypical antipsychotics and metabolic syndrome: From molecular mechanisms to clinical differences. Pharmaceuticals 2021, 14, 238. [Google Scholar] [CrossRef]
  16. Jiang, Y.; Ni, W. Estimating the impact of adherence to and persistence with atypical antipsychotic therapy on health care costs and risk of hospitalization. Pharmacother. J. Hum. Pharmacol. Drug Ther. 2015, 35, 813–822. [Google Scholar] [CrossRef]
  17. Panagioti, M.H.A.; Planner, C.; Dhingra, N.; Gupta, N. Global Burden of Preventable Medication-Related Harm in Health Care: A Systematic Review; World Health Organization (WHO): Geneva, Switzerland, 2024; p. 43. [Google Scholar]
  18. Robinson, M.; Amare, M. Adverse drug reactions. Anaesth. Intensive Care Med. 2023, 24, 210–214. [Google Scholar] [CrossRef]
  19. Lee, L.M.; Carias, D.C.; Gosser, R.; Hannah, A.; Stephens, S.; Templeman, W.A. ASHP guidelines on adverse drug reaction monitoring and reporting. Am. J. Health Syst. Pharm. 2022, 79, e83–e89. [Google Scholar] [CrossRef]
  20. Alenzi, K.A.; Alanazi, N.S.; Almalki, M.; Alatawi, F.O. The evaluation of adverse drug reactions in Saudi Arabia: A retrospective observational study. Saudi Pharm. J. 2022, 30, 735–741. [Google Scholar] [CrossRef]
  21. Aronson, J.K. When I use a word… Medical definitions: Adverse events, effects, and reactions. Br. Med. J. 2023, 381, 917. [Google Scholar] [CrossRef]
  22. World Health Organization. Safety of Medicines: A Guide to Detecting and Reporting Adverse Drug Reactions: Why Health Professionals Need to Take Action; Pharmacy Board of Sierra Leone: Freetown, Sierra Leone, 2002. [Google Scholar]
  23. Uitvlugt, E.B.; Janssen, M.J.; Siegert, C.E.; Kneepkens, E.L.; van den Bemt, B.J.; van den Bemt, P.M.; Karapinar-Çarkit, F. Medication-related hospital readmissions within 30 days of discharge: Prevalence, preventability, type of medication errors and risk factors. Front. Pharmacol. 2021, 12, 567424. [Google Scholar] [CrossRef]
  24. Khan, M.A.A.; Hamid, S.; Babar, Z.-U.-D. Pharmacovigilance in high-income countries: Current developments and a review of literature. Pharmacy 2023, 11, 10. [Google Scholar] [CrossRef]
  25. Osanlou, R.; Walker, L.; Hughes, D.A.; Burnside, G.; Pirmohamed, M. Adverse drug reactions, multimorbidity and polypharmacy: A prospective analysis of 1 month of medical admissions. BMJ Open 2022, 12, e055551. [Google Scholar] [CrossRef] [PubMed]
  26. Abu Esba, L.C.; Al Mardawi, G.; AlJasser, M.I.; Aljohani, B.; Abu Alburak, A. Adverse drug reactions spontaneously reported at a tertiary care hospital and preventable measures implemented. J. Clin. Pharm. Ther. 2021, 46, 460–469. [Google Scholar] [CrossRef] [PubMed]
  27. Syahfitri, H. Adverse Drug Reactions of Atypical Antipsychotics: A Review. IOSR J. Pharm. 2021, 11, 37–45. [Google Scholar]
  28. Matijević, B.; Delalić, Đ.; Meštrović, D.; Petrinović, M.; Jug, J.; Prkačin, I. Side-effects of Medications in Emergency Medicine. Cardiol. Croat. 2022, 17, 96–101. [Google Scholar] [CrossRef]
  29. Srisuriyachanchai, W.; Cox, A.R.; Kampichit, S.; Jarernsiripornkul, N. Severity and management of adverse drug reactions reported by patients and healthcare professionals: A cross-sectional survey. Int. J. Environ. Res. Public Health 2023, 20, 3725. [Google Scholar] [CrossRef]
  30. Palanivel, M.; Suresh, A.; Palaniappan, D.R.; Srinivasan, D. A Step towards Patient Safety by Comparing a Trigger Tool with Pre-Existing Tools in the Pharmacovigilance. Indian. J. Pharm. Pract. 2024, 17, 167–180. [Google Scholar] [CrossRef]
  31. Biswas, M.; Vanwong, N.; Sukasem, C. Pharmacogenomics in clinical practice to prevent risperidone-induced hyperprolactinemia in autism spectrum disorder. Pharmacogenomics 2022, 23, 493–503. [Google Scholar] [CrossRef]
  32. Xiao, T.; Hu, J.Q.; Liu, S.J.; Lu, H.Q.; Li, X.L.; Kong, W.; Huang, S.Q.; Zhu, X.Q.; Zhang, M.; Lu, H.Y. Population pharmacokinetics and dosing optimization of olanzapine in Chinese paediatric patients: Based on the impact of sex and concomitant valproate on clearance. J. Clin. Pharm. Ther. 2022, 47, 1811–1819. [Google Scholar] [CrossRef]
  33. Chieh, A.Y.; Bryant, B.M.; Kim, J.W.; Li, L. Systematic review investigating the relationship between autism spectrum disorder and metabolic dysfunction. Res. Autism Spectr. Disord. 2021, 86, 101821. [Google Scholar] [CrossRef]
  34. Goltz, J.; Ivanov, I.; Rice, T.R. Second generation antipsychotic-induced weight gain in youth with autism spectrum disorders: A brief review of mechanisms, monitoring practices, and indicated treatments. Int. J. Dev. Disabil. 2021, 67, 159–167. [Google Scholar] [CrossRef]
  35. Alsabhan, J.F.; Al Backer, N.B.; Hassan, F.M.; Albaker, A.B.; Assiry, G. Metabolic Side Effects of Risperidone in Pediatric Patients with Neurological Disorders: A Prospective Cohort Study. J. Clin. Med. 2024, 13, 5565. [Google Scholar] [CrossRef]
  36. Carnovale, C.; Battini, V.; Santoro, C.; Riccio, M.P.; Carucci, S.; Nobile, M.; Formisano, P.; Bravaccio, C.; Zuddas, A.; Clementi, E. Umbrella review: Association between antipsychotic drugs and metabolic syndrome hallmarks in children and adolescents. J. Am. Acad. Child Adolesc. Psychiatry 2024, 63, 313–335. [Google Scholar] [CrossRef]
  37. Arango, C.; Ng-Mak, D.; Finn, E.; Byrne, A.; Loebel, A. Lurasidone compared to other atypical antipsychotic monotherapies for adolescent schizophrenia: A systematic literature review and network meta-analysis. Eur. Child Adolesc. Psychiatry 2020, 29, 1195–1205. [Google Scholar] [CrossRef]
  38. Pagsberg, A.K.; Tarp, S.; Glintborg, D.; Stenstrøm, A.D.; Fink-Jensen, A.; Correll, C.U.; Christensen, R. Acute antipsychotic treatment of children and adolescents with schizophrenia-spectrum disorders: A systematic review and network meta-analysis. J. Am. Acad. Child Adolesc. Psychiatry 2017, 56, 191–202. [Google Scholar] [CrossRef]
  39. DelBello, M.P.; Kadakia, A.; Heller, V.; Singh, R.; Hagi, K.; Nosaka, T.; Loebel, A. Systematic review and network meta-analysis: Efficacy and safety of second-generation antipsychotics in youths with bipolar depression. J. Am. Acad. Child Adolesc. Psychiatry 2022, 61, 243–254. [Google Scholar] [CrossRef] [PubMed]
  40. Kim, M.J.; Lee, Y.J.; Choe, Y.; Shin, C.H.; Lee, Y.A. Predictors for thyroid dysfunction after discontinuation of levothyroxine in children and adolescents with Hashimoto thyroiditis. Ann. Pediatr. Endocrinol. Metab. 2024, 29, 337. [Google Scholar] [CrossRef]
  41. Koch, M.T.; Carlson, H.E.; Kazimi, M.M.; Correll, C.U. Antipsychotic-related prolactin levels and sexual dysfunction in mentally ill youth: A 3-month cohort study. J. Am. Acad. Child Adolesc. Psychiatry 2023, 62, 1021–1050. [Google Scholar] [CrossRef]
  42. Krøigaard, S.M.; Clemmensen, L.; Tarp, S.; Pagsberg, A.K. A meta-analysis of antipsychotic-induced hypo-and hyperprolactinemia in children and adolescents. J. Child Adolesc. Psychopharmacol. 2022, 32, 374–389. [Google Scholar] [CrossRef]
  43. Khan, A.A.; Sharma, R.; Ata, F.; Khalil, S.K.; Aldien, A.S.; Hasnain, M.; Sadiq, A.; Bilal, A.B.I.; Mirza, W. Systematic review of the association between thyroid disorders and hyperprolactinemia. Thyroid. Res. 2025, 18, 1. [Google Scholar] [CrossRef]
  44. Menard, M.-L.; Thümmler, S.; Giannitelli, M.; Cruzel, C.; Bonnot, O.; Cohen, D.; Askenazy, F.; Boublil, M.; Chambry, J.; Charvet, D.; et al. Incidence of adverse events in antipsychotic-naïve children and adolescents treated with antipsychotic drugs: Results of a multicenter naturalistic study (ETAPE). Eur. Neuropsychopharmacol. 2019, 29, 1397–1407. [Google Scholar] [CrossRef] [PubMed]
  45. Fraguas, D.; Merchán-Naranjo, J.; Laita, P.; Parellada, M.; Moreno, D.; Ruiz-Sancho, A.; Cifuentes, A.; Giráldez, M.; Arango, C. Metabolic and hormonal side effects in children and adolescents treated with second-generation antipsychotics. J. Clin. Psychiatry 2008, 69, 1166–1175. [Google Scholar] [CrossRef]
  46. Zhang, J.-X.; Li, X. Changes in serum thyroid hormone levels in psychiatric patients treated with second-generation antipsychotics. Endokrynol. Pol. 2020, 71, 292–298. [Google Scholar] [CrossRef]
  47. Kelly, D.L.; Conley, R.R. Thyroid function in treatment-resistant schizophrenia patients treated with quetiapine, risperidone, or fluphenazine. J. Clin. Psychiatry 2005, 66, 20986. [Google Scholar] [CrossRef]
  48. Santos, N.C.; Costa, P.; Ruano, D.; Macedo, A.; Soares, M.J.; Valente, J.; Pereira, A.T.; Azevedo, M.H.; Palha, J.A. Revisiting thyroid hormones in schizophrenia. J. Thyroid. Res. 2012, 2012, 569147. [Google Scholar] [CrossRef]
  49. Jazi, S.; Ben-Amor, L.; Abadie, P.; Menard, M.-L.; Choquette, R.; Berthiaume, C.; Mottron, L.; Ilies, D. Long-term metabolic monitoring of youths treated with second-generation antipsychotics 5 years after publication of the CAMESA guidelines are we making Progress? Surveillance Métabolique à long Terme des Jeunes Traités par Antipsychotiques de Deuxième Génération, Cinq ans Après la publication des Lignes Directrices Camesa: Faisons-nous des Progrès? Can. J. Psychiatry 2021, 66, 645–656. [Google Scholar]
  50. Khoodoruth, M.A.S.; Abdo, A.K.A.; Ouanes, S. Quetiapine-induced thyroid dysfunction: A systematic review. J. Clin. Pharmacol. 2022, 62, 20–35. [Google Scholar] [CrossRef]
  51. Alvarez-Herrera, S.; Escamilla, R.; Medina-Contreras, O.; Saracco, R.; Flores, Y.; Hurtado-Alvarado, G.; Maldonado-García, J.L.; Becerril-Villanueva, E.; Pérez-Sánchez, G.; Pavón, L. Immunoendocrine peripheral effects induced by atypical antipsychotics. Front. Endocrinol. 2020, 11, 195. [Google Scholar] [CrossRef]
  52. Findling, R.L.; Pathak, S.; Earley, W.R.; Liu, S.; DelBello, M.P. Efficacy and safety of extended-release quetiapine fumarate in youth with bipolar depression: An 8 week, double-blind, placebo-controlled trial. J. Child Adolesc. Psychopharmacol. 2014, 24, 325–335. [Google Scholar] [CrossRef]
  53. Beaulieu, J.-M.; Gainetdinov, R.R. The physiology, signaling, and pharmacology of dopamine receptors. Pharmacol. Rev. 2011, 63, 182–217. [Google Scholar] [CrossRef]
  54. Chien, H.-Y.; Chen, S.-M.; Li, W.-C. Dopamine receptor agonists mechanism of actions on glucose lowering and their connections with prolactin actions. Front. Clin. Diabetes Healthc. 2023, 4, 935872. [Google Scholar] [CrossRef]
  55. Libowitz, M.R.; Nurmi, E.L. The burden of antipsychotic-induced weight gain and metabolic syndrome in children. Front. Psychiatry 2021, 12, 623681. [Google Scholar] [CrossRef]
  56. Soria-Chacartegui, P.; Villapalos-García, G.; Zubiaur, P.; Abad-Santos, F.; Koller, D. Genetic polymorphisms associated with the pharmacokinetics, pharmacodynamics and adverse effects of olanzapine, aripiprazole and risperidone. Front. Pharmacol. 2021, 12, 711940. [Google Scholar] [CrossRef]
  57. Al-Zoairy, R.; Pedrini, M.T.; Khan, M.I.; Engl, J.; Tschoner, A.; Ebenbichler, C.; Gstraunthaler, G.; Salzmann, K.; Bakry, R.; Niederwanger, A. Serotonin improves glucose metabolism by Serotonylation of the small GTPase Rab4 in L6 skeletal muscle cells. Diabetol. Metab. Syndr. 2017, 9, 1. [Google Scholar] [CrossRef] [PubMed]
  58. Ferreira, V.; Grajales, D.; Valverde, Á.M. Adipose tissue as a target for second-generation (atypical) antipsychotics: A molecular view. Biochim. Biophys. Acta Mol. Cell Biol. Lipids 2020, 1865, 158534. [Google Scholar] [CrossRef]
  59. Wang, K.-Y.; Tanimoto, A.; Yamada, S.; Guo, X.; Ding, Y.; Watanabe, T.; Watanabe, T.; Kohno, K.; Hirano, K.-I.; Tsukada, H. Histamine regulation in glucose and lipid metabolism via histamine receptors: Model for nonalcoholic steatohepatitis in mice. Am. J. Pathol. 2010, 177, 713–723. [Google Scholar] [CrossRef]
  60. Ren, L.; Zhou, X.; Huang, X.; Wang, C.; Li, Y. The IRS/PI3K/Akt signaling pathway mediates olanzapine-induced hepatic insulin resistance in male rats. Life Sci. 2019, 217, 229–236. [Google Scholar] [CrossRef]
  61. Nagata, M.; Yokooji, T.; Nakai, T.; Miura, Y.; Tomita, T.; Taogoshi, T.; Sugimoto, Y.; Matsuo, H. Blockade of multiple monoamines receptors reduce insulin secretion from pancreatic β-cells. Sci. Rep. 2019, 9, 16438. [Google Scholar] [CrossRef]
  62. Zhao, S.; Lin, Q.; Xiong, W.; Li, L.; Straub, L.; Zhang, D.; Zapata, R.; Zhu, Q.; Sun, X.-N.; Zhang, Z. Hyperleptinemia contributes to antipsychotic drug–associated obesity and metabolic disorders. Sci. Transl. Med. 2023, 15, eade8460. [Google Scholar] [CrossRef]
  63. Khan, M.M.; Khan, Z.A.; Khan, M.A. Metabolic complications of psychotropic medications in psychiatric disorders: Emerging role of de novo lipogenesis and therapeutic consideration. World J. Psychiatry 2024, 14, 767. [Google Scholar] [CrossRef]
  64. Anjum, F.; Ali, M.M.; Jakoby, M.; Williams, V. Abstract# 1401109: Quetiapine-induced Hypoglycemia in an Elderly Non-diabetic Patient with Dementia. Endocr. Pract. 2023, 29, S9–S10. [Google Scholar]
  65. Vasiliu, O. Therapeutic management of atypical antipsychotic-related metabolic dysfunctions using GLP-1 receptor agonists: A systematic review. Exp. Ther. Med. 2023, 26, 355. [Google Scholar] [CrossRef]
  66. Ebrahimian, Z.; Razavi, B.M.; Mousavi Shaegh, S.A.; Hosseinzadeh, H. Exploring the therapeutic potential of chlorogenic acid in alleviating olanzapine-induced metabolic syndrome in rats: A key role of hypothalamic satiety proteins. Nutr. Neurosci. 2025, 28, 1055–1074. [Google Scholar] [CrossRef]
  67. Ballon, J.S.; Pajvani, U.; Freyberg, Z.; Leibel, R.L.; Lieberman, J.A. Molecular pathophysiology of metabolic effects of antipsychotic medications. Trends Endocrinol. Metab. 2014, 25, 593–600. [Google Scholar] [CrossRef] [PubMed]
  68. Zhuo, C.; Xu, Y.; Hou, W.; Chen, J.; Li, Q.; Liu, Z.; Dou, G.; Sun, Y.; Li, R.; Ma, X. Mechanistic/mammalian target of rapamycin and side effects of antipsychotics: Insights into mechanisms and implications for therapy. Transl. Psychiatry 2022, 12, 13. [Google Scholar] [CrossRef] [PubMed]
  69. Sobiś, J.; Kunert, Ł.; Rykaczewska-Czerwińska, M.; Świętochowska, E.; Gorczyca, P. The effect of aripiprazole on leptin levels of patients with chronic schizophrenia and a comparison of leptin, acute phase protein, and cytokine levels with regard to body mass and body composition indexes. Endokrynol. Pol. 2022, 73, 35–42. [Google Scholar] [CrossRef]
  70. Zhao, X.; Zhu, W.; Bu, Y.; Li, J.; Hao, Y.; Bi, Y. Effects of 6-week olanzapine treatment on serum IL-2, IL-4, IL-8, IL-10, and TNF-α levels in drug-naive individuals with first-episode schizophrenia. BMC Psychiatry 2024, 24, 703. [Google Scholar] [CrossRef]
  71. Pillinger, T.; McCutcheon, R.A.; Vano, L.; Mizuno, Y.; Arumuham, A.; Hindley, G.; Beck, K.; Natesan, S.; Efthimiou, O.; Cipriani, A. Comparative effects of 18 antipsychotics on metabolic function in patients with schizophrenia, predictors of metabolic dysregulation, and association with psychopathology: A systematic review and network meta-analysis. Lancet Psychiatry 2020, 7, 64–77. [Google Scholar] [CrossRef]
  72. Spina, E.; Barbieri, M.A.; Cicala, G.; de Leon, J. Clinically relevant interactions between atypical antipsychotics and anti-infective agents. Pharmaceuticals 2020, 13, 439. [Google Scholar] [CrossRef]
  73. Edinoff, A.N.; Silverblatt, N.S.; Vervaeke, H.E.; Horton, C.C.; Girma, E.; Kaye, A.D.; Kaye, A.; Kaye, J.S.; Garcia, A.J.; Neuchat, E.E. Hyperprolactinemia, clinical considerations, and infertility in women on antipsychotic medications. Psychopharmacol. Bull. 2021, 51, 131. [Google Scholar] [CrossRef]
  74. Jumaili, W.A.; Muzwagi, A. Review of the Long-Term Effect of the Atypical Antipsychotic Medication on the Bone Mineral Density of the Pediatric Patient with Consideration of Autism Spectrum Disorder. J. Pharmacol. Pharmacother. 2022, 13, 24–30. [Google Scholar] [CrossRef]
  75. Stojkovic, M.; Radmanovic, B.; Jovanovic, M.; Janjic, V.; Muric, N.; Ristic, D.I. Risperidone induced hyperprolactinemia: From basic to clinical studies. Front. Psychiatry 2022, 13, 874705. [Google Scholar] [CrossRef]
  76. Baykara, H.B.; Güney, S.A.; Avcil, S.; Buran, B.Ş.; Cıray, R.O.; Ermis, C.; Inal, N. Safety of Atypical Antipsychotics in a Child and Adolescent Inpatient Setting: A Naturalistic Study. Psychiatry Clin. Psychopharmacol. 2024, 34, 109. [Google Scholar] [CrossRef] [PubMed]
  77. Mandal, S.; Chowdhury, S.; Saha, P.K.; Chowdhury, J. Hyperprolactinemia vs. Thyroid Autoimmunity: Which is a Greater Concern with Atypical Antipsychotics? Res. Squar 2024. [Google Scholar] [CrossRef]
  78. Pei, X.; Du, X.; Lu, H.; Liu, D. Successful Rescue of Hypotension Shock Induced by Quetiapine: A Case Report. Ann. Clin. Case Rep. 2024, 9, 2653. [Google Scholar]
  79. Theilade, S.; Christensen, M.B.; Vilsbøll, T.; Knop, F.K. An overview of obesity mechanisms in humans: Endocrine regulation of food intake, eating behaviour and common determinants of body weight. Diabetes Obes. Metab. 2021, 23, 17–35. [Google Scholar] [CrossRef]
  80. Kong, L.; Shen, Y.; Hu, S.; Lai, J. The impact of quetiapine monotherapy or in combination with lithium on the thyroid function in patients with bipolar depression: A retrospective study. CNS Neurosci. Ther. 2024, 30, e14342. [Google Scholar] [CrossRef]
  81. Liang, J.; Ringeling, L.T.; Hermans, R.A.; Bayraktar, I.; Bosch, T.M.; Egberts, K.M.; Kloosterboer, S.M.; de Winter, B.; Dierckx, B.; Koch, B.C. Clinical pharmacokinetics of antipsychotics in pediatric populations: A scoping review focusing on dosing regimen. Expert. Opin. Drug Metab. Toxicol. 2023, 19, 501–509. [Google Scholar] [CrossRef]
  82. Chen, F.; Liu, H.; Wang, B.; Yang, L.; Cai, W.; Jiao, Z.; Yang, Z.; Chen, Y.; Quan, Y.; Xiang, X. Physiologically based pharmacokinetic modeling to understand the absorption of risperidone orodispersible film. Front. Pharmacol. 2020, 10, 1692. [Google Scholar] [CrossRef]
  83. Townsend, R.W. Low Dose Risperidone Every 3.8 Hours: Superior Efficacy In Treatment of Bipolar Disorders. Res. Sq. 2021. [Google Scholar] [CrossRef]
  84. Liu, Y.; Zhang, M.-q.; Jia, J.-y.; Liu, Y.-m.; Liu, G.-y.; Li, S.-j.; Wang, W.; Weng, L.-p.; Yu, C. Bioequivalence and pharmacokinetic evaluation of two formulations of risperidone 2 mg: An open-label, single-dose, fasting, randomized-sequence, two-way crossover study in healthy male Chinese volunteers. Drugs R D 2013, 13, 29–36. [Google Scholar] [CrossRef] [PubMed]
  85. de Leon, J. Personalizing dosing of risperidone, paliperidone and clozapine using therapeutic drug monitoring and pharmacogenetics. Neuropharmacology 2020, 168, 107656. [Google Scholar] [CrossRef]
  86. Caccia, S. Safety and pharmacokinetics of atypical antipsychotics in children and adolescents. Pediatr. Drugs. 2013, 15, 217–233. [Google Scholar] [CrossRef]
  87. Stahl, S.M.; Strawn, J.R. Prescriber’s Guide-Children and Adolescents: Stahl’s Essential Psychopharmacology; Cambridge University Press: Singapore, 2024. [Google Scholar]
  88. de Leon, J. Precision psychiatry: The complexity of personalizing antipsychotic dosing. Eur. Neuropsychopharmacol. 2022, 58, 80–85. [Google Scholar] [CrossRef]
  89. Lu, J.; Yang, Y.; Lu, J.; Wang, Z.; He, Y.; Yan, Y.; Fu, K.; Jiang, W.; Xu, Y.; Wu, R. Effect of CYP2D6 polymorphisms on plasma concentration and therapeutic effect of risperidone. BMC Psychiatry 2021, 21, 70. [Google Scholar] [CrossRef]
  90. Xiang, J.; Xu, N.; Wang, X.; Li, S.; Yu, Q.; Liang, M.; Nan, F.; Shu, S.; Yan, R.; Zhu, Y. Bioequivalence of 2 aripiprazole orally disintegrating tablets in healthy Chinese volunteers under fasting and fed conditions. Clin. Pharmacol. Drug Dev. 2021, 10, 840–849. [Google Scholar] [CrossRef]
  91. Kumar, A.; Singh, H.; Mishra, A.; Mishra, A.K. Aripiprazole: An FDA approved bioactive compound to treat schizophrenia-A mini review. Curr. Drug Discov. Technol. 2020, 17, 23–29. [Google Scholar] [CrossRef]
  92. Kirino, E. Profile of aripiprazole in the treatment of bipolar disorder in children and adolescents. Adolesc. Health Med. Ther. 2014, 5, 211–221. [Google Scholar] [CrossRef]
  93. Biswas, M.; Vanwong, N.; Sukasem, C. Pharmacogenomics and non-genetic factors affecting drug response in autism spectrum disorder in Thai and other populations: Current evidence and future implications. Front. Pharmacol. 2024, 14, 1285967. [Google Scholar] [CrossRef]
  94. Egberts, K.; Reuter-Dang, S.-Y.; Fekete, S.; Kulpok, C.; Mehler-Wex, C.; Wewetzer, C.; Karwautz, A.; Mitterer, M.; Holtkamp, K.; Boege, I. Therapeutic drug monitoring of children and adolescents treated with aripiprazole: Observational results from routine patient care. J. Neural Transm. 2020, 127, 1663–1674. [Google Scholar] [CrossRef]
  95. Findling, R.L.; Kauffman, R.E.; Sallee, F.R.; Carson, W.H.; Nyilas, M.; Mallikaarjun, S.; Shoaf, S.E.; Forbes, R.A.; Boulton, D.W.; Pikalov, A. Tolerability and pharmacokinetics of aripiprazole in children and adolescents with psychiatric disorders: An open-label, dose-escalation study. J. Clin. Psychopharmacol. 2008, 28, 441–446. [Google Scholar] [CrossRef] [PubMed]
  96. Jukić, M.M.; Smith, R.L.; Molden, E.; Ingelman-Sundberg, M. Evaluation of the CYP2D6 haplotype activity scores based on metabolic ratios of 4,700 patients treated with three different CYP2D6 substrates. Clin. Pharmacol. Ther. 2021, 110, 750–758. [Google Scholar] [CrossRef]
  97. Shan, Y.; Cheung, L.; Zhou, Y.; Huang, Y.; Huang, R.S. A systematic review on sex differences in adverse drug reactions related to psychotropic, cardiovascular, and analgesic medications. Front. Pharmacol. 2023, 14, 1096366. [Google Scholar] [CrossRef]
  98. Xin, Y.; Gao, L.; Tuo, Y.; Nie, G.; Mei, Y.; Chen, C.; Wang, J.; Li, S.; Sun, D.; Qian, Q. Understanding inter-individual variability in pharmacokinetics/pharmacodynamics of aripiprazole in children with tic disorders: Individualized administration based on physiological development and CYP2D6 genotypes. Front. Pharmacol. 2022, 13, 1048498. [Google Scholar] [CrossRef]
  99. Rudå, D.; Jensen, K.G.; Decara, M.S.; Klauber, D.G.; Fagerlund, B.; Møllegaard, J.R.; Linnet, K.; Werge, T.; Correll, C.U.; Fink-Jensen, A. CYP2D6 genotyping and antipsychotic-associated extrapyramidal adverse effects in a randomized trial of aripiprazole versus quetiapine extended release in children and adolescents, aged 12–17 years, with first episode psychosis. J. Clin. Psychopharmacol. 2021, 41, 667–672. [Google Scholar] [CrossRef] [PubMed]
  100. Zubiaur, P.; Soria-Chacartegui, P.; Koller, D.; Navares-Gómez, M.; Ochoa, D.; Almenara, S.; Saiz-Rodríguez, M.; Mejía-Abril, G.; Villapalos-García, G.; Román, M. Impact of polymorphisms in transporter and metabolizing enzyme genes on olanzapine pharmacokinetics and safety in healthy volunteers. Biomed. Pharmacother. 2021, 133, 111087. [Google Scholar] [CrossRef]
  101. Maharaj, A.R.; Wu, H.; Zimmerman, K.O.; Autmizguine, J.; Kalra, R.; Al-Uzri, A.; Sherwin, C.M.; Goldstein, S.L.; Watt, K.; Erinjeri, J. Population pharmacokinetics of olanzapine in children. Br. J. Clin. Pharmacol. 2021, 87, 542–554. [Google Scholar] [CrossRef] [PubMed]
  102. Penzak, S.R.; Hon, Y.Y.; Lawhorn, W.D.; Shirley, K.L.; Spratlin, V.; Jann, M.W. Influence of ritonavir on olanzapine pharmacokinetics in healthy volunteers. J. Clin. Psychopharmacol. 2002, 22, 366–370. [Google Scholar] [CrossRef]
  103. Kolli, P.; Kelley, G.; Rosales, M.; Faden, J.; Serdenes, R. Olanzapine pharmacokinetics: A clinical review of current insights and remaining questions. Pharmgenomics Pers. Med. 2023, 16, 1097–1108. [Google Scholar] [CrossRef]
  104. Erickson-Ridout, K.K.; Zhu, J.; Lazarus, P. Olanzapine metabolism and the significance of UGT1A448V and UGT2B1067Y variants. Pharmacogenet Genom. 2011, 21, 539–551. [Google Scholar] [CrossRef]
  105. Mao, J.-H.; Han, L.; Liu, X.-Q.; Jiao, Z. Significant predictors for olanzapine pharmacokinetics: A systematic review of population pharmacokinetic studies. Expert. Rev. Clin. Pharmacol. 2023, 16, 575–588. [Google Scholar] [CrossRef] [PubMed]
  106. Fekete, S.; Wewetzer, C.; Mehler-Wex, C.; Holtkamp, K.; Burger, R.; Reichert, S.; Taurines, R.; Romanos, M.; Gerlach, M.; Egberts, K. Therapeutic drug monitoring in children and adolescents under pharmacotherapy with olanzapine in daily clinical practice. Ther. Drug Monit. 2017, 39, 273–281. [Google Scholar] [CrossRef]
  107. Grothe, D.R.; Calis, K.A.; Jacobsen, L.; Kumra, S.; DeVane, C.L.; Rapoport, J.L.; Bergstrom, R.F.; Kurtz, D.L. Olanzapine pharmacokinetics in pediatric and adolescent inpatients with childhood-onset schizophrenia. J. Clin. Psychopharmacol. 2000, 20, 220–225. [Google Scholar] [CrossRef]
  108. Aichhorn, W.; Marksteiner, J.; Walch, T.; Zernig, G.; Hinterhuber, H.; Stuppaeck, C.; Kemmler, G. Age and gender effects on olanzapine and risperidone plasma concentrations in children and adolescents. J. Child Adolesc. Psychopharmacol. 2007, 17, 665–673. [Google Scholar] [CrossRef]
  109. Keepers, G.A.; Fochtmann, L.J.; Anzia, J.M.; Benjamin, S.; Lyness, J.M.; Mojtabai, R.; Servis, M.; Walaszek, A.; Buckley, P.; Lenzenweger, M.F. The American Psychiatric Association practice guideline for the treatment of patients with schizophrenia. Am. J. Psychiatry 2020, 177, 868–872. [Google Scholar] [CrossRef]
  110. Blacker, C.J. Clinical issues to consider for clozapine patients who vape: A case illustration. Focus 2020, 18, 55–57. [Google Scholar] [CrossRef]
  111. Yükselmiş, U.; Akçay, M.; Alomari, O.; Yılmaz, M.K. A case report of combined hemoperfusion and hemodiafiltration utilization in pediatric severe Quetiapine poisoning. J. Med. Surg. Public Health 2024, 4, 100147. [Google Scholar] [CrossRef]
  112. Ortega-Ruiz, M.; Soria-Chacartegui, P.; Villapalos-García, G.; Abad-Santos, F.; Zubiaur, P. The pharmacogenetics of treatment with quetiapine. Future Pharmacol. 2022, 2, 276–286. [Google Scholar] [CrossRef]
  113. Joshi, K.; Rao, S.; Mehta, S. A Review of Pharmacokinetic and Pharmacodynamic Properties of Quetiapine IR and XR: Insights and Clinical Practice Implications. Cureus 2025, 17, e86258. [Google Scholar] [CrossRef] [PubMed]
  114. Bertol, E.; Vaiano, F.; Argo, A.; Zerbo, S.; Trignano, C.; Protani, S.; Favretto, D. Overdose of quetiapine—A case report with QT prolongation. Toxics 2021, 9, 339. [Google Scholar] [CrossRef]
  115. Shnayder, N.A.; Abdyrakhmanova, A.K.; Nasyrova, R.F. Oxidation of antipsychotics. Encyclopedia 2022, 2, 974–989. [Google Scholar] [CrossRef]
  116. Castberg, I.; Skogvoll, E.; Spigset, O. Quetiapine and drug interactions: Evidence from a routine therapeutic drug monitoring service. J. Clin. Psychiatry 2007, 68, 1540–1545. [Google Scholar] [CrossRef]
  117. Fekete, S.; Hiemke, C.; Gerlach, M. Dose-related concentrations of neuroactive/psychoactive drugs expected in blood of children and adolescents. Ther. Drug Monit. 2020, 42, 315–324. [Google Scholar] [CrossRef]
  118. McConville, B.J.; Arvanitis, L.A.; Thyrum, P.T.; Yeh, C.; Wilkinson, L.A.; Chaney, R.O.; Foster, K.D.; Sorter, M.T.; Friedman, L.M.; Brown, K.L. Pharmacokinetics, tolerability, and clinical effectiveness of quetiapine fumarate: An open-label trial in adolescents with psychotic disorders. J. Clin. Psychiatry 2000, 61, 252–260. [Google Scholar] [CrossRef]
  119. Zubiaur, P.; Fernández-Campos, P.; Navares-Gómez, M.; Soria-Chacartegui, P.; Villapalos-García, G.; Román, M.; Mejía-Abril, G.; Ochoa, D.; Abad-Santos, F. Variants in COMT, CYP3A5, CYP2B6, and ABCG2 alter quetiapine pharmacokinetics. Pharmaceutics 2021, 13, 1573. [Google Scholar] [CrossRef]
  120. Yau, K.; McArthur, E.; Jeyakumar, N.; Tsobo Muanda, F.; Kim, R.B.; Clemens, K.K.; Wald, R.; Garg, A.X. Adverse events with quetiapine and clarithromycin coprescription: A population-based retrospective cohort study. Health Sci. Rep. 2023, 6, e1375. [Google Scholar] [CrossRef]
  121. Jang, J.-H.; Jeong, S.-H. Pharmacokinetic Prediction of Immediate-and Extended-Release Tablets for Patients with Liver Disease Using Whole Body Physiologically-Based Pharmacokinetic Modeling for the Antipsychotic Drug Quetiapine. AAPS PharmSciTech 2025, 26, 8. [Google Scholar] [CrossRef]
  122. Li, K.-Y.; Li, X.; Cheng, Z.-N.; Zhang, B.-K.; Peng, W.-X.; Li, H.-D. Effect of erythromycin on metabolism of quetiapine in Chinese suffering from schizophrenia. Eur. J. Clin. Pharmacol. 2005, 60, 791–795. [Google Scholar] [CrossRef] [PubMed]
  123. Lee, S.-M.; Jang, J.-H.; Jeong, S.-H. Exploring gender differences in pharmacokinetics of central nervous system related medicines based on a systematic review approach. Naunyn-Schmiedeberg’s Arch. Pharmacol. 2024, 397, 8311–8347. [Google Scholar] [CrossRef]
  124. McGrane, I.R.; Salyers, L.A.; Molinaro, J.R.; Munjal, R.C. Roux-en-Y gastric bypass and antipsychotic therapeutic drug monitoring: Two cases. J. Pharm. Pract. 2021, 34, 503–506. [Google Scholar] [CrossRef]
  125. Amerio, A.; Giacomini, C.; Fusar-Poli, L.; Aguglia, A.; Costanza, A.; Serafini, G.; Aguglia, E.; Amore, M. Efficacy and safety of lurasidone in children and adolescents: Recommendations for clinical management and future research. Curr. Pharm. Des. 2021, 27, 4062–4069. [Google Scholar] [CrossRef]
  126. Findling, R.L.; Goldman, R.; Chiu, Y.-Y.; Silva, R.; Jin, F.; Pikalov, A.; Loebel, A. Pharmacokinetics and tolerability of lurasidone in children and adolescents with psychiatric disorders. Clin. Ther. 2015, 37, 2788–2797. [Google Scholar] [CrossRef]
  127. Siwek, M.; Krupa, A.; Wasik, A. Lurasidone–pharmacodynamic and pharmacokinetic properties, clinical potential and interaction risk. Pharmacother. Psychiatry Neurol. 2020, 36, 117–134. [Google Scholar] [CrossRef]
  128. Mostafa, Y.E.; Metwally, M.E.S.; Elsebaei, F. Prominently selective fluorescence approach with distinctive biopharmaceutical utility for analysis of lurasidone in human plasma and urine: Application to in vitro dissolution and content uniformity testing. Luminescence 2024, 39, e4887. [Google Scholar] [CrossRef]
  129. Lin, S.-K. Racial/ethnic differences in the pharmacokinetics of antipsychotics: Focusing on East Asians. J. Pers. Med. 2022, 12, 1362. [Google Scholar] [CrossRef]
  130. Shirley, M. Lurasidone in schizophrenia in adolescents: A profile of its use. Drugs Ther. Perspect. 2021, 37, 347–353. [Google Scholar] [CrossRef]
  131. Mole, T.B.; Furlong, Y.; Clarke, R.J.; Rao, P.; Moore, J.K.; Pace, G.; Van Odyck, H.; Chen, W. Lurasidone for adolescents with complex mental disorders: A case series. J. Pharm. Pract. 2022, 35, 800–804. [Google Scholar] [CrossRef]
  132. Yang, Y.; Wang, Z.; Xiao, T.; Ni, X.; Song, E.; Dai, L.; Chen, Y.; Lu, H.; Shang, D.; Wen, Y. Preliminary Determination of the Therapeutic Reference Range of Lurasidone in Chinese Patients and Analysis of the Factors Influencing Lurasidone Dose-Corrected Concentrations. Ther. Drug Monit. 2022, 47, 545–550. [Google Scholar] [CrossRef] [PubMed]
  133. Chiu, Y.-Y.; Ereshefsky, L.; Preskorn, S.H.; Poola, N.; Loebel, A. Lurasidone drug-drug interaction studies: A comprehensive review. Drug Metab. Drug Interact. 2014, 29, 191–202. [Google Scholar] [CrossRef]
  134. Paun, J.S.; Tank, H.M.; Savalia, V.B.; Chaitanya, J.K. Ziprasidone Hydrochloride Nanosuspension for Bioavailability Enhancement: Design, Development and Evaluation. E3S Web Conf. 2025, 619, 05004. [Google Scholar] [CrossRef]
  135. Khan, A.A.; Strawn, J.R.; Croarkin, P.E. Emerging treatment options in bipolar disorder in adolescents: Focus on ziprasidone. Adolesc. Health Med. Ther. 2010, 1, 137–143. [Google Scholar]
  136. Bao, S.; Yang, S.; Hua, Z.; Li, J.; Zang, Y.; Li, X. Ziprasidone population pharmacokinetics and co-medication effects in Chinese patients. Naunyn-Schmiedeberg’s Arch. Pharmacol. 2024, 397, 9811–9821. [Google Scholar] [CrossRef]
  137. Simon, N.; Torrents, R.; Azorin, J.-M. Comorbidities and the right dose: Antipsychotics. Expert. Opin. Drug Metab. Toxicol. 2022, 18, 507–518. [Google Scholar] [CrossRef]
  138. Sallee, F.R.; Miceli, J.J.; Tensfeldt, T.; Robarge, L.; Wilner, K.; Patel, N.C. Single-dose pharmacokinetics and safety of ziprasidone in children and adolescents. J. Am. Acad. Child Adolesc. Psychiatry 2006, 45, 720–728. [Google Scholar] [CrossRef]
  139. Miceli, J.; Smith, M.; Robarge, L.; Morse, T.; Laurent, A. The effects of ketoconazole on ziprasidone pharmacokinetics—A placebo-controlled crossover study in healthy volunteers. Br. J. Clin. Pharmacol. 2000, 49, 71–76. [Google Scholar] [CrossRef]
  140. Cicala, G.; Barbieri, M.A.; Santoro, V.; Tata, C.; Colucci, P.V.; Vanadia, F.; Drago, F.; Russo, C.; Cutroneo, P.M.; Gagliano, A. Safety and tolerability of antipsychotic drugs in pediatric patients: Data from a 1-year naturalistic study. Front. Psychiatry 2020, 11, 152. [Google Scholar] [CrossRef] [PubMed]
  141. Schoretsanitis, G.; de Leon, J.; Correll, C.U. How can we better address the pharmacokinetics of antipsychotics in children and adolescents? Expert. Opin. Drug Metab. Toxicol. 2024, 20, 719–726. [Google Scholar] [CrossRef]
  142. Chapron, B.D.; Chapron, A.; Leeder, J.S. Recent advances in the ontogeny of drug disposition. Br. J. Clin. Pharmacol. 2022, 88, 4267–4284. [Google Scholar] [CrossRef]
  143. O’hara, K. Paediatric pharmacokinetics and drug doses. Aust. Prescr. 2016, 39, 208. [Google Scholar] [CrossRef]
  144. Sanyal, S.; Calarge, C.A.; Rowan, P.J.; Aparasu, R.R.; Abughosh, S.; Chen, H. Impact of the AACAP practice parameters on the metabolic adverse event monitoring for second-generation antipsychotics (SGAs) in children and adolescents. J. Psychiatr. Res. 2023, 165, 170–173. [Google Scholar] [CrossRef]
  145. Hoekstra, S.; Bartz-Johannessen, C.; Sinkeviciute, I.; Reitan, S.K.; Kroken, R.A.; Løberg, E.-M.; Larsen, T.K.; Rettenbacher, M.; Johnsen, E.; Sommer, I.E. Sex differences in antipsychotic efficacy and side effects in schizophrenia spectrum disorder: Results from the BeSt InTro study. npj Schizophr. 2021, 7, 39. [Google Scholar] [CrossRef]
  146. Johansen, I.T.; Steen, N.E.; Rødevand, L.; Werner, M.C.; Lunding, S.H.; Hjell, G.; Ormerod, M.B.; Agartz, I.; Melle, I.; Lagerberg, T.V. Sex-specific associations between metabolic hormones, severe mental disorders and antipsychotic treatment. Psychoneuroendocrinology 2022, 146, 105927. [Google Scholar] [CrossRef]
  147. Zucker, I.; Prendergast, B.J. Sex differences in pharmacokinetics predict adverse drug reactions in women. Biol. Sex. Differ. 2020, 11, 32. [Google Scholar] [CrossRef]
  148. Klein, S.L.; Morgan, R. The impact of sex and gender on immunotherapy outcomes. Biol. Sex. Differ. 2020, 11, 24. [Google Scholar] [CrossRef]
  149. Jovanović, M.; Vučićević, K.; Miljković, B. Understanding variability in the pharmacokinetics of atypical antipsychotics–focus on clozapine, olanzapine and aripiprazole population models. Drug Metab. Rev. 2020, 52, 1–18. [Google Scholar] [CrossRef]
  150. Brand, B.A.; Haveman, Y.R.; De Beer, F.; De Boer, J.N.; Dazzan, P.; Sommer, I.E. Antipsychotic medication for women with schizophrenia spectrum disorders. Psychol. Med. 2022, 52, 649–663. [Google Scholar] [CrossRef] [PubMed]
  151. Elmeliegy, M.; Vourvahis, M.; Guo, C.; Wang, D.D. Effect of P-glycoprotein (P-gp) inducers on exposure of P-gp substrates: Review of clinical drug–drug interaction studies. Clin. Pharmacokinet. 2020, 59, 699–714. [Google Scholar] [CrossRef]
  152. Watson, S.; Caster, O.; Rochon, P.A.; Den Ruijter, H. Reported adverse drug reactions in women and men: Aggregated evidence from globally collected individual case reports during half a century. EClinicalMedicine 2019, 17, 100188. [Google Scholar] [CrossRef] [PubMed]
  153. Shilbayeh, S.A.R.; Adeen, I.S.; Alhazmi, A.S.; Aldilaijan, K.E.; Aloyouni, S.Y. Risperidone pharmacogenetics: The impact of star alleles’ predicted phenotypes on global safety in autistic children. Int. J. Pharmacol. 2023, 19, 485–504. [Google Scholar] [CrossRef]
  154. Beunk, L.; Nijenhuis, M.; Soree, B.; de Boer-Veger, N.J.; Buunk, A.-M.; Guchelaar, H.J.; Houwink, E.J.; Risselada, A.; Rongen, G.A.; van Schaik, R.H. Dutch Pharmacogenetics Working Group (DPWG) guideline for the gene-drug interaction between CYP2D6, CYP3A4 and CYP1A2 and antipsychotics. Eur. J. Hum. Genet. 2024, 32, 278–285. [Google Scholar] [CrossRef]
  155. Tveito, M.; Molden, E.; Høiseth, G.; Correll, C.U.; Smith, R.L. Impact of age and CYP2D6 genetics on exposure of aripiprazole and dehydroaripiprazole in patients using long-acting injectable versus oral formulation: Relevance of poor and intermediate metabolizer status. Eur. J. Clin. Pharmacol. 2020, 76, 41–49. [Google Scholar] [CrossRef]
  156. Jukic, M.M.; Smith, R.L.; Haslemo, T.; Molden, E.; Ingelman-Sundberg, M. Effect of CYP2D6 genotype on exposure and efficacy of risperidone and aripiprazole: A retrospective, cohort study. Lancet Psychiatry 2019, 6, 418–426. [Google Scholar] [CrossRef]
  157. Ahmed, Z.; Hao, S.; Williamson, T.; McMorris, C.A.; Bousman, C.A. Psychotropic prescribing rates and pharmacogenomic testing implications for autism in the Canadian primary care sentinel surveillance network. Pharmacogenet Genom. 2022, 32, 94–100. [Google Scholar] [CrossRef]
  158. Cui, Y.; Yan, H.; Su, Y.; Wang, L.; Lu, T.; Zhang, D.; Yue, W. CYP2D6 genotype-based dose recommendations for risperidone in Asian people. Front. Pharmacol. 2020, 11, 936. [Google Scholar] [CrossRef]
  159. Chamnanphon, M.; Vanwong, N.; Prommas, S.; Koomdee, N.; Sukprasong, R.; Rachanakul, J.; Nuntharadthanaphong, N.; Hongkaew, Y.; John, S.; Ngamsamut, N. Risperidone plasma concentrations are associated with hyperprolactinemia in autism spectrum disorder children: The impact of CYP2D6 polymorphisms. Res. Autism Spectr. Disord. 2022, 96, 102002. [Google Scholar] [CrossRef]
  160. Merino, D.; Fernandez, A.; Gerard, A.O.; Ben Othman, N.; Rocher, F.; Askenazy, F.; Verstuyft, C.; Drici, M.-D.; Thümmler, S. Adverse Drug Reactions of Olanzapine, Clozapine and Loxapine in Children and Youth: A Systematic Pharmacogenetic Review. Pharmaceuticals 2022, 15, 749. [Google Scholar] [CrossRef]
  161. Baldacci, A.; Saguin, E.; Balcerac, A.; Mouchabac, S.; Ferreri, F.; Gaillard, R.; Colas, M.-D.; Delacour, H.; Bourla, A. Pharmacogenetic guidelines for psychotropic drugs: Optimizing prescriptions in clinical practice. Pharmaceutics 2023, 15, 2540. [Google Scholar] [CrossRef] [PubMed]
  162. Dubale, A.T.; Tareke, A.A.; Butta, F.W.; Shibabaw, A.A.; Eniyew, E.B.; Ahmed, M.H.; Kassie, S.Y.; Demsash, A.W.; Chereka, A.A.; Dube, G.N. Healthcare professionals’ willingness to utilize a mobile health application for adverse drug reaction reporting in a limited resource setting: An input for digital health, 2023. Eur. J. Obstet. Gynecol. Reprod. Biol. X 2024, 23, 100324. [Google Scholar] [CrossRef]
  163. Worakunphanich, W.; Youngkong, S.; Suwankesawong, W.; Anderson, C.; Thavorncharoensap, M. Comparison of patient adverse drug reaction reporting systems in nine selected countries. Int. J. Environ. Res. Public Health 2022, 19, 4447. [Google Scholar] [CrossRef]
  164. Alshammari, T.M. Pharmacovigilance and outcomes: Experience from Saudi Arabia narrative review. Expert. Rev. Pharmacoecon. Outcomes Res. 2025, 25, 7–15. [Google Scholar] [CrossRef]
  165. Costa, C.; Abeijon, P.; Rodrigues, D.A.; Figueiras, A.; Herdeiro, M.T.; Torre, C. Factors associated with underreporting of adverse drug reactions by patients: A systematic review. Int. J. Clin. Pharm. 2023, 45, 1349–1358. [Google Scholar] [CrossRef]
  166. Gauci, R. Digitalisation of Adverse Drug Reactions Information Source; University of Malta: Msida, Malta, 2024. [Google Scholar]
  167. Al-Worafi, Y.M. Adverse Drug Reactions (ADRs) in Developing Countries. In Handbook of Medical and Health Sciences in Developing Countries: Education, Practice, and Research; Springer: Cham, Switzerland, 2023; pp. 1–18. [Google Scholar]
  168. Beversdorf, D.Q.; Anagnostou, E.; Hardan, A.; Wang, P.; Erickson, C.A.; Frazier, T.W.; Veenstra-VanderWeele, J. Precision medicine approaches for heterogeneous conditions such as autism spectrum disorders (The need for a biomarker exploration phase in clinical trials-Phase 2m). Front. Psychiatry 2023, 13, 1079006. [Google Scholar] [CrossRef]
  169. Lacivita, E.; Niso, M.; Mastromarino, M.; Garcia Silva, A.; Resch, C.; Zeug, A.; Loza, M.I.; Castro, M.; Ponimaskin, E.; Leopoldo, M. Knowledge-based design of long-chain arylpiperazine derivatives targeting multiple serotonin receptors as potential candidates for treatment of autism spectrum disorder. ACS Chem. Neurosci. 2021, 12, 1313–1327. [Google Scholar] [CrossRef]
  170. Clavenna, A.; Cartabia, M.; Fortino, I.; Bonati, M. Drug prescription profile in children with autism spectrum disorders. Eur. J. Clin. Pharmacol. 2024, 80, 297–299. [Google Scholar] [CrossRef]
  171. Ritter, C.; Hewitt, K.; McMorris, C.A. Psychotropic polypharmacy among children and youth with autism: A systematic review. J. Child Adolesc. Psychopharmacol. 2021, 31, 244–258. [Google Scholar] [CrossRef]
  172. Dargenio, V.N.; Dargenio, C.; Castellaneta, S.; De Giacomo, A.; Laguardia, M.; Schettini, F.; Francavilla, R.; Cristofori, F. Intestinal barrier dysfunction and microbiota–gut–brain axis: Possible implications in the pathogenesis and treatment of autism spectrum disorder. Nutrients 2023, 15, 1620. [Google Scholar] [CrossRef]
  173. Długosz, A.; Wróblewski, M.; Błaszak, B.; Szulc, J. The Role of Nutrition, Oxidative Stress, and Trace Elements in the Pathophysiology of Autism Spectrum Disorders. Int. J. Mol. Sci. 2025, 26, 808. [Google Scholar] [CrossRef] [PubMed]
  174. De Luca, F. Endocrinological abnormalities in autism. Semin. Pediatr. Neurol. 2020, 35, 100582. [Google Scholar] [CrossRef]
  175. Blake, K.V.; Zaccaria, C.; Domergue, F.; La Mache, E.; Saint-Raymond, A.; Hidalgo-Simon, A. Comparison between paediatric and adult suspected adverse drug reactions reported to the European medicines agency: Implications for pharmacovigilance. Pediatr. Drugs. 2014, 16, 309–319. [Google Scholar] [CrossRef]
  176. Dubrall, D.; Leitzen, S.; Toni, I.; Stingl, J.; Schulz, M.; Schmid, M.; Neubert, A.; Sachs, B. Descriptive analysis of adverse drug reaction reports in children and adolescents from Germany: Frequently reported reactions and suspected drugs. BMC Pharmacol. Toxicol. 2021, 22, 56. [Google Scholar] [CrossRef]
  177. Braykova, R.; Toneva, A. Aspects of Insulin Resistance in Children with Autism. J. IMAB. 2025, 31, 6111–6115. [Google Scholar] [CrossRef]
  178. Aljead, M.; Qashta, A.; Jalal, Z.; Jones, A.M. Review of Autism Spectrum Disorder (ASD): Epidemiology, Aetiology, Pathology, and Pharmacological Treatment. Pharmaceuticals 2025, 18, 1644. [Google Scholar] [CrossRef]
  179. Al-Huseini, S.; Al-Barhoumi, A.; Al-Balushi, M.; Al-Hosni, A.; Al-Mahrouqi, T.; Al-Mahrizi, B.; Jaju, S.; Mirza, H. Effectiveness and adverse effects of risperidone in children with autism spectrum disorder in a naturalistic clinical setting at a university hospital in Oman. Autism Res. Treat. 2022, 2022, 2313851. [Google Scholar] [CrossRef]
  180. Rahim Shilbayeh, S.A.; Adeen, I.S. Management of autism spectrum disorder: A pilot study in saudi paediatrics. Mil. Med. Sci. Lett. 2024, 93, 120–130. [Google Scholar] [CrossRef]
  181. Makary, S.; Abd El Moez, K.; Elsayed, M.; Hassan, H. Second-generation antipsychotic medications and metabolic disturbance in children and adolescents. Egypt. J. Neurol. Psychiatry Neurosurg. 2023, 59, 14. [Google Scholar] [CrossRef]
  182. Man, K.K.; Shao, S.-C.; Chang, Y.-C.; Chi, M.-H.; Jeong, H.E.; Lin, S.-J.; Su, C.-C.; Shin, J.-Y.; Wong, K.H.; Wong, I.C. Cardiovascular and metabolic risk of antipsychotics in children and young adults: A multinational self-controlled case series study. Epidemiol. Psychiatr. Sci. 2021, 30, e65. [Google Scholar] [CrossRef]
  183. Iasevoli, F.; Barone, A.; Buonaguro, E.F.; Vellucci, L.; de Bartolomeis, A. Safety and tolerability of antipsychotic agents in neurodevelopmental disorders: A systematic review. Expert. Opin. Drug Saf. 2020, 19, 1419–1444. [Google Scholar] [CrossRef]
  184. Hernandez, M.; Cullell, N.; Cendros, M.; Serra-Llovich, A.; Arranz, M.J. Clinical utility and implementation of pharmacogenomics for the personalisation of antipsychotic treatments. Pharmaceutics 2024, 16, 244. [Google Scholar] [CrossRef]
  185. Mead, L.; Ayres, A.; Blake, J.A.; Scott, J.G. Monitoring of metabolic side-effects in children and adolescents prescribed antipsychotic medication: A systematic review. Aust. N. Z. J. Psychiatry 2021, 55, 763–771. [Google Scholar] [CrossRef]
  186. Fekete, S.; Güntzel, T.; Egberts, K.; Geissler, J.; Neubert, A.; Gerlach, M.; Romanos, M.; Taurines, R. Serious adverse drug reactions to antipsychotics in minors with multiple disabilities: Preventability and potential cost savings by therapeutic drug monitoring. Pharmacopsychiatry 2023, 56, 32–39. [Google Scholar] [CrossRef]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Article Metrics

Citations

Article Access Statistics

Multiple requests from the same IP address are counted as one view.