Next Article in Journal
G3BP1 Regulates the Cell Cycle by Promoting IFNβ Production to Promote PCV2 Replication and Promotes Nuclear Transfer of Viral Proteins by Direct Binding
Previous Article in Journal
Special Issue “Molecular Mechanisms of Bioactive Nutrients Promoting Health Through Gut Microbiota 2.0”
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Pharmaco-Multiomics: A New Frontier in Precision Psychiatry

Pharmaceutical Sciences Department, College of Pharmacy, QU Health, Qatar University, Doha P.O. Box 2713, Qatar
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2025, 26(3), 1082; https://doi.org/10.3390/ijms26031082
Submission received: 20 December 2024 / Revised: 19 January 2025 / Accepted: 21 January 2025 / Published: 26 January 2025
(This article belongs to the Special Issue Innovative Therapeutic Approaches in Neuropsychiatric Disorders)

Abstract

:
The landscape of psychiatric care is poised for transformation through the integration of pharmaco-multiomics, encompassing genomics, proteomics, metabolomics, transcriptomics, epigenomics, and microbiomics. This review discusses how these approaches can revolutionize personalized treatment strategies in psychiatry by providing a nuanced understanding of the molecular bases of psychiatric disorders and individual pharmacotherapy responses. With nearly one billion affected individuals globally, the shortcomings of traditional treatments, characterized by inconsistent efficacy and frequent adverse effects, are increasingly evident. Advanced computational technologies such as artificial intelligence (AI) and machine learning (ML) play crucial roles in processing and integrating complex omics data, enhancing predictive accuracy, and creating tailored therapeutic strategies. To effectively harness the potential of pharmaco-multiomics approaches in psychiatry, it is crucial to address challenges such as high costs, technological demands, and disparate healthcare systems. Additionally, navigating stringent ethical considerations, including data security, potential discrimination, and ensuring equitable access, is essential for the full realization of this approach. This process requires ongoing validation and comprehensive integration efforts. By analyzing recent advances and elucidating how different omic dimensions contribute to therapeutic customization, this review aims to highlight the promising role of pharmaco-multiomics in enhancing patient outcomes and shifting psychiatric treatments from a one-size-fits-all approach towards a more precise and patient-centered model of care.

1. Introduction

Psychiatric disorders encompass a diverse range of conditions that profoundly impact cognitive, emotional, perceptual, behavioral, and mood-related functioning. This broad category includes depressive disorders, bipolar disorder, anxiety disorders, trauma- and stressor-related disorders, psychotic disorders, dissociative disorders, eating disorders, and personality disorders, alongside neurodevelopmental disorders such as ADHD (attention-deficit/hyperactivity disorder) and autism spectrum disorders [1,2,3,4]. These illnesses are often characterized by significant distress, social dysfunction, and a decline in quality of life, underscoring the fundamental importance of mental health for personal well-being, interpersonal relationships, and societal contributions [5]. The ramifications of mental health issues extend beyond psychological suffering, often exacerbating physical ailments and increasing both disability and mortality rates [6,7]. In 2019, 970 million people worldwide, or one in every eight individuals, were affected by mental disorders, with anxiety and depressive disorders identified as the most prevalent [8]. The COVID-19 pandemic further exacerbated these issues, leading to a substantial increase in anxiety and depressive disorders by 26% and 28%, respectively, in 2020 [9]. Regionally, the situation in Qatar mirrors global trends, with about 20% of adults attending primary care facilities presenting diagnosable mental disorders, predominantly anxiety and depression [10,11]. Notably, Qatar ranked second in the Middle East for the increased burden of premature mortality from mental health disorders, highlighting a significant regional health challenge [10]. Economically, the impact of mental disorders is significant. Financial losses due to mental health issues were estimated at about USD 1.3 trillion in 2010, rising to USD 1.6 trillion in 2019, with projections indicating an increase to nearly USD 2.5 trillion by 2030 [12]. Despite their high prevalence and cost, the effectiveness of pharmacological treatments like antidepressants remain limited. In clinical trials, these medications showed modest response rates of only 42% to 53% for major depressive disorder, affecting an estimated 163 million individuals [13,14]. Furthermore, a substantial proportion of patients, up to 60%, display poor responses and achieve low remission rates after initial drug administration, accompanied frequently by severe adverse drug reactions (ADRs) [15,16,17,18]. These ADRs themselves are responsible for a significant number of hospital admissions, representing between 5% and 10% of cases, thereby elevating both the clinical and economic burdens of mental health care [19] and significantly increasing healthcare costs. In the United States, the annual cost associated with these reactions can reach up to USD 30.1 billion [20]. This underscores the shortcomings of the prevailing trial-and-error approach in psychiatric medication management, an approach closely associated with poor treatment adherence and discontinuation [20,21]. Understanding the variability in patient responses is crucial, as it stems from a complex interplay of genetic, environmental, pathophysiological, and dietary factors. Specifically, genetic differences play a significant role in influencing drug efficacy and the likelihood of ADRs [22,23]. Research indicates that 15% to 30% of the variability in treatment response can be attributed to genetic alleles affecting drug absorption, metabolism, transport, and mechanism of action [20]. Consequently, identifying these specific genetic variants can predict drug efficacy and potential adverse reactions, thereby enhancing treatment outcomes [24]. This evidence collectively mandates a paradigm shift towards personalized medicine in psychiatry. Embarking on this transformative path necessitates embracing a comprehensive approach incorporating the diverse biological and molecular factors that shape individual patient experiences [25,26,27]. Through integration of multiple omics layers, clinicians and researchers can unearth previously inaccessible insights, substantially enriching our molecular understanding of psychiatric disorders [28]. Utilizing extensive datasets from extensive pharmaco-multiomics investigations propels psychiatry towards more precise, personalized therapeutic interventions. These enhance clinical outcomes and provide patient-specific care tailored to improving clinical outcomes and aligning treatments with individual biological and environmental contexts. In this review, we discuss recent findings across various omics layers and underscore their implications for the development of personalized treatment strategies in psychiatry. We detail how each omic contributes to the creation of targeted therapeutic approaches, emphasizing its role in promoting personalized care and improving patient outcomes. Ultimately, our examination aims to highlight the transformative potential of pharmaco-multiomics in establishing sophisticated, patient-centered therapeutic strategies that not only accommodate the biological individuality of each patient but also set new paradigms in the management of psychiatric disorders.

2. Pharmacogenomics in Psychiatry

Pharmacogenomics (PGx) stands at the frontier of personalized medicine in psychiatry, dedicated to understanding how genetic variation affects individual responses to psychiatric drugs [29]. Over the past four decades, extensive research focusing on both direct and indirect evidence has underscored the clinical utility of PGx in guiding medication dosing and selection decisions, particularly for mental health treatments [30]. This research has examined a multitude of genetic variants associated with drug targets such as the serotonin reuptake transporter (SLC6A4), serotonin receptors (HTR2A and HTR2C), and the dopamine D2 receptor (DRD2). Additional significant pharmacogenes include transporter genes like ATP-binding cassette subfamily B member 1 (ABCB1) and organic cation transporter 1 (OCT1). The study of drug-metabolizing enzymes such as cytochrome P450 2C19 (CYP2C19), cytochrome P450 2D6 (CYP2D6), cytochrome P450 2B6 (CYP2B6), and catechol-O-methyltransferase (COMT) has also been extensive. Moreover, the major histocompatibility complex genes, including HLA-A and HLA-B, have been recognized for their roles related to immunogenic responses potentially triggered by psychiatric medications. This broad array of genes and their variants has been incorporated into PGx-driven evidence-based guidelines for many psychiatric drugs, including antidepressants [31,32,33], antipsychotics [34], ADHD treatments [35,36], and mood stabilizers [37]. The integration of genetic testing into prescribing practices has become increasingly necessary, paving the way for personalized medicine. The International Society of Psychiatric Genetics (ISPG) highlights the importance of pharmacogenetic testing in psychiatry, specifically recommending genetic testing for enzymes such as CYP2C19 and CYP2D6. This testing is particularly valuable for patients who have previously exhibited inadequate responses or adverse reactions to certain antidepressants and antipsychotics [38]. The U.S. Food and Drug Administration (FDA) and the Clinical Pharmacogenetics Implementation Consortium (CPIC) advocate for genetic testing prior to the prescription of specific drugs like carbamazepine to optimize therapeutic outcomes and mitigate adverse effects [39]. Guidelines supported by the CPIC along with international resources such as the Dutch Pharmacogenetics Working Group (DPWG) and the Pharmacogenomics Knowledgebase (PharmGKB) are instrumental in translating PGx findings into clinical practice [40,41]. The FDA has also developed a reference table of pharmacogenetic associations, an essential tool for healthcare providers to evaluate drug–gene interactions for optimal therapeutic strategies, including medication dosage and avoidance of toxicities. These guidelines and resources have significantly influenced the labeling of many psychiatric medications in relation to specific genotypes of CYP2C19 and CYP2D6 [40,42]. Despite these advancements, consistent implementation of these guidelines across different clinical settings remains a critical challenge [43]. Pharmacogenomics has uncovered substantial variations in drug pharmacokinetics and pharmacodynamics across individuals and ethnic groups. The acknowledgment of these differences is crucial for optimizing drug dosages, minimizing ADRs, and enhancing overall treatment outcomes. This section of our review pertains to pharmacogenomics, and particularly focuses on the influence of the CYP450 gene family and the HLA gene family in psychiatry, given their proven clinical relevance. By leveraging the actionable insights provided by genotyping these genes, we can refine and personalize treatment strategies, potentially revolutionizing psychiatric care by aligning drug therapies more closely with individual genetic profiles.

2.1. Cytochrome P450 Variability and Its Implications in Psychiatric Pharmacotherapy

Genetic polymorphisms within CYP450 enzymes introduce significant variability in drug metabolism, thereby impacting therapeutic efficacy and the risk of adverse drug reactions (ADRs) [44,45]. These variations create distinct metabolic phenotypes among patients, ranging from poor to ultra-rapid metabolizers, each exhibiting unique responses to medication [46]. Poor metabolizers (PMs), for example, may experience more adverse reactions at standard doses, due to reduced enzyme activity from homozygous non-functional alleles or gene deletion [47]. Conversely, ultra-rapid metabolizers (UMs) might show an inadequate response to drugs administrated at standard doses because of elevated enzyme activity due to multiple active gene copies, posing significant challenges in attaining therapeutic efficacy [48]. Moreover, genetic mutations such as alternative splicing or frame-shifting events can profoundly alter enzyme structure and function. These alterations consequently affect the metabolism of key psychiatric drugs, adding another layer of complexity to managing patient-specific treatment plans [49]. Table 1 presents a comprehensive synthesis of psychotropic medications, including actionable and informative genes, alongside recommendations from the Clinical Pharmacogenetics Implementation Consortium (CPIC) and the Dutch Pharmacogenetics Working Group (DPWG). All data are sourced from the Pharmacogenomics Knowledgebase (PharmGKB). This table is instrumental in guiding clinicians on genotype-specific drug recommendations, enhancing the precision of psychiatric pharmacotherapy. This underlines the critical need to incorporate pharmacogenetic testing into routine psychiatric practice. By tailoring drug dosages and therapy plans to individual genetic profiles, healthcare providers can optimize treatment outcomes and minimize adverse effects, advancing towards a more personalized approach in psychiatry [25,32].

2.1.1. CYP450 Genetic Variations and Antidepressant Treatment

Antidepressants, particularly selective serotonin reuptake inhibitors (SSRIs) and serotonin–norepinephrine reuptake inhibitors (SNRIs), are metabolized extensively by the CYP450 enzyme system. These pharmacotherapeutic agents serve as frontline treatments for a variety of anxiety and depressive disorders [50,51,52]. SSRIs, which include fluoxetine, citalopram, escitalopram, paroxetine, sertraline, and fluvoxamine, share a mechanistic pathway that involves the inhibition of serotonin reuptake, resulting in increased synaptic serotonin levels [53]. Notably, each SSRI is characterized by distinct pharmacokinetic profiles, pharmacodynamics attributes, and a unique spectrum of side effects, emphasizing the need for individualized therapeutic considerations [53].
The role of specific CYP450 enzymes in the metabolism of SSRIs has been extensively documented. Cacabelos et al. noted that approximately 24% of these antidepressants are metabolized by CYP1A2, 5% by CYP2B6, 38% by CYP2C19, 85% by CYP2D6, and 38% by CYP3A4 [54]. All SSRIs inhibit CYP2D6, with fluoxetine and paroxetine being the most potent inhibitors, though interactions with CYP3A4 are generally insignificant [55]. Citalopram and escitalopram are primarily metabolized by the highly polymorphic enzyme CYP2C19. The Clinical Pharmacogenetics Implementation Consortium (CPIC) has established gene-based therapeutic guidelines for these SSRIs based on the CYP2C19 genotype [56,57]. Genetic variations in CYP2C19 can significantly alter the enzyme’s metabolic activity, affecting drug plasma concentrations. For instance, the CYP2C19*1 allele encodes a fully functional enzyme, while the *17 variant is associated with higher activity and the *2 variant with no activity. Patients with poor metabolizing phenotypes exhibit high plasma concentrations of citalopram, prompting FDA recommendations for a 50% dose reduction to avoid the risk of QT prolongation [58,59,60]. Similarly, fluvoxamine is metabolized by CYP2D6. CPIC guidelines recommend a 25–50% initial dose reduction for fluvoxamine in poor metabolizers, allowing for titration to the maximum effective dose while minimizing adverse effects, or considering alternative medications not metabolized by CYP2D6 [58,61]. Similarly, SNRIs present complex metabolic profiles requiring careful consideration of drug–drug interactions and patient-specific factors [62,63]. For example, venlafaxine and duloxetine are extensively metabolized by CYP2D6, with variations in this enzyme drastically affecting their pharmacokinetic behaviors and necessitating individualized dosing schedules, especially in patients with hepatic or renal impairments [64,65,66]. The pharmacokinetics of newer agents such as levomilnacipran, which is preferentially metabolized by CYP3A4, also exemplifies the intricate considerations required in managing treatments due to its significant impact on norepinephrine compared to serotonin reuptake [58,61]. This detailed attention to metabolic pathways ensures efficacy and safety, particularly in populations with genetic predispositions affecting enzyme activity. Additionally, Tricyclic antidepressants (TCAs), such as imipramine, amitriptyline, trimipramine, and clomipramine, represent some of the earliest forms of antidepressant drugs and are primarily metabolized by the CYP2C19 enzyme. However, their biotransformation into active or inert metabolites crucially depends on subsequent processing by the CYP2D6 enzyme [67,68]. This dual-stage metabolism not only illustrates the complexity of drug interactions within the body but also highlights how genetic variations influencing these enzymes can significantly affect both the efficacy and safety profiles of these medications. In contrast to TCAs’ reliance mainly on two cytochrome P450 enzymes for their metabolism, newer serotonin modulators like Mirtazapine involve a broader spectrum of these isoenzymes in its metabolic pathway, including CYP2D6, CYP1A2, and CYP3A4 [69]. In the context of treatment-resistant depression, ketamine and esketamine introduce complex considerations due to genetic polymorphisms affecting their metabolism. These agents are primarily metabolized by polymorphic enzymes such as CYP2B6, CYP3A4, CYP2A6, CYP2C9, and CYP3A5, which influence the conversion of ketamine to norketamine (NK) and then to hydroxynorketamine (HNK), as well as the transformation of esketamine to noresketamine and hydroxynoresketamine. Notably, CYP2B6, which exhibits numerous single-nucleotide polymorphisms leading to 38 variant proteins, demonstrates varying catalytic activities that significantly impact drug exposure and pharmacokinetics. Specifically, decreased-function CYP2B6 variants like CYP2B6*6 and CYP2B6*9 show reduced metabolic activity, directly affecting the efficacy and safety of treatments. Studies indicate that the wild-type CYP2B6 enzymes exhibit the highest activity for the N-demethylation of ketamine and its derivatives, with some variant enzymes showing up to 35% lower activity compared to the wild type. Such genetic discrepancies highlight the necessity for personalized therapeutic strategies similar to those developed for SSRIs and SNRIs based on CYP2C19 and CYP2D6 variations [70]. These insights into the pharmacogenomics of ketamine and esketamine underscore the critical need for clinical validation of genetic influences on therapy outcomes. Understanding these genetic and pharmacological profiles is essential to optimizing treatment strategies in personalized psychiatry, ensuring efficacy and safety for individuals with specific genetic predispositions affecting metabolic enzyme activity.

2.1.2. CYP450 Genetic Variations and Antipsychotic Treatment

Antipsychotics, which are pivotal for managing psychiatric conditions such as schizophrenia, bipolar disorder, and major depression, demonstrate significant pharmacogenetic diversity. Approximately 40% of these drugs are metabolized by the polymorphic enzyme CYP2D6, whose variants, along with those in CYP2C19, critically influence drug metabolism, efficacy, and safety. Key enzymes involved in antipsychotic metabolism include CYP1A2, CYP2D6, CYP3A4, and CYP3A5, each contributing uniquely to these medications’ metabolic pathways [71,72,73]. CYP2D6 plays a primary role in the metabolism of antipsychotics like aripiprazole and risperidone, where genetic polymorphisms can result in a range of metabolic phenotypes from poor to ultra-rapid metabolizers, significantly affecting drug exposure and therapeutic efficacy [74,75]. Moreover, CYP1A2 is essential for the metabolism of medications such as olanzapine, clozapine, and asenapine, with genetic variants potentially altering plasma concentrations and increasing the risk of side effects [76,77]. The enzyme CYP3A4, responsible for the metabolism of several antipsychotics, harbors mutations like the rs680055 variant, which can profoundly impact drug response [77,78]. Less prevalent but still influential, CYP3A5 variations affect drug ratios and plasma levels essential for optimal therapeutic outcomes [79,80]. Additionally, the ABCB1 gene encodes transporters crucial for drug efflux, affecting drug exposure, with variants such as rs1045642 and rs1128503 modifying the clearance and plasma levels of antipsychotics [80,81,82,83]. The interplay of these enzymes underscores the complexity of antipsychotic drug metabolism, highlighting the importance of personalized treatment approaches. For example, poor metabolizers of CYP2D6 may experience higher plasma drug concentrations, necessitating dosage adjustments to mitigate side effects. The FDA has issued dosing recommendations for 24 antipsychotic drugs based on the CYP2D6 phenotype, illustrating the clinical relevance of genetic profiling [74,75]. While pharmacogenetic testing offers significant potential for enhancing treatment outcomes, its role in predicting adverse reactions remains uncertain. Research in this domain has yielded mixed outcomes, underscoring the complexity of genetic influences on drug response. For example, studies show that CYP450 gene genotyping has limited utility in predicting adverse reactions for drugs like nortriptyline or escitalopram [84]. Conversely, other evidence suggests improved outcomes with pharmacogenetically guided treatment decisions, as one study reported a 23% improvement when treatment was tailored based on genetic testing, with no cases of worsened conditions [73]. Nonetheless, some studies indicate that pharmacogenetic analysis may not significantly reduce certain side effects, such as hyperprolactinemia, which highlights the need for further research in this area [85]. Aside from CYP450 enzymes, genetic variations in other molecular targets such as the HTR2C gene and the DRD2*A1 allele also modulate the efficacy and side effect profiles of antipsychotics. For example, variations in HTR2C are associated with metabolic disturbances and weight gain, whereas the DRD2*A1 allele can influence prolactin levels in patients treated with antipsychotics such as clozapine [86,87].

2.1.3. CYP450 Genetic Variations and Mood Stabilizer Treatments

The treatment efficacy and safety of mood stabilizers, pivotal in managing bipolar disorder, are significantly influenced by genetic variations [88]. Drugs such as valproic acid (VPA), carbamazepine (CBZ), and lithium each have distinct metabolic pathways affected by genetic predispositions. Valproic acid is primarily metabolized by CYP2C9, along with contributions of 20–25% from CYP2B6 and CYP2A6 enzymes, but notable variants like CYP2C19*3 and CYP2C19*2 are particularly impactful [89]. These variants alter plasma concentrations of VPA and are linked to an increased risk of side effects, such as significant weight gain observed in Japanese women. Additionally, the CYP2C9*3 variant is associated with the formation of hepatotoxic metabolites, presenting further challenges in the clinical management of VPA dosing. Carbamazepine metabolism involves a more extensive network of enzymes, including CYP2C8, CYP3A4, and epoxide hydrolase gene (EPHX1), which is crucial for converting CBZ into its active metabolite. Variants such as the non-functional CYP3A5*3 allele significantly affect CBZ plasma levels and, by extension, patient responses to therapy, illustrating the critical need for genetic profiling in optimizing drug dosages and reducing adverse effects [90,91,92,93]. Lithium, uniquely among mood stabilizers, does not undergo metabolism by the CYP450 system, as it is excreted renally. Nevertheless, the long-term use of lithium is associated with potential impairments in thyroid and renal function, highlighting the importance of monitoring and managing these effects during treatment [90,91,92].

2.1.4. Human Leukocyte Antigen (HLA) Gene and Psychiatric Treatment

The human leukocyte antigen (HLA) gene family encodes major histocompatibility complex (MHC) proteins, which are crucial for immune and inflammatory responses [94]. Recent studies highlight the significance of HLA genetic variations in the etiology of psychiatric disorders such as bipolar disorder and schizophrenia, though not in depressive disorders and ADHD [95,96]. HLA molecules modulate neural signaling and synaptic integration, influencing behaviors, learning, and memory [97]. Specific HLA alleles have been linked to psychotropic treatment responses and the development of ADRs. For instance, certain Class I and II HLA alleles are implicated in clozapine-induced agranulocytosis [98] and correlate with better treatment outcomes for risperidone in schizophrenia patients due to specific amino acid variants in the HLA-A peptide-binding groove [99]. Additionally, HLA polymorphisms are associated with severe cutaneous adverse reactions (SCARs), like Stevens–Johnson syndrome (SJS) and toxic epidermal necrolysis (TEN), particularly in Asian populations treated with anticonvulsants such as carbamazepine and oxcarbazepine [100], and phenytoin, which are linked to biomarkers HLA-B*15:02 and HLA-A*31:01 [101]. These severe reactions include blistering rashes, hemorrhagic erosions of mucous membranes, skin detachment, fever, and internal organ lesions [102]. The nucleotide sequences of HLA-A and HLA-B are highly polymorphic, with hundreds of variants that influence susceptibility to various diseases and adverse reactions to pharmaceuticals [101,103]. Given the significant impact of HLA genetic diversity on treatment outcomes and ADRs in psychiatry, incorporating HLA genotyping into pharmacogenomic panels for psychiatric patients can enhance individualized therapy. Preemptive HLA testing could help prevent life-threatening side effects and optimize treatment efficacy, thereby improving overall patient care.

3. Pharmacometabolomics in Psychiatry

While pharmacogenomics has been instrumental in advancing precision medicine by predicting drug responses based on genetic profiles, it does not fully capture the influence of environmental and lifestyle factors such as diet, age, nutrition, gender, and the gut microbiome [104], which substantially impact drug efficacy and safety. Introduced in 2006, pharmacometabolomics fill this gap by analyzing both baseline and dynamic post-treatment metabolic profiles, thereby enhancing the predictability of individual drug response variations [105].
A previous study encompassed nearly 800 patients diagnosed with major depressive disorder (MDD) who were treated with the selective serotonin reuptake inhibitors (SSRIs) citalopram and escitalopram. Clinical depression symptoms were evaluated using the Hamilton Depression Rating Scale (HAMD) and the Quick Inventory of Depressive Symptomatology–Clinician Rated (QIDS-C16) at baseline and after 4 and 8 weeks of treatment. Genomic and metabolomic analyses were conducted on blood samples collected at these time points. Pharmacometabolomics-informed pharmacogenomic analyses in this context have explored both exogenous and endogenous metabolomics. A genome-wide association study (GWAS) of 529 patients has been conducted to uncover genetic variants associated with drug response phenotypes such as remission and response, considering covariates such as age, sex, drug, and dose [106,107]. One significant study identified SNPs related to blood concentrations of escitalopram and its metabolite S-didesmethylcitalopram. This GWAS identified significant correlations between SNPs in the CYP2C19 and CYP2D6 genes with the concentrations of escitalopram and S-didesmethylcitalopram, respectively, both of which are enzymes known to metabolize these drugs. Interestingly, the study also pinpointed a novel association with the TRIML1 gene, suggesting its potential involvement in drug metabolism [107,108]. In addition, the PGRN-AMPS study also investigated endogenous metabolites. One pivotal metabolomic analysis focused on glycine within the nitrogen metabolism pathway. In this analysis, plasma samples from 40 patients (20 escitalopram remitters and 20 non-remitters) were screened using a semi-quantitative gas chromatography–mass spectrometry platform. Glycine was identified as significantly associated with clinical response, prompting a follow-up with a tag SNP genotyping study across glycine metabolism pathway genes in a larger cohort of 512 MDD patients. This study revealed associations between SNPs in the GLDC gene and several clinical outcomes, a finding further validated in a replication cohort and functional genomic studies demonstrating altered protein binding in central nervous system-derived cell lines [106]. Subsequent research targeted serotonin, a key metabolite involved in the mechanism of action of SSRIs. Using plasma samples from 290 patients, a quantitative, targeted liquid chromatography–electrochemical coulometric array (LCECA) platform was employed to analyze metabolites primarily from the tryptophan, tyrosine, and tocopherol pathways. Among the 31 metabolites identified, serotonin levels were most significantly correlated with SSRI clinical outcomes. Consequently, baseline serotonin concentrations and their changes after 4 or 8 weeks of SSRI treatment were used as phenotypes for GWAS. This serotonin-focused GWAS identified two major SNP signals in the TSPAN5 and ERICH3 genes, both of which were associated with plasma serotonin levels. Functional genomic studies further revealed that these SNPs influenced serotonin concentrations in cell culture media, highlighting their potential functional relevance in SSRI response [109,110,111].
On the other hand, the data derived from metabolomic studies are pivotal in bridging critical gaps, as they map the regulation of metabolic pathways. This mapping is essential for illustrating interactions between genome-encoded proteins and environmental factors such as medication exposure. As such, the metabolome acts as a dynamic indicator of an individual’s metabolic status at specific times or in response to certain environmental stimuli. Metabolomic studies on ketamine and esketamine have shown that these antidepressants alter key metabolic pathways, such as the tricarboxylic acid cycle and lipid metabolism, influencing mitochondrial function and neurotransmission. These findings, derived from both preclinical and clinical studies, suggest that metabolic changes are integral to the antidepressant effects observed with these compounds. As research advances, these insights could inform personalized treatment strategies for treatment-resistant depression, enhancing clinical management approaches [112]. A focus in untargeted metabolomic studies, particularly within the realm of antipsychotic treatments, reveals frequent associations with carboxylic acids, a group that includes essential molecules like amino acids, citric acid cycle intermediates, and fatty acids. These compounds are vital for neurotransmitter synthesis and functionality [113]. Research underscores the potential links between amino acids and both the pathophysiology of schizophrenia and the therapeutic responses to antipsychotic drugs [114]. Genetic studies linking amino acid binding efficiency with schizophrenia symptoms have spurred further research into the potential therapeutic applications of amino acids and related compounds [115]. Furthermore, studies have explored other metabolite classes like keto acids and organic compounds in the context of antipsychotic treatments. Keto acids, which feature ketone and carboxylic acid groups, are influenced by both medications and dietary factors [116,117]. In some cases, ketogenic diets have shown potential in managing antipsychotic-associated hyperglycemia and improving symptoms. Organic compounds, particularly those containing organooxygen like glucose and glucuronic acid, and organonitrogen compounds such as trimethylamine N-oxide (TMAO), have been linked to the efficacy and side effects of antipsychotics, including their effects on metabolic and cardiovascular health [118,119,120]. Targeted metabolomic studies have highlighted acylcarnitines as important elements associated with antipsychotic use, implicated in various biological functions, including energy production, lipid metabolism, inflammation, and insulin sensitivity [121,122,123,124,125]. Additionally, these studies have found significant changes in neurotransmitter levels in plasma, suggesting their potential as biomarkers for psychopharmacological treatments [126,127]. However, discrepancies observed in brain sample analyses point to the need for further research to clarify these findings [128].
Pharmacometabolomics adds a crucial dimension to the broader field of pharmacogenomics by offering a more intricate understanding of how metabolic profiles and genetic polymorphisms interact to influence drug efficacy and safety. The continual advancements in metabolomics analytical platforms and their integration with other “omics” data ensure that this field will remain at the forefront of personalized medicine, offering new opportunities to optimize therapeutic outcomes for complex disorders.

4. Pharmacotranscriptomics in Psychiatry

The quest for novel targets and reliable biomarkers to predict therapeutic efficacy in psychiatric treatments remains a significant challenge [129,130]. A deeper understanding of how antidepressants modulate gene expression over time in key tissues could dramatically refine treatment strategies and spearhead the development of more effective therapies [131]. RNA, as the immediate product of gene expression and epigenetic modifications, serves as an accurate indicator of an individual’s functional status, making it a prime target for research studies [132]. Studies have compared peripheral RNA expression profiles between patients with major depression and healthy controls, as well as across varied treatment stages. Such comparisons aim to identify potential biomarkers that can prognosticate treatment responses [133].
The complexity of human studies, influenced by variables such as age and comorbidities, requires large cohorts for consistent biomarker identification. Conversely, animal models offer a controlled environment to study antidepressant effects, allowing for precise manipulation of treatment variables [134,135]. These models facilitate deeper cellular and molecular investigations, enhancing our understanding of psychiatric disorders [135,136]. Recent efforts using animal models have focused on deciphering mechanisms behind stress vulnerability and resilience, particularly looking at transcriptional signatures [137,138,139,140,141]. These studies have identified a varied response to treatments, enabling the selection of animal subjects that showcase strong antidepressant effects for further studies on the biological pathways involved in behavioral normalization [142]. Despite the prevalent use of SSRIs in treating major depression, there is a notable scarcity of comprehensive reviews concerning their transcriptional effects in animal models [143,144]. This gap highlights an urgent need to identify common gene targets across species, which could enhance therapeutic effectiveness. Large-scale transcriptome studies have revealed numerous differentially expressed genes in conditions like schizophrenia, particularly in the dorsolateral prefrontal cortex [145,146]. Yet, interpreting these changes remains complicated due to confounders such as antipsychotic drug exposure [147,148].
Pharmacotranscriptomics seeks to elucidate the interactions between drug effects and psychiatric disorders by investigating the transcriptional impact of antidepressants and antipsychotic drugs. Interestingly, these drugs can induce gene expression changes that either reflect or counter the alterations observed in disorders like schizophrenia [149,150,151,152,153]. This complexity underscores the importance of a nuanced understanding of how these medications influence the brain’s transcriptome, informing both treatment strategies and drug development. Transcriptome-wide association studies (TWAS) provide a methodology to directly quantify the impact of gene expression on psychiatric traits. This approach simplifies complex genetic data into gene-level insights, having already offered significant revelations concerning conditions such as ADHD, major depression, treatment-resistant depression, and autism spectrum disorder [154]. Furthermore, transcriptomic data can streamline drug repurposing efforts by utilizing open-source databases that depict interactions between existing drugs and specific gene products. This strategy can accelerate the drug development process, reducing some of the costs and time typically required [155,156,157]. Given the high incidence of comorbidity in psychiatric conditions and frequent polypharmacy, identifying drugs that target consistent gene expression patterns across various disorders could optimize treatment approaches and minimize the need for multiple medications [154].

5. Pharmacomicrobiomics in Psychiatry

The exploration of pharmacomicrobiomics within the realm of personalized psychiatry offers a promising frontier in enhancing the efficacy and specificity of psychiatric treatments [158]. Pharmacomicrobiomics research has highlighted that gut microorganisms affect psychotropic drug absorption, distribution, metabolism, and excretion, leading to interindividual variability in therapeutic outcomes and adverse effects [159]. For instance, variations in microbial composition can influence the pharmacokinetics of antidepressants and antipsychotics, altering their availability and action [159,160].
The gut microbiome significantly influences psychiatric drug efficacy through mechanisms such as bioaccumulation and modulation of metabolic pathways. Strains like Streptococcus salivarius can uptake drugs like aripiprazole and duloxetine, affecting their availability and, hence, treatment efficacy, a process further complicated by interactions like cross-feeding, which can alter microbial composition [161]. Moreover, the gut microbiome impacts brain function indirectly by modifying tryptophan metabolism through the kynurenine pathway, a process linked to conditions like major depressive disorder and schizophrenia [161]. This interaction not only alters neurotransmitter levels such as serotonin but also influences the efficacy of SSRIs, with drugs like escitalopram increasing beneficial microbial metabolites in patients [162].
A broad spectrum of studies collectively illuminate the intricate role of the gut microbiome in modulating drug responses. Foundational research such as the Human Microbiome Project, an initiative by the NIH, provides a comprehensive catalog of microbial profiles, laying the groundwork for understanding microbiome variations that influence pharmacological outcomes [163]. Notably, European initiatives like ONCOBIOME and The Global Microbiome Conservancy extend this understanding by focusing on the microbiome’s role in specific health conditions, including psychiatric disorders [164]. Cussotto et al. (2019) underscores how psychotropic drugs such as antidepressants and antipsychotics can induce significant shifts in gut microbiota composition, which in turn can affect drug efficacy and tolerability [165]. In addition, there is increasing emphasis on the bidirectional interaction between drugs and the gut microbiome, highlighting that microbes can metabolize drugs, and conversely, drugs can modify the gut microbiota composition [166]. One study reviews the effects of atypical antipsychotics on the gut microbiome, suggesting that treatments themselves can influence microbial compositions, thus potentially opening up new avenues for therapeutic strategies [167]. Despite the promising insights these studies provide, the field acknowledges significant variability and challenges in methodology, as pointed out in systematic evaluations using tools like the STORMS checklist. Future research directions emphasize the need for more stringent clinical studies and the development of standardized protocols to reliably harness the gut microbiome’s potential in improving psychiatric treatment outcomes.
Thus, pharmacomicrobiomics stands at the forefront of a transformative shift in precision psychiatry, where thorough scientific exploration promises to unlock tailored therapeutic strategies that consider the microbiome a central player in drug response variability. The potential role of gut microbiota-derived metabolites as biomarkers, as highlighted in various studies, presents another promising avenue for research. Such metabolites might not only clarify the microbiome’s impact on drug responses but also offer new targets for therapeutic intervention, potentially leading to the development of novel psychobiotics [168]. Future research efforts will likely focus on elucidating the mechanisms of gut microbiota to brain communication, which emphasize gut neuropeptides and their systemic effects [169]. The intricate interplay between the gut microbiome and psychiatric pharmacology, supported by a robust foundation of research, heralds a future where treatments are not only centered on individual genetic makeup but also tailored to microbial profiles. This integrated approach promises more effective management of psychiatric disorders, with fewer side effects and improved patient outcomes, marking a significant advance in the pursuit of precision psychiatry. Harnessing the microbiome’s role in pharmacology could revolutionize treatment personalization, allowing clinicians to predict patient responses to drugs and potentially adjust therapies based on microbiome profiles. Despite promising findings, inconsistency in methodology and the need for robust study designs remain challenges. Thus, future research in pharmacomicrobiomics aims to establish clearer guidelines and integrate microbiome analysis into clinical practice, ultimately enhancing psychiatric care through personalized medicine.

6. Pharmacoepigenomics in Psychiatry

Pharmacoepigenomics in psychiatry elucidates the complex interplay between epigenetic mechanisms such as DNA methylation (DNAm) and histone modifications, thereby advancing our understanding of drug responses and therapeutic outcomes in mental health disorders [170]. Epigenetic alterations at gene promoters generally reduce gene expression, whereas methylation within gene bodies may enhance it. These modifications variably influence drug efficacy and patient responses [171,172,173].
As summarized in Table 2, a variety of studies have explored the relationship between DNA methylation patterns and treatment responses in psychiatric conditions. Depression has been notably highlighted in this context, where baseline hypomethylation of two CpG sites within the serotonin receptor type 1A (HTR1A) and 1B (HTR1B) genes derived from whole blood was predictive of poor escitalopram response in a sample of Han Chinese patients with MDD after 8 weeks of treatment [174]. Furthermore, research on DNA methylation (DNAm) of the BDNF gene in relation to antidepressant treatment response offers varied yet crucial insights. Hypomethylation of BDNF promoter regions has been linked to less favorable responses to a range of antidepressants, including fluoxetine, venlafaxine, and escitalopram, underscoring its potential influence on treatment efficacy [174,175]. On the other hand, studies have shown that higher methylation levels at specific BDNF promoter CpGs correlate with better outcomes, particularly in patients with severe depression treated with escitalopram or venlafaxine, suggesting the modulation of methylation could be a targeted approach for enhancing treatment responses [176,177]. The majority of studies focusing on the genetic basis of antidepressant response have concentrated on genes within the serotonin pathway, specifically the serotonin transporter (SLC6A4) and serotonin receptors (HTR1A/1B). The action of many antidepressants involves the inhibition of SLC6A4, which is crucial for mediating the reuptake of serotonin, while HTR1A and HTR1B receptors are integral in amplifying the therapeutic effects of these drugs [178,179,180]. Recent investigations have expanded the analysis into the interaction of DNAm of HTR1A and HTR1B with environmental factors such as childhood adversity in predicting response to antidepressants. Xu et al. examined DNAm in 181 CpG sites of HTR1A and HTR1B, stratifying their analysis into SSRI and non-SSRI groups. This study revealed that hypomethylation of a particular HTR1A CpG site was associated with an improved response in patients treated with non-SSRI antidepressants (SNRIs and NaSSAs), and those with a specific HTR1B rs6298 SNP and high methylation levels showed better SSRI treatment efficacy. Additionally, an interaction between hypermethylation of HTR1A and high levels of childhood adversity correlated significantly with poor treatment outcomes across both medication groups [181]. Despite extensive research, findings from studies on SLC6A4 methylation and antidepressant treatments, such as SSRIs, TCAs, SNRIs, including escitalopram and venlafaxine, have varied. Initial research in Korean patients suggested minimal hypermethylation at a single CpG site linked with non-response [182], whereas subsequent studies in Japanese and European cohorts reported mixed outcomes, correlating SLC6A4 methylation patterns variably with treatment success [183,184,185,186]. In a subset of five MDD and four panic disorder patients, two CpG sites (14 and 15 in Region A) were significantly hypermethylated following three-month treatment with SSRIs (citalopram, fluoxetine, or sertraline) compared to pre-treatment, although the authors reported that the methodology did not permit reliability of results [187]. Depression was associated with decreased IL6 methylation compared to controls, while antidepressant usage, particularly SSRI-class drugs, was associated with an increase in IL6. [188]. Another study conversely observed decreased IL-6 levels after antidepressant treatment [189]. At the genome scale, a study from Japan investigated the potential of predicting the treatment response to paroxetine based on DNA methylation patterns in the peripheral blood of 68 MDD patients. In their analysis, the authors contrasted the 10 best responders and the 10 worst responders to paroxetine treatment and identified the hypermethylation of the genes liprin-alpha-4 (PPFIA4) and heparan sulfate-glucosamine 3-sulfotransferase 1 (HS3ST1) at baseline in patients with the worst treatment outcomes in relation to the best treatment responders [190]. The role of non-coding RNAs (ncRNAs), particularly microRNAs and long non-coding RNAs, is becoming increasingly recognized within psychiatric pharmacoepigenomics. These ncRNAs modulate transcriptional and translational processes and present novel pathways through which drugs can exert pharmacological effects, independent of direct changes to the DNA sequence [191,192]. More ongoing research efforts aim to identify how these epigenetic mechanisms, influenced or modified by pharmacological treatments, may serve as predictive biomarkers for treatment responses or targets for novel therapeutic interventions. Notably, the use of HDAC and DNMT inhibitors has shown promise in enhancing the efficacy of antidepressants in both preclinical models and clinical settings by modifying epigenetic states [193,194,195,196].

7. Pharmacoproteomics in Psychiatry

Pharmacoproteomics is an emerging field within the larger context of biomedical sciences focused on identifying changes in the proteomic landscape as a response to pharmacological treatment. This discipline is particularly relevant in the field of psychiatry, where proteomic profiling has concentrated on elucidating the alterations in specific proteins crucial for cellular communication, signaling, inflammation, and the maintenance and growth of cells following treatment with psychotropic drugs. These insights guide the development of new therapeutic targets, refine drug efficacy assessments, and aid in crafting personalized treatment plans based on protein profiles, thus advancing precision medicine.
Significant advancement has been made in pinpointing proteins that can forecast therapeutic outcomes in psychiatric conditions [198]. For instance, research documented by Schwarz et al. in 2012 identified several proteins, including interleukin-16, fatty acid-binding protein, ferritin, C-reactive protein, myoglobin, prolactin, and complement factor H, that are indicative of symptomatic improvements in schizophrenia patients undergoing antipsychotic treatment [199]. Additionally, other proteins, such as matrix metalloproteinase 2 and insulin, have been associated with enhancements in negative symptoms within this patient population. In the context of depressive disorders, findings from the GENDEP (genome-based therapeutic drugs for depression) study reveal that the levels of C-reactive protein can predict differential responses to pharmacotherapies such as nortriptyline and escitalopram, which have distinct pharmacological profiles [200]. These findings allow clinicians to customize treatments more accurately, potentially minimizing adverse effects and enhancing outcomes.
However, despite these insights, the field has yet to establish proteomic markers validated sufficiently to guide clinical decision-making regarding the selection of psychotropic medications. The GENDEP project consists of a comprehensive series of multidisciplinary studies aimed at discovering predictors of therapeutic response to antidepressants, employing rigorous methodologies across genetics, transcriptomics, and proteomics [201]. For example, a study analyzing antidepressant response in two mouse strains has shed light on molecular mechanisms and potential biomarkers for therapeutic response using advanced quantitative proteomic techniques [202].
Despite its potential, pharmacoproteomics faces hurdles such as the evolving nature of proteomic technologies, the lack of standardized protocols, and the complex proteome of human plasma. Future efforts should focus on rigorous validation across various patient populations and drug classes and establishing interdisciplinary frameworks to standardize proteomic methodologies. By overcoming these challenges, pharmacoproteomics could significantly enhance therapeutic outcomes and minimize adverse effects, thereby refining psychiatric treatment paradigms through precision medicine.

8. Harnessing AI and Machine Learning to Enhance Precision Psychiatry Through Multiomics Integration

The burgeoning field of multiomics presents significant complexities due to its expansive and data-rich nature, posing substantial challenges in data integration and interpretation. To manage this abundance of biological data, machine learning and AI have emerged as pivotal tools. These technologies foster novel approaches to effectively integrate disparate omics layers. Advanced computational techniques enable the extraction of meaningful biological insights and facilitate the prediction of disease phenotypes and treatment outcomes, which will enhance precision psychiatry [203,204,205,206,207,208]. This approach leverages detailed molecular and clinical information to craft personalized treatment regimens and improve diagnostic accuracy beyond traditional methods.

8.1. Tailoring Treatments in Psychiatry Using Integrated Multiomics Data

In psychiatry, the integration of multiomics data holds potential for tailoring treatments effectively across a spectrum of disorders, such as major depressive disorder (MDD) and bipolar disorder [209,210]. For instance, a study developed predictive models using genomics and plasma metabolomics to forecast outcomes of combination pharmacotherapies in treating MDD. They employed penalized linear regression and XGBoost algorithms to construct two distinct models: one utilizing only metabolomics data, and another integrating both metabolomics and genomics. Their findings indicated that multiomics models achieved superior area under the curve (AUC) measures compared to models using only metabolomics data, underscoring the enhanced accuracy of multiomics in predicting treatment responses [210]. Further supporting this, meta-reviews consolidated findings from various studies, demonstrating how combined multiomics and clinical data can effectively identify predictors of treatment response in unipolar depression [211,212]. Additionally, pharmacogenomic reviews have advocated for including machine learning techniques early in clinical trial planning for psychiatric disorders. These reviews emphasize the significant role of genomic data in elucidating the impact of psychotropic drugs, highlighting studies using machine learning to study genomic influences on the efficacy of treatments for MDD and bipolar disorder using lithium [213,214].
Furthermore, the application of machine learning in psychiatry extends beyond mood disorders. Research demonstrated the use of gradient boosting algorithms to combine exome sequencing data with clinical scores, achieving an AUC of 0.75 in predicting treatment responses in treatment-resistant depression, illustrating the robust potential of integrating genetic data with clinical assessments [215]. Similarly, research on treatment-resistant schizophrenia shows parallel efforts across different psychiatric conditions where predictive models are enriched with omics data [216]. For example, a key study investigated modeling responses to combination antidepressant therapies, examining data from MDD outpatients treated with citalopram or escitalopram in the Mayo Clinic PGRN-AMPS and the CO-MED studies. It focused on enhancing prediction accuracy by integrating metabolomics with validated single-nucleotide polymorphisms (SNPs) compared to using only metabolomics. When trained and tested on both patient cohorts, the models achieved accuracies of 76.6% (AUC: 0.85) without SNPs and 77.5% (AUC: 0.86) with SNPs, demonstrating robust cross-trial prediction replication [210]. In a notable study, Lin et al. [217] utilized a sophisticated deep learning framework that combines genetic data such as SNPs, demographic factors like age and sex, and clinical insights including depression severity scores and suicide attempt statuses in a genome-wide association study (GWAS). Employing multi-layer feedforward neural networks (MFNNs) equipped with multiple hidden layers, they achieved promising predictive accuracies, demonstrating the capability of deep learning frameworks to capture the complex interplay between diverse biomarkers and treatment outcomes, thus offering enhanced predictive precision essential for clinical applications. Additionally, various studies have explored the use of conventional AI and machine learning techniques, such as random forest and decision tree algorithms, to improve the predictability of antidepressant responses. These methods analyze combined datasets consisting of genetic, demographic, and clinical data, generating new insights into patient-specific treatment outcomes and enhancing the potential of personalized psychiatric treatment protocols [205,206,207,208,218].
Table 3 provides a comprehensive summary of pivotal studies involving AI and machine learning in psychiatry, specifically focusing on predictive modeling for treatment responses in psychiatric disorders.

8.2. Challenges in AI and Machine Learning Deployment for Personalized Psychiatry

The deployment of AI and machine learning presents striking predictive possibilities that are tempered by compelling limitations. One of the most significant challenges is the “black box” nature of these models, where the lack of transparency in decision-making processes can hinder trust and understanding among clinicians and patients, thereby affecting their ethical integration into clinical practice. Additionally, these models are constrained by data heterogeneity and potential biases; models trained on non-representative datasets may yield skewed predictions that exacerbate health disparities. The issue of generalizability also arises, as models developed in specific clinical or demographic environments often fail to perform accurately across different populations due to genetic, environmental, and disease prevalence variations. Interoperability issues and ethical concerns such as data privacy, autonomy, and informed consent present further barriers to integration within existing healthcare systems. Ethical deliberations in the deployment of AI and machine learning in healthcare encompass examining biases in training datasets and algorithmic decisions, ensuring transparency in AI methodologies, clarifying accountability for AI-driven outcomes, and safeguarding patient privacy. These considerations are crucial, as they address fundamental ethical principles and help foster a climate of trust and reliability in AI and machine learning applications. Critical questions about how biases within these systems are identified and mitigated, the extent of transparency required to maintain patient and clinician trust, how accountability is apportioned in cases of AI error, and the mechanisms protecting patient data must be rigorously examined and addressed by a multidisciplinary team. Furthermore, ethical concerns extend to issues of equity and justice, particularly examining who benefits from these advancements and who may be inadvertently harmed. The emotional and psychological impacts of AI and machine learning decisions on patients also require careful attention, especially in fields like psychiatry, where the implications of errors are profound [219,220,221]. These challenges underscore the necessity for a multidisciplinary approach involving data scientists, clinicians, ethicists, and policymakers to forge AI and ML tools that are transparent, equitable, adaptable, and aligned with both ethical standards and societal values, thereby ensuring their successful implementation in personalized psychiatry.
Table 3. Representative studies on AI and machine learning applications for predictive modeling in psychiatric treatment responses.
Table 3. Representative studies on AI and machine learning applications for predictive modeling in psychiatric treatment responses.
ReferenceSample DescriptionOutcome MeasureModel(s) UsedPrimary Performance MetricData Used
Lin et al. [217]MDD patients
Antidepressants
Treatment response
(8 weeks)
Deep learning architectureAUC: 0.82 for treatment response, 0.81 for remission-
Joyce et al. [210]MDD patients
(PGRN-AMPS)
SSRI
Treatment response
(8 weeks)
Linear penalized regression, XGBoostAUC (multiomics > metabolomics)Multiomics, metabolomics
Chekroud et al. [218]MDD patients; various antidepressantsRemission
(12 weeks)
Tree-based ensembleAccuracy: 0.59Top 25 predictors
Chang et al. [204]MDD patients;
antidepressants
Treatment response
(8 weeks)
Linear regression (ARPNet)Accuracy: 0.84Neuroimaging biomarkers, genetic variants, DNA methylation, demographics information
Athreya et al. [203]MDD patients;
SSRI
Treatment response
(8 weeks)
Random forestAUC: >0.7, accuracy: >0.69SNP datasets
Eugene et al. [222]Bipolar patients
Lithium
Treatment response Decision tree, random forestAUC:0.92 (male; 2 genes)
AUC: 1 (female; 3 genes)
Gene expression data
Cepeda et al. [223]Patients with and without proxy for treatment-resistant depression (TDR); antidepressants and antipsychoticsTreatment resistance
(12 months)
Decision treeAUC: 0.81Health claims databases (CCAE, MDCR, Optum)
Kautzky et al. [224]MDD patients;
antidepressants
Treatment resistance Random forestAccuracy: 0.73 (TRD);
accuracy: 0.85 (remission)
Not specified
Pradier et al. [225]Bipolar patients; antidepressantsConversion to bipolar diagnosis within 3 monthsLASSO logistic regression, random forestAUC: 0.80-

9. Challenges and Future Directions in Personalized Psychiatry

Personalized psychiatry stands at the cusp of a transformative era, significantly propelled by the integration of multiomics approaches. These methods are pioneering enhanced understanding and fostering the development of more effective treatment modalities for psychiatric conditions. However, translating such advanced methodologies into clinical paradigms presents formidable challenges. The inherent complexity and considerable costs associated with multiomics technologies [218] necessitate the implementation of sophisticated, adaptable analytical frameworks. Traditional genomic analyses often fall short in capturing dynamic interactions between biological and environmental factors, further emphasizing the need for advancements in this domain.

9.1. Global Efforts and Initiatives

Amid these challenges, a broad spectrum of coordinated, multi-level efforts underpins the progression of personalized psychiatry. Key global consortiums such as the Psychiatric Genomics Consortium (PGC) [226], the European College of Neuropsychopharmacology (ECNP) [227], and the UK Biobank [228] play pivotal roles in amassing extensive biological and phenotypic datasets from various populations. These data are crucial for delineating the intricate interplay of genetic, environmental, and lifestyle factors. Concurrently, established clinical guidelines set forth by authoritative entities such as the Canadian Pharmacogenomics Network for Drug Safety (CPNDS) [229] and the French National Network of Pharmacogenetics (RNPGx) [230], endorsed by organizations like the American Society of Health-System Pharmacists (ASHP) [231], guide the safe and effective integration of genomic insights into clinical frameworks. Moreover, implementation networks such as IGNITE actively address logistical and operational hurdles in genomic medicine, utilizing tools like the Consolidated Framework for Implementation Research (CFIR) to facilitate practical applications [232,233]. On the frontier of innovation, projects like the PRIME Care program [234] and the PREPARE study [235] within the U-PGx initiative lead the way in integrating preemptive genotyping and pharmacogenomics into clinical trials and healthcare practice. These initiatives and efforts are reinforced by advancements related to the progress of AI/ML in precision medicine, driven by collaborative endeavors across the modeling community, regulatory bodies, and scientific institutions. The Innovation and Quality (IQ) Consortium’s 2022 workshop exemplifies this synergy, promoting AI/ML adoption and advocating for the integration of domain-specific knowledge to enhance model applicability in pharmaceutical research [236]. The prevailing approach favors synergistic integration where AI/ML complements existing pharmacokinetic–pharmacodynamic (PK-PD) models rather than replacing them, thereby enhancing predictive accuracy. This method aggregates AI’s robust data processing capabilities with the biological insights of traditional models, exemplified by hybrid ML-PK/PD/toxicodynamic approaches that optimize therapeutic strategies using complex biomedical data. Regulatory engagement, illustrated by the collaborative workshop with the FDA and the University of Maryland, further highlights AI/ML’s significant role in refining patient-specific treatments [237]. The increasing number of AI/ML-related regulatory submissions to the FDA underscores the growing trust in these technologies to improve clinical trial designs, patient stratification, and treatment adherence [238]. Additionally, the advancement of emerging disciplines such as connectomics, a nascent field crucial for mapping neural connections and understanding how various psychiatric treatments interact with brain systems [239], and the implementation of pharmacogenomic (PGx) scoring systems [240] are integral to ongoing efforts aimed at refining therapeutic approaches for complex psychiatric disorders. These scientific advancements are crucial in aligning with and pushing the frontiers of personalized medicine to develop more targeted and effective treatment protocols for a range of psychiatric conditions.

9.2. Considerations and Perspectives

These multifaceted initiatives exemplify a robust, concerted endeavor to overcome existing barriers such as high costs, technological complexity, and disparate healthcare infrastructures.
Financially, the utilization of omics technologies necessitates significant investment in both cutting-edge equipment and specialized personnel training, making it prohibitive for many institutions, especially those in resource-limited settings. Technologically, the integration and standardization of sophisticated genomic data systems require advanced IT infrastructure and data management skills that are not uniformly available across different healthcare providers. Regional disparities in healthcare infrastructure further accentuate these challenges, with differences in healthcare quality, technological access, and professional training potentially creating significant gaps in the effectiveness of these strategy applications. Such disparities necessitate tailored implementation strategies that consider the specific capacities and needs of local contexts. Ethical considerations also come to the forefront, particularly concerning the security of patient data, potential psychosocial effects, and the risk of discrimination. To ensure the responsible implementation of clinical applications, it is vital that decisions be based on solid evidence weighing potential risks against benefits. This involves strict data governance, comprehensive patient education, and the use of explainable AI and machine learning models. Additionally, policy frameworks must promote equitable access, particularly for vulnerable groups, to mitigate the risk of widening health disparities. Integrating insights from all stakeholders and maintaining transparency throughout all phases of implementation are essential, complemented by continuous validation and cost-effectiveness analyses to ethically advance precision psychiatry. The future success of these endeavors hinges on sustained investments in technological innovation, interdisciplinary research, and global collaboration. Knowledge exchange, facilitated through symposia, workshops, and publications, along with equipping clinicians with necessary tools for targeted interventions, holds the promise of reshaping psychiatric practices. Ultimately, the aim is to meet the unique treatment needs of each patient more effectively and precisely, thereby emphasizing the critical importance of continued validation, research, and implementation of these sophisticated strategies.

10. Conclusions

In this review, we have delved into the multifaceted realm of pharmaco-omics and examined its pivotal contributions to demystifying the complexities inherent in psychiatric disorders. We emphasize the transformative potential of integrating diverse omics fields, including genomics, proteomics, transcriptomics, the microbiome, and metabolomics, into a unified analytical framework. This holistic approach advocates for a personalized treatment strategy meticulously tailored to each patient’s unique biological profile, moving past the conventional trial-and-error methods prevalent in psychiatric practice. Looking to the future, the ability of pharmaco-multiomics to revolutionize psychiatric treatment relies on overcoming several critical challenges. These include the integration of complex multiomics data to yield clinically actionable insights, bridging technological gaps, and upholding ethical standards in data management and privacy (Figure 1). To fully realize and harness the capabilities of a personalized pharmaco-multiomics approach in psychiatry, key strategic measures are crucial:
  • Interdisciplinary Collaborations: Establish robust collaborations among researchers, clinicians, bioinformaticians, and ethicists. Such interdisciplinary partnerships are essential for developing treatment modalities that are predictive, preventive, and deeply personalized.
  • Investment in Technological Infrastructure: Commit substantial investment into developing technological infrastructures and research ecosystems. This is particularly critical in regions like the Middle East, where numerous challenges obstruct the adoption of advanced multiomics strategies.
  • Enhanced International Cooperation: Strengthen international cooperation to facilitate efficient data sharing and the pooling of global expertise. Such collaboration is vital for leveraging diverse knowledge and resources to advance psychiatric treatment modalities.
  • Integration of AI and Machine Learning: Deepen the integration of AI and machine learning technologies within pharmaco-multiomics. These technologies are key to managing large, complex datasets and enabling sophisticated predictive modeling that can inform personalized treatment plans.
  • Overcoming Methodological Barriers: Address critical barriers such as data diversity, sample size, and methodology standardization. It is imperative to resolve these issues to ensure that research findings are replicable and interpretable across different settings.
  • Translation of Multiomics Data: With a robust technological and collaborative framework in place, focus on the effective integration of multiomics approaches to convert complex biological data into tailored therapeutic strategies. This will facilitate the creation of customized treatment plans that meet individual patient needs.
By overcoming existing challenges and implementing key strategic measures, we can fully unlock the potential of pharmaco-multiomics. This heralds a new era in personalized psychiatry that respects the complex biological individuality of each patient, signifying substantial scientific progression and shifting toward a more compassionate, individualized, and effective approach to mental health care.

Author Contributions

Conceptualization, D.D. and K.B.; writing—original draft preparation, D.D.; writing—review and editing, D.D. and K.B.; visualization, K.B. and D.D.; supervision, K.B.; project administration, K.B.; funding acquisition, K.B. All authors have read and agreed to the published version of the manuscript.

Funding

This study was funded by the Qatar National Research Fund (QNRF)/QRDI under grant number PPM 06-0530-230047. Additionally, this grant covered the article-processing charges (APCs).

Acknowledgments

We gratefully acknowledge the invaluable support provided by the Qatar Genome Program (QGP) and the Qatar Precision Health Institute (QPHI).

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

The following abbreviations are used in this manuscript:
CBZCarbamazepine
CPICClinical Pharmacogenetics Implementation Consortium
DNAmDNA methylation
DPWGDutch Pharmacogenetics Working Group
FDAU.S. Food and Drug Administration
HLAHuman leukocyte antigen
MDDMajor depressive disorder
ncRNANon-coding RNA
PGxPharmacogenomics
PharmGKBPharmacogenomics Knowledgebase
PMsPoor metabolizers
SNPSingle-nucleotide polymorphism
SSRIsSelective serotonin reuptake inhibitors
UMsUltra-rapid metabolizers
VPAValproic acid

References

  1. American Psychiatric Association. Diagnostic and Statistical Manual of Mental Disorders; American Psychiatric Association: Washington, WA, USA, 2013; ISBN 0-89042-555-8. [Google Scholar]
  2. National Academies of Sciences, Engineering, and Medicine. Selected Health Conditions and Likelihood of Improvement with Treatment. 2020. Available online: https://www.ncbi.nlm.nih.gov/books/NBK559510/ (accessed on 24 November 2024).
  3. WHO. Mental Disorders; WHO: Geneva, Switzerland, 2022. [Google Scholar]
  4. Healthy People. Mental Health and Mental Disorders; Healthy People: Frankston, Australia, 2020. [Google Scholar]
  5. Kirkbride, J.B.; Anglin, D.M.; Colman, I.; Dykxhoorn, J.; Jones, P.B.; Patalay, P.; Pitman, A.; Soneson, E.; Steare, T.; Wright, T.; et al. The Social Determinants of Mental Health and Disorder: Evidence, Prevention and Recommendations. World Psychiatry 2024, 23, 58–90. [Google Scholar] [CrossRef] [PubMed]
  6. Luppino, F.S.; de Wit, L.M.; Bouvy, P.F.; Stijnen, T.; Cuijpers, P.; Penninx, B.W.J.H.; Zitman, F.G. Overweight, Obesity, and Depression. Arch. Gen. Psychiatry 2010, 67, 220. [Google Scholar] [CrossRef] [PubMed]
  7. Walker, E.R.; McGee, R.E.; Druss, B.G. Mortality in Mental Disorders and Global Disease Burden Implications. JAMA Psychiatry 2015, 72, 334. [Google Scholar] [CrossRef] [PubMed]
  8. Institute of Health Metrics and Evaluation. Global Health Data Exchange (GHDx); Institute of Health Metrics and Evaluation: Seattle, WA, USA, 2021. [Google Scholar]
  9. WHO. Mental Health and COVID-19: Early Evidence of the Pandemic’s Impact: Scientific Brief, 2 March 2022; WHO: Geneva, Switzerland, 2022. [Google Scholar]
  10. Ghuloum, S.; Bener, A.; Abou-Saleh, M.T. Prevalence of Mental Disorders in Adult Population Attending Primary Health Care Setting in Qatari Population. J. Pak. Med. Assoc. 2011, 61, 216–221. [Google Scholar]
  11. Wadoo, O.; Ahmed, M.A.S.; Reagu, S.; Al Abdulla, S.A.; Al Abdulla, M.A.Y.A. Primary Care Mental Health Services in Qatar. BJPsych Int. 2021, 18, 15–18. [Google Scholar] [CrossRef]
  12. Arias, D.; Saxena, S.; Verguet, S. Quantifying the Global Burden of Mental Disorders and Their Economic Value. EClinicalMedicine 2022, 54, 101675. [Google Scholar] [CrossRef] [PubMed]
  13. Khan, A.; Fahl Mar, K.; Faucett, J.; Khan Schilling, S.; Brown, W.A. Has the Rising Placebo Response Impacted Antidepressant Clinical Trial Outcome? Data from the US Food and Drug Administration 1987–2013. World Psychiatry 2017, 16, 181–192. [Google Scholar] [CrossRef] [PubMed]
  14. Cipriani, A.; Furukawa, T.A.; Salanti, G.; Chaimani, A.; Atkinson, L.Z.; Ogawa, Y.; Leucht, S.; Ruhe, H.G.; Turner, E.H.; Higgins, J.P.T.; et al. Comparative Efficacy and Acceptability of 21 Antidepressant Drugs for the Acute Treatment of Adults with Major Depressive Disorder: A Systematic Review and Network Meta-Analysis. Lancet 2018, 391, 1357–1366. [Google Scholar] [CrossRef]
  15. Fabbri, C.; Corponi, F.; Souery, D.; Kasper, S.; Montgomery, S.; Zohar, J.; Rujescu, D.; Mendlewicz, J.; Serretti, A. The Genetics of Treatment-Resistant Depression: A Critical Review and Future Perspectives. Int. J. Neuropsychopharmacol. 2019, 22, 93–104. [Google Scholar] [CrossRef] [PubMed]
  16. Rush, A.J.; Trivedi, M.H.; Wisniewski, S.R.; Nierenberg, A.A.; Stewart, J.W.; Warden, D.; Niederehe, G.; Thase, M.E.; Lavori, P.W.; Lebowitz, B.D.; et al. Acute and Longer-Term Outcomes in Depressed Outpatients Requiring One or Several Treatment Steps: A STAR*D Report. Am. J. Psychiatry 2006, 163, 1905–1917. [Google Scholar] [CrossRef]
  17. Undurraga, J.; Baldessarini, R.J. Randomized, Placebo-Controlled Trials of Antidepressants for Acute Major Depression: Thirty-Year Meta-Analytic Review. Neuropsychopharmacology 2012, 37, 851–864. [Google Scholar] [CrossRef]
  18. Fava, M. Diagnosis and Definition of Treatment-Resistant Depression. Biol. Psychiatry 2003, 53, 649–659. [Google Scholar] [CrossRef] [PubMed]
  19. Sultana, J.; Cutroneo, P.; Trifirò, G. Clinical and Economic Burden of Adverse Drug Reactions. J. Pharmacol. Pharmacother. 2013, 4, S73–S77. [Google Scholar] [CrossRef] [PubMed]
  20. Eichelbaum, M.; Ingelman-Sundberg, M.; Evans, W.E. Pharmacogenomics and Individualized Drug Therapy. Annu. Rev. Med. 2006, 57, 119–137. [Google Scholar] [CrossRef]
  21. van Westrhenen, R.; van Schaik, R.H.N.; van Gelder, T.; Birkenhager, T.K.; Bakker, P.R.; Houwink, E.J.F.; Bet, P.M.; Hoogendijk, W.J.G.; van Weelden-Hulshof, M.J.M. Policy and Practice Review: A First Guideline on the Use of Pharmacogenetics in Clinical Psychiatric Practice. Front. Pharmacol. 2021, 12, 640032. [Google Scholar] [CrossRef]
  22. Ahmed, S.; Zhou, Z.; Zhou, J.; Chen, S.-Q. Pharmacogenomics of Drug Metabolizing Enzymes and Transporters: Relevance to Precision Medicine. Genom. Proteom. Bioinform. 2016, 14, 298–313. [Google Scholar] [CrossRef] [PubMed]
  23. Jukic, M.; Milosavljević, F.; Molden, E.; Ingelman-Sundberg, M. Pharmacogenomics in Treatment of Depression and Psychosis: An Update. Trends Pharmacol. Sci. 2022, 43, 1055–1069. [Google Scholar] [CrossRef]
  24. Singh, D.B. The Impact of Pharmacogenomics in Personalized Medicine. In Current Applications of Pharmaceutical Biotechnology; Springer: Berlin/Heidelberg, Germany, 2019; pp. 369–394. [Google Scholar]
  25. Sim, S.C.; Ingelman-Sundberg, M. The Human Cytochrome P450 (CYP) Allele Nomenclature Website: A Peer-Reviewed Database of CYP Variants and Their Associated Effects. Hum. Genom. 2010, 4, 278. [Google Scholar] [CrossRef]
  26. Zhou, S.-F. Polymorphism of Human Cytochrome P450 2D6 and Its Clinical Significance. Clin. Pharmacokinet. 2009, 48, 689–723. [Google Scholar] [CrossRef]
  27. Češková, E.; Šilhán, P. From Personalized Medicine to Precision Psychiatry? Neuropsychiatr. Dis. Treat. 2021, 17, 3663–3668. [Google Scholar] [CrossRef] [PubMed]
  28. Auwerx, C.; Sadler, M.C.; Reymond, A.; Kutalik, Z. From Pharmacogenetics to Pharmaco-Omics: Milestones and Future Directions. Human Genet. Genom. Adv. 2022, 3, 100100. [Google Scholar] [CrossRef] [PubMed]
  29. Evans, W.E.; McLeod, H.L. Pharmacogenomics—Drug Disposition, Drug Targets, and Side Effects. N. Engl. J. Med. 2003, 348, 538–549. [Google Scholar] [CrossRef]
  30. Bousman, C.A.; Bengesser, S.A.; Aitchison, K.J.; Amare, A.T.; Aschauer, H.; Baune, B.T.; Asl, B.B.; Bishop, J.R.; Burmeister, M.; Chaumette, B.; et al. Review and Consensus on Pharmacogenomic Testing in Psychiatry. Pharmacopsychiatry 2021, 54, 5–17. [Google Scholar] [CrossRef]
  31. Bousman, C.A.; Stevenson, J.M.; Ramsey, L.B.; Sangkuhl, K.; Hicks, J.K.; Strawn, J.R.; Singh, A.B.; Ruaño, G.; Mueller, D.J.; Tsermpini, E.E.; et al. Clinical Pharmacogenetics Implementation Consortium (CPIC) Guideline for CYP2D6, CYP2C19, CYP2B6, SLC6A4, and HTR2A Genotypes and Serotonin Reuptake Inhibitor Antidepressants. Clin. Pharmacol. Ther. 2023, 114, 51–68. [Google Scholar] [CrossRef] [PubMed]
  32. Hicks, J.; Sangkuhl, K.; Swen, J.; Ellingrod, V.; Müller, D.; Shimoda, K.; Bishop, J.; Kharasch, E.; Skaar, T.; Gaedigk, A.; et al. Clinical Pharmacogenetics Implementation Consortium Guideline (CPIC) for CYP2D6 and CYP2C19 Genotypes and Dosing of Tricyclic Antidepressants: 2016 Update. Clin. Pharmacol. Ther. 2017, 102, 37–44. [Google Scholar] [CrossRef]
  33. Brouwer, J.M.J.L.; Nijenhuis, M.; Soree, B.; Guchelaar, H.-J.; Swen, J.J.; van Schaik, R.H.N.; Weide, J.; van der Rongen, G.A.P.J.M.; Buunk, A.-M.; de Boer-Veger, N.J.; et al. Dutch Pharmacogenetics Working Group (DPWG) Guideline for the Gene-Drug Interaction between CYP2C19 and CYP2D6 and SSRIs. Eur. J. Human Genet. 2022, 30, 1114–1120. [Google Scholar] [CrossRef]
  34. Beunk, L.; Nijenhuis, M.; Soree, B.; de Boer-Veger, N.J.; Buunk, A.-M.; Guchelaar, H.J.; Houwink, E.J.F.; Risselada, A.; Rongen, G.A.P.J.M.; van Schaik, R.H.N.; et al. Dutch Pharmacogenetics Working Group (DPWG) Guideline for the Gene-Drug Interaction between CYP2D6, CYP3A4 and CYP1A2 and Antipsychotics. Eur. J. Human Genet. 2024, 32, 278–285. [Google Scholar] [CrossRef]
  35. Nijenhuis, M.; Soree, B.; Jama, W.O.M.; de Boer-Veger, N.J.; Buunk, A.M.; Guchelaar, H.-J.; Houwink, E.J.F.; Rongen, G.A.; van Schaik, R.H.N.; Swen, J.J.; et al. Dutch Pharmacogenetics Working Group (DPWG) Guideline for the Gene-Drug Interaction of CYP2D6 and COMT with Atomoxetine and Methylphenidate. Eur. J. Human Genet. 2023, 31, 1364–1370. [Google Scholar] [CrossRef] [PubMed]
  36. Brown, J.T.; Bishop, J.R.; Sangkuhl, K.; Nurmi, E.L.; Mueller, D.J.; Dinh, J.C.; Gaedigk, A.; Klein, T.E.; Caudle, K.E.; McCracken, J.T.; et al. Clinical Pharmacogenetics Implementation Consortium Guideline for Cytochrome P450 (CYP) 2D6 Genotype and Atomoxetine Therapy. Clin. Pharmacol. Ther. 2019, 106, 94–102. [Google Scholar] [CrossRef] [PubMed]
  37. Phillips, E.J.; Sukasem, C.; Whirl-Carrillo, M.; Müller, D.J.; Dunnenberger, H.M.; Chantratita, W.; Goldspiel, B.; Chen, Y.; Carleton, B.C.; George, A.L.; et al. Clinical Pharmacogenetics Implementation Consortium Guideline for HLA Genotype and Use of Carbamazepine and Oxcarbazepine: 2017 Update. Clin. Pharmacol. Ther. 2018, 103, 574–581. [Google Scholar] [CrossRef]
  38. ISPG. The International Society of Psychiatric Genetics; ISPG: Singapore, 2019. [Google Scholar]
  39. Sims, R.J.; Chen, C.-F.; Santos-Rosa, H.; Kouzarides, T.; Patel, S.S.; Reinberg, D. Human but Not Yeast CHD1 Binds Directly and Selectively to Histone H3 Methylated at Lysine 4 via Its Tandem Chromodomains. J. Biol. Chem. 2005, 280, 41789–41792. [Google Scholar] [CrossRef]
  40. Abdullah-Koolmees, H.; van Keulen, A.M.; Nijenhuis, M.; Deneer, V.H.M. Pharmacogenetics Guidelines: Overview and Comparison of the DPWG, CPIC, CPNDS, and RNPGx Guidelines. Front. Pharmacol. 2021, 11, 595219. [Google Scholar] [CrossRef] [PubMed]
  41. Caudle, K.; Klein, T.; Hoffman, J.; Muller, D.; Whirl-Carrillo, M.; Gong, L.; McDonagh, E.; Sangkuhl, K.; Thorn, C.; Schwab, M.; et al. Incorporation of Pharmacogenomics into Routine Clinical Practice: The Clinical Pharmacogenetics Implementation Consortium (CPIC) Guideline Development Process. Curr. Drug Metab. 2014, 15, 209–217. [Google Scholar] [CrossRef]
  42. Yoon, D.Y.; Lee, S.; Ban, M.S.; Jang, I.-J.; Lee, S. Pharmacogenomic Information from CPIC and DPWG Guidelines and Its Application on Drug Labels. Transl. Clin. Pharmacol. 2020, 28, 189. [Google Scholar] [CrossRef]
  43. Shekhani, R.; Steinacher, L.; Swen, J.J.; Ingelman-Sundberg, M. Evaluation of Current Regulation and Guidelines of Pharmacogenomic Drug Labels: Opportunities for Improvements. Clin. Pharmacol. Ther. 2020, 107, 1240–1255. [Google Scholar] [CrossRef]
  44. Stavropoulou, E.; Pircalabioru, G.G.; Bezirtzoglou, E. The Role of Cytochromes P450 in Infection. Front. Immunol. 2018, 9, 89. [Google Scholar] [CrossRef]
  45. Guengerich, F.P.; Waterman, M.R.; Egli, M. Recent Structural Insights into Cytochrome P450 Function. Trends Pharmacol. Sci. 2016, 37, 625–640. [Google Scholar] [CrossRef] [PubMed]
  46. Riaz, S.; Muhammad Din, S.; Usman Tareen, M.; Tariq, F.; Latif, Y.; Siddiqi, S.; Sultan, A.; Mansoor, A. Genetic Polymorphism of CYP2C19 in Pakistani Population. Iran. J. Pharm. Res. 2019, 18, 1097–1102. [Google Scholar] [CrossRef] [PubMed]
  47. Foster, A.; Mobley, E.; Wang, Z. Complicated Pain Management in a CYP450 2D6 Poor Metabolizer. Pain. Pract. 2007, 7, 352–356. [Google Scholar] [CrossRef]
  48. Zanger, U.M.; Schwab, M. Cytochrome P450 Enzymes in Drug Metabolism: Regulation of Gene Expression, Enzyme Activities, and Impact of Genetic Variation. Pharmacol. Ther. 2013, 138, 103–141. [Google Scholar] [CrossRef]
  49. Taylor, C.; Crosby, I.; Yip, V.; Maguire, P.; Pirmohamed, M.; Turner, R.M. A Review of the Important Role of CYP2D6 in Pharmacogenomics. Genes 2020, 11, 1295. [Google Scholar] [CrossRef] [PubMed]
  50. Kendrick, T.; Taylor, D.; Johnson, C.F. Which First-Line Antidepressant? Br. J. General. Pract. 2019, 69, 114–115. [Google Scholar] [CrossRef]
  51. Darby-Stewart, A.; Dachs, R.J.; Graber, M.A. Antidepressants for Initial Treatment of Depression. Am. Fam. Physician 2010, 81, 1205. [Google Scholar] [PubMed]
  52. Koenig, A.M.; Thase, M.E. First-Line Pharmacotherapies for Depression—What Is the Best Choice? Pol. Arch. Med. Wewn. 2009, 119, 478–486. [Google Scholar] [CrossRef] [PubMed]
  53. Edinoff, A.N.; Akuly, H.A.; Hanna, T.A.; Ochoa, C.O.; Patti, S.J.; Ghaffar, Y.A.; Kaye, A.D.; Viswanath, O.; Urits, I.; Boyer, A.G.; et al. Selective Serotonin Reuptake Inhibitors and Adverse Effects: A Narrative Review. Neurol. Int. 2021, 13, 387–401. [Google Scholar] [CrossRef]
  54. Cacabelos, R.; Torrellas, C. Epigenetics of Aging and Alzheimer’s Disease: Implications for Pharmacogenomics and Drug Response. Int. J. Mol. Sci. 2015, 16, 30483–30543. [Google Scholar] [CrossRef]
  55. Brøsen, K. Differences in Interactions of SSRIs. Int. Clin. Psychopharmacol. 1998, 13, S45–S48. [Google Scholar] [CrossRef] [PubMed]
  56. Vo, N.; Goodman, R.H. CREB-Binding Protein and P300 in Transcriptional Regulation. J. Biol. Chem. 2001, 276, 13505–13508. [Google Scholar] [CrossRef] [PubMed]
  57. Fricke-Galindo, I.; Céspedes-Garro, C.; Rodrigues-Soares, F.; Naranjo, M.E.G.; Delgado, Á.; de Andrés, F.; López-López, M.; Peñas-Lledó, E.; LLerena, A. Interethnic Variation of CYP2C19 Alleles, ‘Predicted’ Phenotypes and ‘Measured’ Metabolic Phenotypes across World Populations. Pharmacogenomics J. 2016, 16, 113–123. [Google Scholar] [CrossRef]
  58. Hicks, J.; Bishop, J.; Sangkuhl, K.; Müller, D.; Ji, Y.; Leckband, S.; Leeder, J.; Graham, R.; Chiulli, D.; LLerena, A.; et al. Clinical Pharmacogenetics Implementation Consortium (CPIC) Guideline for CYP2D6 and CYP2C19 Genotypes and Dosing of Selective Serotonin Reuptake Inhibitors. Clin. Pharmacol. Ther. 2015, 98, 127–134. [Google Scholar] [CrossRef]
  59. Whirl-Carrillo, M.; Huddart, R.; Gong, L.; Sangkuhl, K.; Thorn, C.F.; Whaley, R.; Klein, T.E. An Evidence-Based Framework for Evaluating Pharmacogenomics Knowledge for Personalized Medicine. Clin. Pharmacol. 2021, 110, 563–572. [Google Scholar] [CrossRef]
  60. Schenk, P.W.; van Vliet, M.; Mathot, R.A.A.; van Gelder, T.; Vulto, A.G.; van Fessem, M.A.C.; Verploegh-Van Rij, S.; Lindemans, J.; Bruijn, J.A.; van Schaik, R.H.N. The CYP2C19*17 Genotype Is Associated with Lower Imipramine Plasma Concentrations in a Large Group of Depressed Patients. Pharmacogenomics J. 2010, 10, 219–225. [Google Scholar] [CrossRef]
  61. Kirchheiner, J.; Nickchen, K.; Bauer, M.; Wong, M.-L.; Licinio, J.; Roots, I.; Brockmöller, J. Pharmacogenetics of Antidepressants and Antipsychotics: The Contribution of Allelic Variations to the Phenotype of Drug Response. Mol. Psychiatry 2004, 9, 442–473. [Google Scholar] [CrossRef]
  62. Fanelli, D.; Weller, G.; Liu, H. New Serotonin-Norepinephrine Reuptake Inhibitors and Their Anesthetic and Analgesic Considerations. Neurol. Int. 2021, 13, 497–509. [Google Scholar] [CrossRef] [PubMed]
  63. Hillhouse, T.M.; Porter, J.H. A Brief History of the Development of Antidepressant Drugs: From Monoamines to Glutamate. Exp. Clin. Psychopharmacol. 2015, 23, 1–21. [Google Scholar] [CrossRef] [PubMed]
  64. Bruno, A.; Morabito, P.; Spina, E.; Rosaria Muscatello, M. The Role of Levomilnacipran in the Management of Major Depressive Disorder: A Comprehensive Review. Curr. Neuropharmacol. 2016, 14, 191–199. [Google Scholar] [CrossRef]
  65. Eap, C.B.; Gründer, G.; Baumann, P.; Ansermot, N.; Conca, A.; Corruble, E.; Crettol, S.; Dahl, M.L.; de Leon, J.; Greiner, C.; et al. Tools for Optimising Pharmacotherapy in Psychiatry (Therapeutic Drug Monitoring, Molecular Brain Imaging and Pharmacogenetic Tests): Focus on Antidepressants. World J. Biol. Psychiatry 2021, 22, 561–628. [Google Scholar] [CrossRef]
  66. Puozzo, C.; Panconi, E.; Deprez, D. Pharmacology and Pharmacokinetics of Milnacipran. Int. Clin. Psychopharmacol. 2002, 17, S25–S35. [Google Scholar] [CrossRef] [PubMed]
  67. Khalid, M.M.; Waseem, M. Tricyclic Antidepressant Toxicity; StatPearls: St. Petersburg, FL, USA, 2023. [Google Scholar]
  68. Laura Dean, M.D. Imipramine Therapy and CYP2D6 and CYP2C19 Genotype. In Medical Genetics Summaries; NIH: Bethesda, MD, USA, 2017. [Google Scholar]
  69. Anttila, S.A.K.; Leinonen, E.V.J. A Review of the Pharmacological and Clinical Profile of Mirtazapine. CNS Drug Rev. 2001, 7, 249–264. [Google Scholar] [CrossRef]
  70. Langmia, I.M.; Just, K.S.; Yamoune, S.; Müller, J.P.; Stingl, J.C. Pharmacogenetic and Drug Interaction Aspects on Ketamine Safety in Its Use as Antidepressant—Implications for Precision Dosing in a Global Perspective. Br. J. Clin. Pharmacol. 2022, 88, 5149–5165. [Google Scholar] [CrossRef] [PubMed]
  71. Lally, J.; MacCabe, J.H. Antipsychotic Medication in Schizophrenia: A Review. Br. Med. Bull. 2015, 114, 169–179. [Google Scholar] [CrossRef]
  72. Altar, C.A.; Hornberger, J.; Shewade, A.; Cruz, V.; Garrison, J.; Mrazek, D. Clinical Validity of Cytochrome P450 Metaboliand Serotonin Gene Variants in Psychiatric Pharmacotherapy. Int. Rev. Psychiatry 2013, 25, 509–533. [Google Scholar] [CrossRef]
  73. Walden, L.M.; Brandl, E.J.; Tiwari, A.K.; Cheema, S.; Freeman, N.; Braganza, N.; Kennedy, J.L.; Müller, D.J. Genetic Testing for CYP2D6 and CYP2C19 Suggests Improved Outcome for Antidepressant and Antipsychotic Medication. Psychiatry Res. 2019, 279, 111–115. [Google Scholar] [CrossRef]
  74. Ravyn, D.; Ravyn, V.; Lowney, R.; Nasrallah, H.A. CYP450 Pharmacogenetic Treatment Strategies for Antipsychotics: A Review of the Evidence. Schizophr. Res. 2013, 149, 1–14. [Google Scholar] [CrossRef] [PubMed]
  75. Spina, E.; de Leon, J. Clinical Applications of CYP Genotyping in Psychiatry. J. Neural Transm. 2015, 122, 5–28. [Google Scholar] [CrossRef] [PubMed]
  76. Hoffmann, M.F.; Preissner, S.C.; Nickel, J.; Dunkel, M.; Preissner, R.; Preissner, S. The Transformer Database: Biotransformation of Xenobiotics. Nucleic Acids Res. 2014, 42, D1113–D1117. [Google Scholar] [CrossRef] [PubMed]
  77. Carrascal-Laso, L.; Isidoro-García, M.; Ramos-Gallego, I.; Franco-Martín, M. Review: Influence of the CYP450 Genetic Variation on the Treatment of Psychotic Disorders. J. Clin. Med. 2021, 10, 4275. [Google Scholar] [CrossRef]
  78. Brandl, E.J.; Chowdhury, N.I.; Tiwari, A.K.; Lett, T.A.P.; Meltzer, H.Y.; Kennedy, J.L.; Müller, D.J. Genetic Variation in CYP3A43 Is Associated with Response to Antipsychotic Medication. J. Neural Transm. 2015, 122, 29–34. [Google Scholar] [CrossRef]
  79. Kim, K.-A.; Joo, H.-J.; Lee, H.-M.; Park, J.-Y. Influence of ABCB1 and CYP3A5 Genetic Polymorphisms on the Pharmacokinetics of Quetiapine in Healthy Volunteers. Pharmacogenet Genom. 2014, 24, 35–42. [Google Scholar] [CrossRef] [PubMed]
  80. Belmonte, C.; Ochoa, D.; Román, M.; Saiz-Rodríguez, M.; Wojnicz, A.; Gómez-Sánchez, C.I.; Martín-Vílchez, S.; Abad-Santos, F. Influence of CYP2D6,CYP3A4,CYP3A5 and ABCB1 Polymorphisms on Pharmacokinetics and Safety of Aripiprazole in Healthy Volunteers. Basic Clin. Pharmacol. Toxicol. 2018, 122, 596–605. [Google Scholar] [CrossRef] [PubMed]
  81. Soranzo, N.; Cavalleri, G.L.; Weale, M.E.; Wood, N.W.; Depondt, C.; Marguerie, R.; Sisodiya, S.M.; Goldstein, D.B. Identifying Candidate Causal Variants Responsible for Altered Activity of the ABCB1 Multidrug Resistance Gene. Genome Res. 2004, 14, 1333–1344. [Google Scholar] [CrossRef]
  82. Suzuki, Y.; Tsuneyama, N.; Fukui, N.; Sugai, T.; Watanabe, J.; Ono, S.; Saito, M.; Someya, T. Impact of the ABCB1 Gene Polymorphism on Plasma 9-Hydroxyrisperidone and Active Moiety Levels in Japanese Patients with Schizophrenia. J. Clin. Psychopharmacol. 2013, 33, 411–414. [Google Scholar] [CrossRef] [PubMed]
  83. Hoffmeyer, S.; Burk, O.; von Richter, O.; Arnold, H.P.; Brockmöller, J.; Johne, A.; Cascorbi, I.; Gerloff, T.; Roots, I.; Eichelbaum, M.; et al. Functional Polymorphisms of the Human Multidrug-Resistance Gene: Multiple Sequence Variations and Correlation of One Allele with P-Glycoprotein Expression and Activity in vivo. Proc. Natl. Acad. Sci. USA 2000, 97, 3473–3478. [Google Scholar] [CrossRef] [PubMed]
  84. Hodgson, K.; Tansey, K.; Dernovšek, M.Z.; Hauser, J.; Henigsberg, N.; Maier, W.; Mors, O.; Placentino, A.; Rietschel, M.; Souery, D.; et al. Genetic Differences in Cytochrome P450 Enzymes and Antidepressant Treatment Response. J. Psychopharmacol. 2014, 28, 133–141. [Google Scholar] [CrossRef] [PubMed]
  85. Austin-Zimmerman, I.; Wronska, M.; Wang, B.; Irizar, H.; Thygesen, J.H.; Bhat, A.; Denaxas, S.; Fatemifar, G.; Finan, C.; Harju-Seppänen, J.; et al. The Influence of CYP2D6 and CYP2C19 Genetic Variation on Diabetes Mellitus Risk in People Taking Antidepressants and Antipsychotics. Genes 2021, 12, 1758. [Google Scholar] [CrossRef]
  86. Young, R.M.; Lawford, B.R.; Barnes, M.; Burton, S.C.; Ritchie, T.; Ward, W.K.; Noble, E.P. Prolactin Levels in Antipsychotic Treatment of Patients with Schizophrenia Carrying the DRD2*A1 Allele. Br. J. Psychiatry 2004, 185, 147–151. [Google Scholar] [CrossRef]
  87. Shad, M.U. Genetic Testing for Antipsychotic Pharmacotherapy: Bench to Bedside. Behav. Sci. 2021, 11, 97. [Google Scholar] [CrossRef] [PubMed]
  88. Nath, M.; Gupta, V. Mood Stabilizers; StatPearls: St. Petersburg, FL, USA, 2023. [Google Scholar]
  89. Iannaccone, T.; Sellitto, C.; Manzo, V.; Colucci, F.; Giudice, V.; Stefanelli, B.; Iuliano, A.; Corrivetti, G.; Filippelli, A. Pharmacogenetics of Carbamazepine and Valproate: Focus on Polymorphisms of Drug Metabolizing Enzymes and Transporters. Pharmaceuticals 2021, 14, 204. [Google Scholar] [CrossRef] [PubMed]
  90. Wang, P.; Yin, T.; Ma, H.-Y.; Liu, D.-Q.; Sheng, Y.-H.; Wang, C.; Zhou, B.-T. Effects of CYP3A4/5 and ABCB1 Genetic Polymorphisms on Carbamazepine Metabolism and Transport in Chinese Patients with Epilepsy Treated with Carbamazepine in Monotherapy and Bitherapy. Epilepsy Res. 2015, 117, 52–57. [Google Scholar] [CrossRef]
  91. Panomvana, D.; Traiyawong, T.; Towanabut, S. Effect of CYP3A5 Genotypes on the Pharmacokinetics of Carbamazepine When Used as Monotherapy or Co-Administered with Phenytoin, Phenobarbital or Valproic Acid in Thai Patients. J. Pharm. Pharm. Sci. 2013, 16, 502. [Google Scholar] [CrossRef] [PubMed]
  92. Saiz-Rodríguez, M.; Almenara, S.; Navares-Gómez, M.; Ochoa, D.; Román, M.; Zubiaur, P.; Koller, D.; Santos, M.; Mejía, G.; Borobia, A.M.; et al. Effect of the Most Relevant CYP3A4 and CYP3A5 Polymorphisms on the Pharmacokinetic Parameters of 10 CYP3A Substrates. Biomedicines 2020, 8, 94. [Google Scholar] [CrossRef] [PubMed]
  93. Franco, V.; Perucca, E. The Pharmacogenomics of Epilepsy. Expert. Rev. Neurother. 2015, 15, 1161–1170. [Google Scholar] [CrossRef] [PubMed]
  94. Paola Cruz-Tapias, J.C.; Anaya, J.-M. Major Histocompatibility Complex: Antigen Processing and Presentation. In Autoimmunity: From Bench to Bedside; NIH: Bethesda, MD, USA, 2013. [Google Scholar]
  95. Nudel, R.; Benros, M.E.; Krebs, M.D.; Allesøe, R.L.; Lemvigh, C.K.; Bybjerg-Grauholm, J.; Børglum, A.D.; Daly, M.J.; Nordentoft, M.; Mors, O.; et al. Immunity and Mental Illness: Findings from a Danish Population-Based Immunogenetic Study of Seven Psychiatric and Neurodevelopmental Disorders. Eur. J. Human Genet. 2019, 27, 1445–1455. [Google Scholar] [CrossRef] [PubMed]
  96. Tamouza, R.; Krishnamoorthy, R.; Leboyer, M. Understanding the Genetic Contribution of the Human Leukocyte Antigen System to Common Major Psychiatric Disorders in a World Pandemic Context. Brain Behav. Immun. 2021, 91, 731–739. [Google Scholar] [CrossRef] [PubMed]
  97. Boulanger, L.M. Immune Proteins in Brain Development and Synaptic Plasticity. Neuron 2009, 64, 93–109. [Google Scholar] [CrossRef] [PubMed]
  98. JJeiziner, C.; Wernli, U.; Suter, K.; Hersberger, K.E.; zu Schwabedissen, H.E.M. HLA-associated Adverse Drug Reactions—Scoping Review. Clin. Transl. Sci. 2021, 14, 1648–1658. [Google Scholar] [CrossRef]
  99. Le Clerc, S.; Taing, L.; Fond, G.; Meary, A.; Llorca, P.-M.; Blanc, O.; Beaune, P.; Rajagopal, K.; Jamain, S.; Tamouza, R.; et al. A Double Amino-Acid Change in the HLA-A Peptide-Binding Groove Is Associated with Response to Psychotropic Treatment in Patients with Schizophrenia. Transl. Psychiatry 2015, 5, e608. [Google Scholar] [CrossRef]
  100. Yang, S.-C.; Chen, C.-B.; Lin, M.-Y.; Zhang, Z.-Y.; Jia, X.-Y.; Huang, M.; Zou, Y.-F.; Chung, W.-H. Genetics of Severe Cutaneous Adverse Reactions. Front. Med. 2021, 8, 652091. [Google Scholar] [CrossRef] [PubMed]
  101. Amstutz, U.; Ross, C.J.D.; Castro-Pastrana, L.I.; Rieder, M.J.; Shear, N.H.; Hayden, M.R.; Carleton, B.C. HLA-A*31:01 and HLA-B*15:02 as Genetic Markers for Carbamazepine Hypersensitivity in Children. Clin. Pharmacol. Ther. 2013, 94, 142–149. [Google Scholar] [CrossRef]
  102. Wallace, M.L.; Frank, E.; Kraemer, H.C. A Novel Approach for Developing and Interpreting Treatment Moderator Profiles in Randomized Clinical Trials. JAMA Psychiatry 2013, 70, 1241. [Google Scholar] [CrossRef] [PubMed]
  103. Shiina, T.; Hosomichi, K.; Inoko, H.; Kulski, J.K. The HLA Genomic Loci Map: Expression, Interaction, Diversity and Disease. J. Hum. Genet. 2009, 54, 15–39. [Google Scholar] [CrossRef]
  104. Pang, H.; Jia, W.; Hu, Z. Emerging Applications of Metabolomics in Clinical Pharmacology. Clin. Pharmacol. Ther. 2019, 106, 544–556. [Google Scholar] [CrossRef]
  105. Andrew Clayton, T.; Lindon, J.C.; Cloarec, O.; Antti, H.; Charuel, C.; Hanton, G.; Provost, J.-P.; Le Net, J.-L.; Baker, D.; Walley, R.J.; et al. Pharmaco-Metabonomic Phenotyping and Personalized Drug Treatment. Nature 2006, 440, 1073–1077. [Google Scholar] [CrossRef]
  106. Ji, Y.; Schaid, D.J.; Desta, Z.; Kubo, M.; Batzler, A.J.; Snyder, K.; Mushiroda, T.; Kamatani, N.; Ogburn, E.; Hall-Flavin, D.; et al. Citalopram and Escitalopram Plasma Drug and Metabolite Concentrations: Genome-wide Associations. Br. J. Clin. Pharmacol. 2014, 78, 373–383. [Google Scholar] [CrossRef] [PubMed]
  107. Neavin, D.; Kaddurah-Daouk, R.; Weinshilboum, R. Pharmacometabolomics Informs Pharmacogenomics. Metabolomics 2016, 12, 121. [Google Scholar] [CrossRef] [PubMed]
  108. Lander, E.S.; Linton, L.M.; Birren, B.; Nusbaum, C.; Zody, M.C.; Baldwin, J.; Devon, K.; Dewar, K.; Doyle, M.; FitzHugh, W.; et al. Initial Sequencing and Analysis of the Human Genome. Nature 2001, 409, 860–921. [Google Scholar] [CrossRef] [PubMed]
  109. Gupta, M.; Neavin, D.; Liu, D.; Biernacka, J.; Hall-Flavin, D.; Bobo, W.V.; Frye, M.A.; Skime, M.; Jenkins, G.D.; Batzler, A.; et al. TSPAN5, ERICH3 and Selective Serotonin Reuptake Inhibitors in Major Depressive Disorder: Pharmacometabolomics-Informed Pharmacogenomics. Mol. Psychiatry 2016, 21, 1717–1725. [Google Scholar] [CrossRef] [PubMed]
  110. Zhong, H.; Hansen, K.B.; Boyle, N.J.; Han, K.; Muske, G.; Huang, X.; Egebjerg, J.; Sánchez, C. An Allosteric Binding Site at the Human Serotonin Transporter Mediates the Inhibition of Escitalopram by R-Citalopram: Kinetic Binding Studies with the ALI/VFL–SI/TT Mutant. Neurosci. Lett. 2009, 462, 207–212. [Google Scholar] [CrossRef] [PubMed]
  111. Jacobsen, J.P.R.; Plenge, P.; Sachs, B.D.; Pehrson, A.L.; Cajina, M.; Du, Y.; Roberts, W.; Rudder, M.L.; Dalvi, P.; Robinson, T.J.; et al. The Interaction of Escitalopram and R-Citalopram at the Human Serotonin Transporter Investigated in the Mouse. Psychopharmacology 2014, 231, 4527–4540. [Google Scholar] [CrossRef] [PubMed]
  112. Cavaleri, D.; Riboldi, I.; Crocamo, C.; Paglia, G.; Carrà, G.; Bartoli, F. Evidence from Preclinical and Clinical Metabolomics Studies on the Antidepressant Effects of Ketamine and Esketamine. Neurosci. Lett. 2024, 831, 137791. [Google Scholar] [CrossRef] [PubMed]
  113. Burghardt, K.J.; Kajy, M.; Ward, K.M.; Burghardt, P.R. Metabolomics, Lipidomics, and Antipsychotics: A Systematic Review. Biomedicines 2023, 11, 3295. [Google Scholar] [CrossRef] [PubMed]
  114. de Bartolomeis, A.; Vellucci, L.; Austin, M.C.; De Simone, G.; Barone, A. Rational and Translational Implications of D-Amino Acids for Treatment-Resistant Schizophrenia: From Neurobiology to the Clinics. Biomolecules 2022, 12, 909. [Google Scholar] [CrossRef]
  115. Labrie, V.; Lipina, T.; Roder, J.C. Mice with Reduced NMDA Receptor Glycine Affinity Model Some of the Negative and Cognitive Symptoms of Schizophrenia. Psychopharmacology 2008, 200, 217–230. [Google Scholar] [CrossRef] [PubMed]
  116. Effinger, D.; Hirschberger, S.; Yoncheva, P.; Schmid, A.; Heine, T.; Newels, P.; Schütz, B.; Meng, C.; Gigl, M.; Kleigrewe, K.; et al. A Ketogenic Diet Substantially Reshapes the Human Metabolome. Clin. Nutr. 2023, 42, 1202–1212. [Google Scholar] [CrossRef]
  117. Yurista, S.R.; Chong, C.-R.; Badimon, J.J.; Kelly, D.P.; de Boer, R.A.; Westenbrink, B.D. Therapeutic Potential of Ketone Bodies for Patients With Cardiovascular Disease. J. Am. Coll. Cardiol. 2021, 77, 1660–1669. [Google Scholar] [CrossRef] [PubMed]
  118. O’Donnell, C.P.; Allott, K.A.; Murphy, B.P.; Yuen, H.P.; Proffitt, T.-M.; Papas, A.; Moral, J.; Pham, T.; O’Regan, M.K.; Phassouliotis, C.; et al. Adjunctive Taurine in First-Episode Psychosis. J. Clin. Psychiatry 2016, 77, e1610–e1617. [Google Scholar] [CrossRef] [PubMed]
  119. Canyelles, M.; Borràs, C.; Rotllan, N.; Tondo, M.; Escolà-Gil, J.C.; Blanco-Vaca, F. Gut Microbiota-Derived TMAO: A Causal Factor Promoting Atherosclerotic Cardiovascular Disease? Int. J. Mol. Sci. 2023, 24, 1940. [Google Scholar] [CrossRef] [PubMed]
  120. Hoyles, L.; Pontifex, M.G.; Rodriguez-Ramiro, I.; Anis-Alavi, M.A.; Jelane, K.S.; Snelling, T.; Solito, E.; Fonseca, S.; Carvalho, A.L.; Carding, S.R.; et al. Regulation of Blood–Brain Barrier Integrity by Microbiome-Associated Methylamines and Cognition by Trimethylamine N-Oxide. Microbiome 2021, 9, 235. [Google Scholar] [CrossRef]
  121. Schooneman, M.G.; Vaz, F.M.; Houten, S.M.; Soeters, M.R. Acylcarnitines: Reflecting or inflicting insulin resistance? Diabetes 2013, 62, 1–8. [Google Scholar] [CrossRef]
  122. McCoin, C.S.; Knotts, T.A.; Adams, S.H. Acylcarnitines—Old Actors Auditioning for New Roles in Metabolic Physiology. Nat. Rev. Endocrinol. 2015, 11, 617–625. [Google Scholar] [CrossRef] [PubMed]
  123. Dambrova, M.; Makrecka-Kuka, M.; Kuka, J.; Vilskersts, R.; Nordberg, D.; Attwood, M.M.; Smesny, S.; Sen, Z.D.; Guo, A.C.; Oler, E.; et al. Acylcarnitines: Nomenclature, Biomarkers, Therapeutic Potential, Drug Targets, and Clinical Trials. Pharmacol. Rev. 2022, 74, 506–551. [Google Scholar] [CrossRef] [PubMed]
  124. Li, S.; Gao, D.; Jiang, Y. Function, Detection and Alteration of Acylcarnitine Metabolism in Hepatocellular Carcinoma. Metabolites 2019, 9, 36. [Google Scholar] [CrossRef] [PubMed]
  125. Rutkowsky, J.M.; Knotts, T.A.; Ono-Moore, K.D.; McCoin, C.S.; Huang, S.; Schneider, D.; Singh, S.; Adams, S.H.; Hwang, D.H. Acylcarnitines Activate Proinflammatory Signaling Pathways. Am. J. Physiol.-Endocrinol. Metab. 2014, 306, E1378–E1387. [Google Scholar] [CrossRef]
  126. Cai, H.-L.; Li, H.-D.; Yan, X.-Z.; Sun, B.; Zhang, Q.; Yan, M.; Zhang, W.-Y.; Jiang, P.; Zhu, R.-H.; Liu, Y.-P.; et al. Metabolomic Analysis of Biochemical Changes in the Plasma and Urine of First-Episode Neuroleptic-Naïve Schizophrenia Patients after Treatment with Risperidone. J. Proteome Res. 2012, 11, 4338–4350. [Google Scholar] [CrossRef]
  127. Liu, D.; An, Z.; Li, P.; Chen, Y.; Zhang, R.; Liu, L.; He, J.; Abliz, Z. A Targeted Neurotransmitter Quantification and Nontargeted Metabolic Profiling Method for Pharmacometabolomics Analysis of Olanzapine by Using UPLC-HRMS. RSC Adv. 2020, 10, 18305–18314. [Google Scholar] [CrossRef]
  128. Atagün, M.İ.; Şıkoğlu, E.M.; Can, S.S.; Uğurlu, G.K.; Kaymak, S.U.; Çayköylü, A.; Algın, O.; Phillips, M.L.; Moore, C.M.; Öngür, D. Neurochemical Differences between Bipolar Disorder Type I and II in Superior Temporal Cortices: A Proton Magnetic Resonance Spectroscopy Study. J. Affect. Disord. 2018, 235, 15–19. [Google Scholar] [CrossRef] [PubMed]
  129. Kraus, C.; Kadriu, B.; Lanzenberger, R.; Zarate, C.A.; Kasper, S. Prognosis and Improved Outcomes in Major Depression: A Review. Transl. Psychiatry 2019, 9, 127. [Google Scholar] [CrossRef]
  130. Le-Niculescu, H.; Roseberry, K.; Gill, S.S.; Levey, D.F.; Phalen, P.L.; Mullen, J.; Williams, A.; Bhairo, S.; Voegtline, T.; Davis, H.; et al. Precision Medicine for Mood Disorders: Objective Assessment, Risk Prediction, Pharmacogenomics, and Repurposed Drugs. Mol. Psychiatry 2021, 26, 2776–2804. [Google Scholar] [CrossRef] [PubMed]
  131. Cook, I.A.; Congdon, E.; Krantz, D.E.; Hunter, A.M.; Coppola, G.; Hamilton, S.P.; Leuchter, A.F. Time Course of Changes in Peripheral Blood Gene Expression During Medication Treatment for Major Depressive Disorder. Front. Genet. 2019, 10, 870. [Google Scholar] [CrossRef] [PubMed]
  132. Mangiavacchi, A.; Morelli, G.; Orlando, V. Behind the Scenes: How RNA Orchestrates the Epigenetic Regulation of Gene Expression. Front. Cell Dev. Biol. 2023, 11, 1123975. [Google Scholar] [CrossRef]
  133. Mariani, N.; Cattane, N.; Pariante, C.; Cattaneo, A. Gene Expression Studies in Depression Development and Treatment: An Overview of the Underlying Molecular Mechanisms and Biological Processes to Identify Biomarkers. Transl. Psychiatry 2021, 11, 354. [Google Scholar] [CrossRef]
  134. Carrillo-Roa, T.; Labermaier, C.; Weber, P.; Herzog, D.P.; Lareau, C.; Santarelli, S.; Wagner, K.V.; Rex-Haffner, M.; Harbich, D.; Scharf, S.H.; et al. Common Genes Associated with Antidepressant Response in Mouse and Man Identify Key Role of Glucocorticoid Receptor Sensitivity. PLoS Biol. 2017, 15, e2002690. [Google Scholar] [CrossRef] [PubMed]
  135. Wang, Q.; Timberlake, M.A.; Prall, K.; Dwivedi, Y. The Recent Progress in Animal Models of Depression. Prog. Neuropsychopharmacol. Biol. Psychiatry 2017, 77, 99–109. [Google Scholar] [CrossRef] [PubMed]
  136. Baker, M.; Hong, S.-I.; Kang, S.; Choi, D.-S. Rodent Models for Psychiatric Disorders: Problems and Promises. Lab. Anim. Res. 2020, 36, 9. [Google Scholar] [CrossRef] [PubMed]
  137. Krishnan, V.; Nestler, E.J. Animal Models of Depression: Molecular Perspectives. In Molecular and Functional Models in Neuropsychiatry; Springer: Berlin/Heidelberg, Germany, 2011; pp. 121–147. [Google Scholar]
  138. Dudek, K.A.; Dion-Albert, L.; Kaufmann, F.N.; Tuck, E.; Lebel, M.; Menard, C. Neurobiology of Resilience in Depression: Immune and Vascular Insights from Human and Animal Studies. Eur. J. Neurosci. 2021, 53, 183–221. [Google Scholar] [CrossRef]
  139. Lorsch, Z.S.; Hamilton, P.J.; Ramakrishnan, A.; Parise, E.M.; Salery, M.; Wright, W.J.; Lepack, A.E.; Mews, P.; Issler, O.; McKenzie, A.; et al. Stress Resilience Is Promoted by a Zfp189-Driven Transcriptional Network in Prefrontal Cortex. Nat. Neurosci. 2019, 22, 1413–1423. [Google Scholar] [CrossRef]
  140. Cathomas, F.; Murrough, J.W.; Nestler, E.J.; Han, M.-H.; Russo, S.J. Neurobiology of Resilience: Interface Between Mind and Body. Biol. Psychiatry 2019, 86, 410–420. [Google Scholar] [CrossRef]
  141. Scarpa, J.R.; Fatma, M.; Loh, Y.-H.E.; Traore, S.R.; Stefan, T.; Chen, T.H.; Nestler, E.J.; Labonté, B. Shared Transcriptional Signatures in Major Depressive Disorder and Mouse Chronic Stress Models. Biol. Psychiatry 2020, 88, 159–168. [Google Scholar] [CrossRef]
  142. Hervé, M.; Bergon, A.; Le Guisquet, A.-M.; Leman, S.; Consoloni, J.-L.; Fernandez-Nunez, N.; Lefebvre, M.-N.; El-Hage, W.; Belzeaux, R.; Belzung, C.; et al. Translational Identification of Transcriptional Signatures of Major Depression and Antidepressant Response. Front. Mol. Neurosci. 2017, 10, 248. [Google Scholar] [CrossRef]
  143. Ramos-da-Silva, L.; Carlson, P.T.; Silva-Costa, L.C.; Martins-de-Souza, D.; de Almeida, V. Molecular Mechanisms Associated with Antidepressant Treatment on Major Depression. Complex. Psychiatry 2021, 7, 49–59. [Google Scholar] [CrossRef]
  144. Kumar, S.; Gal, Z.; Gonda, X.; Huse, R.J.; Juhasz, G.; Bagdy, G.; Petschner, P. Transcriptomic Changes Following Chronic Administration of Selective Serotonin Reuptake Inhibitors: A Review of Animal Studies. Neuropsychopharmacol. Hung. 2019, 21, 26–35. [Google Scholar]
  145. Gandal, M.J.; Zhang, P.; Hadjimichael, E.; Walker, R.L.; Chen, C.; Liu, S.; Won, H.; van Bakel, H.; Varghese, M.; Wang, Y.; et al. Transcriptome-Wide Isoform-Level Dysregulation in ASD, Schizophrenia, and Bipolar Disorder. Science 2018, 362, eaat8127. [Google Scholar] [CrossRef]
  146. Fromer, M.; Roussos, P.; Sieberts, S.K.; Johnson, J.S.; Kavanagh, D.H.; Perumal, T.M.; Ruderfer, D.M.; Oh, E.C.; Topol, A.; Shah, H.R.; et al. Gene Expression Elucidates Functional Impact of Polygenic Risk for Schizophrenia. Nat. Neurosci. 2016, 19, 1442–1453. [Google Scholar] [CrossRef] [PubMed]
  147. Wu, X.; Shukla, R.; Alganem, K.; Zhang, X.; Eby, H.M.; Devine, E.A.; Depasquale, E.; Reigle, J.; Simmons, M.; Hahn, M.K.; et al. Transcriptional Profile of Pyramidal Neurons in Chronic Schizophrenia Reveals Lamina-Specific Dysfunction of Neuronal Immunity. Mol. Psychiatry 2021, 26, 7699–7708. [Google Scholar] [CrossRef]
  148. Perzel Mandell, K.A.; Eagles, N.J.; Deep-Soboslay, A.; Tao, R.; Han, S.; Wilton, R.; Szalay, A.S.; Hyde, T.M.; Kleinman, J.E.; Jaffe, A.E.; et al. Molecular Phenotypes Associated with Antipsychotic Drugs in the Human Caudate Nucleus. Mol. Psychiatry 2022, 27, 2061–2067. [Google Scholar] [CrossRef] [PubMed]
  149. Martin, M.V.; Mirnics, K.; Nisenbaum, L.K.; Vawter, M.P. Olanzapine Reversed Brain Gene Expression Changes Induced by Phencyclidine Treatment in Non-Human Primates. Complex. Psychiatry 2015, 1, 82–93. [Google Scholar] [CrossRef] [PubMed]
  150. Chan, M.K.; Tsang, T.M.; Harris, L.W.; Guest, P.C.; Holmes, E.; Bahn, S. Evidence for Disease and Antipsychotic Medication Effects in Post-Mortem Brain from Schizophrenia Patients. Mol. Psychiatry 2011, 16, 1189–1202. [Google Scholar] [CrossRef] [PubMed]
  151. Alnafisah, R.S.; Reigle, J.; Eladawi, M.A.; O’Donovan, S.M.; Funk, A.J.; Meller, J.; Mccullumsmith, R.E.; Shukla, R. Assessing the Effects of Antipsychotic Medications on Schizophrenia Functional Analysis: A Postmortem Proteome Study. Neuropsychopharmacology 2022, 47, 2033–2041. [Google Scholar] [CrossRef]
  152. Choi, K.H.; Higgs, B.W.; Weis, S.; Song, J.; Llenos, I.C.; Dulay, J.R.; Yolken, R.H.; Webster, M.J. Effects of Typical and Atypical Antipsychotic Drugs on Gene Expression Profiles in the Liver of Schizophrenia Subjects. BMC Psychiatry 2009, 9, 57. [Google Scholar] [CrossRef]
  153. Mehler-Wex, C.; Grünblatt, E.; Zeiske, S.; Gille, G.; Rausch, D.; Warnke, A.; Gerlach, M. Microarray Analysis Reveals Distinct Gene Expression Patterns in the Mouse Cortex Following Chronic Neuroleptic and Stimulant Treatment: Implications for Body Weight Changes. J. Neural Transm. 2006, 113, 1383–1393. [Google Scholar] [CrossRef]
  154. Grotzinger, A.D.; Singh, K.; Miller-Fleming, T.W.; Lam, M.; Mallard, T.T.; Chen, Y.; Liu, Z.; Ge, T.; Smoller, J.W. Transcriptome-Wide Structural Equation Modeling of 13 Major Psychiatric Disorders for Cross-Disorder Risk and Drug Repurposing. JAMA Psychiatry 2023, 80, 811. [Google Scholar] [CrossRef]
  155. Dudley, J.T.; Deshpande, T.; Butte, A.J. Exploiting Drug-Disease Relationships for Computational Drug Repositioning. Brief. Bioinform. 2011, 12, 303–311. [Google Scholar] [CrossRef] [PubMed]
  156. Reay, W.R.; Cairns, M.J. Advancing the Use of Genome-Wide Association Studies for Drug Repurposing. Nat. Rev. Genet. 2021, 22, 658–671. [Google Scholar] [CrossRef]
  157. So, H.-C.; Chau, C.K.-L.; Chiu, W.-T.; Ho, K.-S.; Lo, C.-P.; Yim, S.H.-Y.; Sham, P.-C. Analysis of Genome-Wide Association Data Highlights Candidates for Drug Repositioning in Psychiatry. Nat. Neurosci. 2017, 20, 1342–1349. [Google Scholar] [CrossRef]
  158. Wang, S.; Ju, D.; Zeng, X. Mechanisms and Clinical Implications of Human Gut Microbiota-Drug Interactions in the Precision Medicine Era. Biomedicines 2024, 12, 194. [Google Scholar] [CrossRef]
  159. Cryan, J.F.; O’Riordan, K.J.; Cowan, C.S.M.; Sandhu, K.V.; Bastiaanssen, T.F.S.; Boehme, M.; Codagnone, M.G.; Cussotto, S.; Fulling, C.; Golubeva, A.V.; et al. The Microbiota-Gut-Brain Axis. Physiol. Rev. 2019, 99, 1877–2013. [Google Scholar] [CrossRef]
  160. Dikeocha, I.J.; Al-Kabsi, A.M.; Miftahussurur, M.; Alshawsh, M.A. Pharmacomicrobiomics: Influence of Gut Microbiota on Drug and Xenobiotic Metabolism. FASEB J. 2022, 36, e22350. [Google Scholar] [CrossRef]
  161. Klünemann, M.; Andrejev, S.; Blasche, S.; Mateus, A.; Phapale, P.; Devendran, S.; Vappiani, J.; Simon, B.; Scott, T.A.; Kafkia, E.; et al. Bioaccumulation of Therapeutic Drugs by Human Gut Bacteria. Nature 2021, 597, 533–538. [Google Scholar] [CrossRef]
  162. Wang, Y.; Zhou, J.; Ye, J.; Sun, Z.; He, Y.; Zhao, Y.; Ren, S.; Zhang, G.; Liu, M.; Zheng, P.; et al. Multi-Omics Reveal Microbial Determinants Impacting the Treatment Outcome of Antidepressants in Major Depressive Disorder. Microbiome 2023, 11, 195. [Google Scholar] [CrossRef]
  163. Tiano, J.P. The Human Microbiome Project … and the Intramural Connection; NIH: Bethesda, MD, USA, 2013. [Google Scholar]
  164. Astudillo-de la Vega, H.; Alonso-Luna, O.; Ali-Pérez, J.; López-Camarillo, C.; Ruiz-Garcia, E. Oncobiome at the Forefront of a Novel Molecular Mechanism to Understand the Microbiome and Cancer. In Translational Research and Onco-Omics Applications in the Era of Cancer Personal Genomics; Springer: Berlin/Heidelberg, Germany, 2019; pp. 147–156. [Google Scholar]
  165. Cussotto, S.; Strain, C.R.; Fouhy, F.; Strain, R.G.; Peterson, V.L.; Clarke, G.; Stanton, C.; Dinan, T.G.; Cryan, J.F. Differential Effects of Psychotropic Drugs on Microbiome Composition and Gastrointestinal Function. Psychopharmacology 2019, 236, 1671–1685. [Google Scholar] [CrossRef] [PubMed]
  166. Clarke, G.; Sandhu, K.V.; Griffin, B.T.; Dinan, T.G.; Cryan, J.F.; Hyland, N.P. Gut Reactions: Breaking Down Xenobiotic–Microbiome Interactions. Pharmacol. Rev. 2019, 71, 198–224. [Google Scholar] [CrossRef]
  167. Skonieczna-Żydecka, K.; Łoniewski, I.; Misera, A.; Stachowska, E.; Maciejewska, D.; Marlicz, W.; Galling, B. Second-Generation Antipsychotics and Metabolism Alterations: A Systematic Review of the Role of the Gut Microbiome. Psychopharmacology 2019, 236, 1491–1512. [Google Scholar] [CrossRef] [PubMed]
  168. Mosquera, F.E.C.; Lizcano Martinez, S.; Liscano, Y. Effectiveness of Psychobiotics in the Treatment of Psychiatric and Cognitive Disorders: A Systematic Review of Randomized Clinical Trials. Nutrients 2024, 16, 1352. [Google Scholar] [CrossRef]
  169. Aresti Sanz, J.; El Aidy, S. Microbiota and Gut Neuropeptides: A Dual Action of Antimicrobial Activity and Neuroimmune Response. Psychopharmacology 2019, 236, 1597–1609. [Google Scholar] [CrossRef]
  170. Smith, D.A.; Sadler, M.C.; Altman, R.B. Promises and Challenges in Pharmacoepigenetics. Camb. Prism. Precis. Med. 2023, 1, e18. [Google Scholar] [CrossRef] [PubMed]
  171. Tyrka, A.R.; Price, L.H.; Marsit, C.; Walters, O.C.; Carpenter, L.L. Childhood Adversity and Epigenetic Modulation of the Leukocyte Glucocorticoid Receptor: Preliminary Findings in Healthy Adults. PLoS ONE 2012, 7, e30148. [Google Scholar] [CrossRef]
  172. Andari, E.; Nishitani, S.; Kaundinya, G.; Caceres, G.A.; Morrier, M.J.; Ousley, O.; Smith, A.K.; Cubells, J.F.; Young, L.J. Epigenetic Modification of the Oxytocin Receptor Gene: Implications for Autism Symptom Severity and Brain Functional Connectivity. Neuropsychopharmacology 2020, 45, 1150–1158. [Google Scholar] [CrossRef] [PubMed]
  173. Zhou, J.; Li, M.; Wang, X.; He, Y.; Xia, Y.; Sweeney, J.A.; Kopp, R.F.; Liu, C.; Chen, C. Drug Response-Related DNA Methylation Changes in Schizophrenia, Bipolar Disorder, and Major Depressive Disorder. Front. Neurosci. 2021, 15, 674273. [Google Scholar] [CrossRef]
  174. Wang, P.; Zhang, C.; Lv, Q.; Bao, C.; Sun, H.; Ma, G.; Fang, Y.; Yi, Z.; Cai, W. Association of DNA Methylation in BDNF with Escitalopram Treatment Response in Depressed Chinese Han Patients. Eur. J. Clin. Pharmacol. 2018, 74, 1011–1020. [Google Scholar] [CrossRef]
  175. Tadić, A.; Müller-Engling, L.; Schlicht, K.F.; Kotsiari, A.; Dreimüller, N.; Kleimann, A.; Bleich, S.; Lieb, K.; Frieling, H. Methylation of the Promoter of Brain-Derived Neurotrophic Factor Exon IV and Antidepressant Response in Major Depression. Mol. Psychiatry 2014, 19, 281–283. [Google Scholar] [CrossRef]
  176. Li, L.; Wang, T.; Chen, S.; Yue, Y.; Xu, Z.; Yuan, Y. DNA Methylations of Brain-Derived Neurotrophic Factor Exon VI Are Associated with Major Depressive Disorder and Antidepressant-Induced Remission in Females. J. Affect. Disord. 2021, 295, 101–107. [Google Scholar] [CrossRef]
  177. Lieb, K.; Dreimüller, N.; Wagner, S.; Schlicht, K.; Falter, T.; Neyazi, A.; Müller-Engling, L.; Bleich, S.; Tadić, A.; Frieling, H. BDNF Plasma Levels and BDNF Exon IV Promoter Methylation as Predictors for Antidepressant Treatment Response. Front. Psychiatry 2018, 9, 511. [Google Scholar] [CrossRef]
  178. Fakhoury, M. New Insights into the Neurobiological Mechanisms of Major Depressive Disorders. Gen. Hosp. Psychiatry 2015, 37, 172–177. [Google Scholar] [CrossRef]
  179. Chilmonczyk, Z.; Bojarski, A.; Pilc, A.; Sylte, I. Functional Selectivity and Antidepressant Activity of Serotonin 1A Receptor Ligands. Int. J. Mol. Sci. 2015, 16, 18474–18506. [Google Scholar] [CrossRef] [PubMed]
  180. Tiger, M.; Varnäs, K.; Okubo, Y.; Lundberg, J. The 5-HT1B Receptor—A Potential Target for Antidepressant Treatment. Psychopharmacology 2018, 235, 1317–1334. [Google Scholar] [CrossRef]
  181. Xu, Z.; Chen, Z.; Shen, T.; Chen, L.; Tan, T.; Gao, C.; Chen, B.; Yuan, Y.; Zhang, Z. The Impact of HTR1A and HTR1B Methylation Combined with Stress/Genotype on Early Antidepressant Efficacy. Psychiatry Clin. Neurosci. 2022, 76, 51–57. [Google Scholar] [CrossRef]
  182. Kang, H.-J.; Kim, J.-M.; Stewart, R.; Kim, S.-Y.; Bae, K.-Y.; Kim, S.-W.; Shin, I.-S.; Shin, M.-G.; Yoon, J.-S. Association of SLC6A4 Methylation with Early Adversity, Characteristics and Outcomes in Depression. Prog. Neuropsychopharmacol. Biol. Psychiatry 2013, 44, 23–28. [Google Scholar] [CrossRef] [PubMed]
  183. Okada, S.; Morinobu, S.; Fuchikami, M.; Segawa, M.; Yokomaku, K.; Kataoka, T.; Okamoto, Y.; Yamawaki, S.; Inoue, T.; Kusumi, I.; et al. The Potential of SLC6A4 Gene Methylation Analysis for the Diagnosis and Treatment of Major Depression. J. Psychiatr. Res. 2014, 53, 47–53. [Google Scholar] [CrossRef]
  184. Iga, J.; Watanabe, S.; Numata, S.; Umehara, H.; Nishi, A.; Kinoshita, M.; Inoshita, M.; Shimodera, S.; Fujita, H.; Ohmori, T. Association Study of Polymorphism in the Serotonin Transporter Gene Promoter, Methylation Profiles, and Expression in Patients with Major Depressive Disorder. Human Psychopharmacol. Clin. Exp. 2016, 31, 193–199. [Google Scholar] [CrossRef] [PubMed]
  185. Domschke, K.; Tidow, N.; Schwarte, K.; Deckert, J.; Lesch, K.-P.; Arolt, V.; Zwanzger, P.; Baune, B.T. Serotonin Transporter Gene Hypomethylation Predicts Impaired Antidepressant Treatment Response. Int. J. Neuropsychopharmacol. 2014, 17, 1167–1176. [Google Scholar] [CrossRef]
  186. Schiele, M.A.; Zwanzger, P.; Schwarte, K.; Arolt, V.; Baune, B.T.; Domschke, K. Serotonin Transporter Gene Promoter Hypomethylation as a Predictor of Antidepressant Treatment Response in Major Depression: A Replication Study. Int. J. Neuropsychopharmacol. 2021, 24, 191–199. [Google Scholar] [CrossRef]
  187. Bayles, R.; Baker, E.K.; Jowett, J.B.M.; Barton, D.; Esler, M.; El-Osta, A.; Lambert, G. Methylation of the SLC6a2 Gene Promoter in Major Depression and Panic Disorder. PLoS ONE 2013, 8, e83223. [Google Scholar] [CrossRef]
  188. Ryan, J.; Pilkington, L.; Neuhaus, K.; Ritchie, K.; Ancelin, M.-L.; Saffery, R. Investigating the Epigenetic Profile of the Inflammatory Gene IL-6 in Late-Life Depression. BMC Psychiatry 2017, 17, 354. [Google Scholar] [CrossRef]
  189. Dahl, J.; Ormstad, H.; Aass, H.C.D.; Malt, U.F.; Bendz, L.T.; Sandvik, L.; Brundin, L.; Andreassen, O.A. The Plasma Levels of Various Cytokines Are Increased during Ongoing Depression and Are Reduced to Normal Levels after Recovery. Psychoneuroendocrinology 2014, 45, 77–86. [Google Scholar] [CrossRef] [PubMed]
  190. Takeuchi, N.; Nonen, S.; Kato, M.; Wakeno, M.; Takekita, Y.; Kinoshita, T.; Kugawa, F. Therapeutic Response to Paroxetine in Major Depressive Disorder Predicted by DNA Methylation. Neuropsychobiology 2017, 75, 81–88. [Google Scholar] [CrossRef] [PubMed]
  191. Belzeaux, R.; Lin, R.; Ju, C.; Chay, M.-A.; Fiori, L.M.; Lutz, P.-E.; Turecki, G. Transcriptomic and Epigenomic Biomarkers of Antidepressant Response. J. Affect. Disord. 2018, 233, 36–44. [Google Scholar] [CrossRef] [PubMed]
  192. Fiori, L.M.; Lin, R.; Ju, C.; Belzeaux, R.; Turecki, G. Using Epigenetic Tools to Investigate Antidepressant Response. In Progress in Molecular Biology and Translational Science; Elsevier: Amsterdam, The Netherlands, 2018; pp. 255–272. [Google Scholar]
  193. Sales, A.J.; Joca, S.R.L. Effects of DNA Methylation Inhibitors and Conventional Antidepressants on Mice Behaviour and Brain DNA Methylation Levels. Acta Neuropsychiatr. 2016, 28, 11–22. [Google Scholar] [CrossRef] [PubMed]
  194. Wei, Y.B.; Melas, P.A.; Wegener, G.; Mathe, A.A.; Lavebratt, C. Antidepressant-Like Effect of Sodium Butyrate Is Associated with an Increase in TET1 and in 5-Hydroxymethylation Levels in the Bdnf Gene. Int. J. Neuropsychopharmacol. 2015, 18, pyu032. [Google Scholar] [CrossRef] [PubMed]
  195. Qiao, M.; Jiang, Q.-S.; Liu, Y.-J.; Hu, X.-Y.; Wang, L.-J.; Zhou, Q.-X.; Qiu, H.-M. Antidepressant Mechanisms of Venlafaxine Involving Increasing Histone Acetylation and Modulating Tyrosine Hydroxylase and Tryptophan Hydroxylase Expression in Hippocampus of Depressive Rats. Neuroreport 2019, 30, 255–261. [Google Scholar] [CrossRef]
  196. Sales, A.J.; Biojone, C.; Terceti, M.S.; Guimarães, F.S.; Gomes, M.V.; Joca, S.R. Antidepressant-like Effect Induced by Systemic and Intra-hippocampal Administration of DNA Methylation Inhibitors. Br. J. Pharmacol. 2011, 164, 1711–1721. [Google Scholar] [CrossRef] [PubMed]
  197. Hsieh, M.-C.; Lin, K.-D.; Tien, K.-J.; Tu, S.-T.; Hsiao, J.-Y.; Chang, S.-J.; Lin, S.-R.; Shing, S.-J.; Chen, H.-C. Common Polymorphisms of the Peroxisome Proliferator-Activated Receptor–γ (Pro12Ala) and Peroxisome Proliferator-Activated Receptor–γ Coactivator–1 (Gly482Ser) and the Response to Pioglitazone in Chinese Patients with Type 2 Diabetes Mellitus. Metabolism 2010, 59, 1139–1144. [Google Scholar] [CrossRef] [PubMed]
  198. Cassoli, J.S.; Guest, P.C.; Santana, A.G.; Martins-de-Souza, D. Employing Proteomics to Unravel the Molecular Effects of Antipsychotics and Their Role in Schizophrenia. Proteom. Clin. Appl. 2016, 10, 442–455. [Google Scholar] [CrossRef]
  199. Schwarz, E.; Guest, P.C.; Steiner, J.; Bogerts, B.; Bahn, S. Identification of Blood-Based Molecular Signatures for Prediction of Response and Relapse in Schizophrenia Patients. Transl. Psychiatry 2012, 2, e82. [Google Scholar] [CrossRef] [PubMed]
  200. Uher, R.; Maier, W.; Hauser, J.; Marušič, A.; Schmael, C.; Mors, O.; Henigsberg, N.; Souery, D.; Placentino, A.; Rietschel, M.; et al. Differential Efficacy of Escitalopram and Nortriptyline on Dimensional Measures of Depression. Br. J. Psychiatry 2009, 194, 252–259. [Google Scholar] [CrossRef]
  201. McGuffin, P. GENome-Based Therapeutic Drugs for DEPression. Available online: http://isrctn.org/ (accessed on 27 November 2024).
  202. Malki, K.; Campbell, J.; Davies, M.; Keers, R.; Uher, R.; Ward, M.; Paya-Cano, J.; Aitchinson, K.J.; Binder, E.; Sluyter, F.; et al. Pharmacoproteomic Investigation into Antidepressant Response in Two Mouse Inbred Strains. Proteomics 2012, 12, 2355–2365. [Google Scholar] [CrossRef]
  203. Athreya, A.P.; Neavin, D.; Carrillo-Roa, T.; Skime, M.; Biernacka, J.; Frye, M.A.; Rush, A.J.; Wang, L.; Binder, E.B.; Iyer, R.K.; et al. Pharmacogenomics-Driven Prediction of Antidepressant Treatment Outcomes: A Machine-Learning Approach with Multi-trial Replication. Clin. Pharmacol. Ther. 2019, 106, 855–865. [Google Scholar] [CrossRef]
  204. Chang, B.; Choi, Y.; Jeon, M.; Lee, J.; Han, K.-M.; Kim, A.; Ham, B.-J.; Kang, J. ARPNet: Antidepressant Response Prediction Network for Major Depressive Disorder. Genes 2019, 10, 907. [Google Scholar] [CrossRef]
  205. Maciukiewicz, M.; Marshe, V.S.; Hauschild, A.-C.; Foster, J.A.; Rotzinger, S.; Kennedy, J.L.; Kennedy, S.H.; Müller, D.J.; Geraci, J. GWAS-Based Machine Learning Approach to Predict Duloxetine Response in Major Depressive Disorder. J. Psychiatr. Res. 2018, 99, 62–68. [Google Scholar] [CrossRef] [PubMed]
  206. Iniesta, R.; Malki, K.; Maier, W.; Rietschel, M.; Mors, O.; Hauser, J.; Henigsberg, N.; Dernovsek, M.Z.; Souery, D.; Stahl, D.; et al. Combining Clinical Variables to Optimize Prediction of Antidepressant Treatment Outcomes. J. Psychiatr. Res. 2016, 78, 94–102. [Google Scholar] [CrossRef] [PubMed]
  207. Patel, M.J.; Andreescu, C.; Price, J.C.; Edelman, K.L.; Reynolds, C.F.; Aizenstein, H.J. Machine Learning Approaches for Integrating Clinical and Imaging Features in Late-life Depression Classification and Response Prediction. Int. J. Geriatr. Psychiatry 2015, 30, 1056–1067. [Google Scholar] [CrossRef] [PubMed]
  208. Kautzky, A.; Baldinger, P.; Souery, D.; Montgomery, S.; Mendlewicz, J.; Zohar, J.; Serretti, A.; Lanzenberger, R.; Kasper, S. The Combined Effect of Genetic Polymorphisms and Clinical Parameters on Treatment Outcome in Treatment-Resistant Depression. Eur. Neuropsychopharmacol. 2015, 25, 441–453. [Google Scholar] [CrossRef]
  209. Dwyer, D.B.; Falkai, P.; Koutsouleris, N. Machine Learning Approaches for Clinical Psychology and Psychiatry. Annu. Rev. Clin. Psychol. 2018, 14, 91–118. [Google Scholar] [CrossRef]
  210. Joyce, J.B.; Grant, C.W.; Liu, D.; MahmoudianDehkordi, S.; Kaddurah-Daouk, R.; Skime, M.; Biernacka, J.; Frye, M.A.; Mayes, T.; Carmody, T.; et al. Multi-Omics Driven Predictions of Response to Acute Phase Combination Antidepressant Therapy: A Machine Learning Approach with Cross-Trial Replication. Transl. Psychiatry 2021, 11, 513. [Google Scholar] [CrossRef]
  211. Gillett, G.; Tomlinson, A.; Efthimiou, O.; Cipriani, A. Predicting Treatment Effects in Unipolar Depression: A Meta-Review. Pharmacol. Ther. 2020, 212, 107557. [Google Scholar] [CrossRef]
  212. Perlman, K.; Benrimoh, D.; Israel, S.; Rollins, C.; Brown, E.; Tunteng, J.-F.; You, R.; You, E.; Tanguay-Sela, M.; Snook, E.; et al. A Systematic Meta-Review of Predictors of Antidepressant Treatment Outcome in Major Depressive Disorder. J. Affect. Disord. 2019, 243, 503–515. [Google Scholar] [CrossRef] [PubMed]
  213. Lin, E.; Lin, C.-H.; Lane, H.-Y. Precision Psychiatry Applications with Pharmacogenomics: Artificial Intelligence and Machine Learning Approaches. Int. J. Mol. Sci. 2020, 21, 969. [Google Scholar] [CrossRef]
  214. van Westrhenen, R.; Aitchison, K.J.; Ingelman-Sundberg, M.; Jukić, M.M. Pharmacogenomics of Antidepressant and Antipsychotic Treatment: How Far Have We Got and Where Are We Going? Front. Psychiatry 2020, 11, 94. [Google Scholar] [CrossRef]
  215. Fabbri, C.; Kasper, S.; Kautzky, A.; Zohar, J.; Souery, D.; Montgomery, S.; Albani, D.; Forloni, G.; Ferentinos, P.; Rujescu, D.; et al. A Polygenic Predictor of Treatment-Resistant Depression Using Whole Exome Sequencing and Genome-Wide Genotyping. Transl. Psychiatry 2020, 10, 50. [Google Scholar] [CrossRef]
  216. Pisanu, C.; Squassina, A. Treatment-Resistant Schizophrenia: Insights From Genetic Studies and Machine Learning Approaches. Front. Pharmacol. 2019, 10, 617. [Google Scholar] [CrossRef]
  217. Lin, E.; Kuo, P.-H.; Liu, Y.-L.; Yu, Y.W.-Y.; Yang, A.C.; Tsai, S.-J. A Deep Learning Approach for Predicting Antidepressant Response in Major Depression Using Clinical and Genetic Biomarkers. Front. Psychiatry 2018, 9, 290. [Google Scholar] [CrossRef]
  218. Chekroud, A.M.; Zotti, R.J.; Shehzad, Z.; Gueorguieva, R.; Johnson, M.K.; Trivedi, M.H.; Cannon, T.D.; Krystal, J.H.; Corlett, P.R. Cross-Trial Prediction of Treatment Outcome in Depression: A Machine Learning Approach. Lancet Psychiatry 2016, 3, 243–250. [Google Scholar] [CrossRef]
  219. Thakkar, A.; Gupta, A.; De Sousa, A. Artificial Intelligence in Positive Mental Health: A Narrative Review. Front. Digit. Health 2024, 6, 1280235. [Google Scholar] [CrossRef]
  220. Ostojic, D.; Lalousis, P.A.; Donohoe, G.; Morris, D.W. The Challenges of Using Machine Learning Models in Psychiatric Research and Clinical Practice. Eur. Neuropsychopharmacol. 2024, 88, 53–65. [Google Scholar] [CrossRef] [PubMed]
  221. Sanchez, T.W.; Brenman, M.; Ye, X. The Ethical Concerns of Artificial Intelligence in Urban Planning. In Journal of the American Planning Association; Taylor & Francis: Milton Park, UK, 2024; pp. 1–14. [Google Scholar] [CrossRef]
  222. Eugene, A.R.; Masiak, J.; Eugene, B. Predicting Lithium Treatment Response in Bipolar Patients Using Gender-Specific Gene Expression Biomarkers and Machine Learning. F1000Research 2018, 7, 474. [Google Scholar] [CrossRef] [PubMed]
  223. Cepeda, M.S.; Reps, J.; Fife, D.; Blacketer, C.; Stang, P.; Ryan, P. Finding Treatment-Resistant Depression in Real-World Data: How a Data-Driven Approach Compares with Expert-Based Heuristics. Depress. Anxiety 2018, 35, 220–228. [Google Scholar] [CrossRef]
  224. Kautzky, A.; Baldinger-Melich, P.; Kranz, G.S.; Vanicek, T.; Souery, D.; Montgomery, S.; Mendlewicz, J.; Zohar, J.; Serretti, A.; Lanzenberger, R.; et al. A New Prediction Model for Evaluating Treatment-Resistant Depression. J. Clin. Psychiatry 2017, 78, 215–222. [Google Scholar] [CrossRef] [PubMed]
  225. Pradier, M.F.; Hughes, M.C.; McCoy, T.H.; Barroilhet, S.A.; Doshi-Velez, F.; Perlis, R.H. Predicting Change in Diagnosis from Major Depression to Bipolar Disorder after Antidepressant Initiation. Neuropsychopharmacology 2021, 46, 455–461. [Google Scholar] [CrossRef] [PubMed]
  226. Sullivan, P.F.; Agrawal, A.; Bulik, C.M.; Andreassen, O.A.; Børglum, A.D.; Breen, G.; Cichon, S.; Edenberg, H.J.; Faraone, S.V.; Gelernter, J.; et al. Psychiatric Genomics: An Update and an Agenda. Am. J. Psychiatry 2018, 175, 15–27. [Google Scholar] [CrossRef]
  227. FENS. European College of Neuropsychopharmacology (ECNP); FENS: Madrid, Spain. Available online: https://www.fens.org/membership/societies/european-college-of-neuropsychopharmacology-ecnp (accessed on 17 October 2024).
  228. Elliott, L.T.; Sharp, K.; Alfaro-Almagro, F.; Shi, S.; Miller, K.L.; Douaud, G.; Marchini, J.; Smith, S.M. Genome-Wide Association Studies of Brain Imaging Phenotypes in UK Biobank. Nature 2018, 562, 210–216. [Google Scholar] [CrossRef]
  229. Ross, C.J.D.; Visscher, H.; Sistonen, J.; Brunham, L.R.; Pussegoda, K.; Loo, T.T.; Rieder, M.J.; Koren, G.; Carleton, B.C.; Hayden, M.R. The Canadian Pharmacogenomics Network for Drug Safety: A Model for Safety Pharmacology. Thyroid 2010, 20, 681–687. [Google Scholar] [CrossRef]
  230. Picard, N.; Boyer, J.-C.; Etienne-Grimaldi, M.-C.; Barin-Le Guellec, C.; Thomas, F.; Loriot, M.-A. Pharmacogenetics-Based Personalized Therapy: Levels of Evidence and Recommendations from the French Network of Pharmacogenetics (RNPGx). Therapies 2017, 72, 185–192. [Google Scholar] [CrossRef]
  231. ASHP. Endorsed Documents; ASHP: Bethesda, MD, USA. Available online: https://www.ashp.org/pharmacy-practice/policy-positions-and-guidelines/browse-by-document-type/endorsed-documents?loginreturnUrl=SSOCheckOnly (accessed on 20 November 2024).
  232. Duong, B.Q.; Arwood, M.J.; Hicks, J.K.; Beitelshees, A.L.; Franchi, F.; Houder, J.T.; Limdi, N.A.; Cook, K.J.; Owusu Obeng, A.; Petry, N.; et al. Development of Customizable Implementation Guides to Support Clinical Adoption of Pharmacogenomics: Experiences of the Implementing GeNomics in PracTicE (IGNITE) Network. Pharmgenom. Pers. Med. 2020, 13, 217–226. [Google Scholar] [CrossRef] [PubMed]
  233. Orlando, L.A.; Sperber, N.R.; Voils, C.; Nichols, M.; Myers, R.A.; Wu, R.R.; Rakhra-Burris, T.; Levy, K.D.; Levy, M.; Pollin, T.I.; et al. Developing a Common Framework for Evaluating the Implementation of Genomic Medicine Interventions in Clinical Care: The IGNITE Network’s Common Measures Working Group. Genet. Med. 2018, 20, 655–663. [Google Scholar] [CrossRef]
  234. Oslin, D.W.; Chapman, S.; Duvall, S.L.; Gelernter, J.; Ingram, E.P.; Kranzler, H.R.; Lehmann, L.S.; Lynch, J.A.; Lynch, K.G.; Pyne, J.M.; et al. Study Design and Implementation of the PRecision Medicine In MEntal Health Care (PRIME Care) Trial. Contemp. Clin. Trials 2021, 101, 106247. [Google Scholar] [CrossRef]
  235. Swen, J.J.; van der Wouden, C.H.; Manson, L.E.; Abdullah-Koolmees, H.; Blagec, K.; Blagus, T.; Böhringer, S.; Cambon-Thomsen, A.; Cecchin, E.; Cheung, K.-C.; et al. A 12-Gene Pharmacogenetic Panel to Prevent Adverse Drug Reactions: An Open-Label, Multicentre, Controlled, Cluster-Randomised Crossover Implementation Study. Lancet 2023, 401, 347–356. [Google Scholar] [CrossRef] [PubMed]
  236. IQ Consortium. IQ Machine Intelligence for Quantitative Modeling in Drug Discovery & Development Applications; IQ Consortium: Washington, WA, USA, 2022. [Google Scholar]
  237. FDA; University of Maryland. Application of Artificial Intelligence & Machine Learning for Precision Medicine; FDA & University of Maryland: Silver Spring, MD, USA, 2023. [Google Scholar]
  238. Liu, Q.; Joshi, A.; Standing, J.F.; van der Graaf, P.H. Artificial Intelligence/Machine Learning: The New Frontier of Clinical Pharmacology and Precision Medicine. Clin. Pharmacol. Ther. 2024, 115, 637–642. [Google Scholar] [CrossRef] [PubMed]
  239. Siddiqi, S.H.; Taylor, J.J.; Horn, A.; Fox, M.D. Bringing Human Brain Connectomics to Clinical Practice in Psychiatry. Biol. Psychiatry 2023, 93, 386–387. [Google Scholar] [CrossRef] [PubMed]
  240. Sharew, N.T.; Clark, S.R.; Schubert, K.O.; Amare, A.T. Pharmacogenomic Scores in Psychiatry: Systematic Review of Current Evidence. Transl. Psychiatry 2024, 14, 322. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Navigating the path to precision psychiatry. This figure outlines a novel approach in personalized psychiatry, predicated on the integration of diverse datasets and encompassing pharmaco-multiomics. Utilizing advanced AI and ML technologies, these integrated data are leveraged to yield precise insights that enable the formulation of tailored therapeutic interventions and dynamic management strategies. This methodology distinctly addresses the intricate needs of psychiatric care and emphasizes patient-centered care.
Figure 1. Navigating the path to precision psychiatry. This figure outlines a novel approach in personalized psychiatry, predicated on the integration of diverse datasets and encompassing pharmaco-multiomics. Utilizing advanced AI and ML technologies, these integrated data are leveraged to yield precise insights that enable the formulation of tailored therapeutic interventions and dynamic management strategies. This methodology distinctly addresses the intricate needs of psychiatric care and emphasizes patient-centered care.
Ijms 26 01082 g001
Table 1. Key genes and guidelines for psychotropic medications (PharmGKB).
Table 1. Key genes and guidelines for psychotropic medications (PharmGKB).
ClassDrugInformative PGx
(PharmGKB Level 2, 3, and 4)
Actionable PGx
(PharmGKB Level 1)
VariantRecommendation CPIC/DPWG
AntidepressantsCitalopramABCB1
BDNF
CACNA1C
COL26A1
CREB1, METTL21A
CRHR2
CYP2D6
DTNBP1
ERICH3
FKBP5
GLDC
GRIA3
GRK5
GSK3B
HTR1B
HTR2A
NEDD4L
PAPLN
REEP5
RFK
RORA
SERPINE1
SRP19
TPH2
BDNF
CYP2D19
CYP2D6
HTR2A
SLC6A4
TPH1
TSPAN5
CYP2C19CYP2C19*1; CYP2C19*2; CYP2C19*3; CYP2C19*17
CYP2C19*4
UM, LP, and PM: consideration of a clinically appropriate antidepressant not predominantly metabolized by CYP2C19 for CYP2C19
In case citalopram or escitalopram are clinically appropriate, dose alterations are recommended.
CPIC
AntidepressantsEscitalopramBDNF
BMP5
CYP1A2
CYP2D6
ERICH3
FKBP5
GLDC
GRK5
HTR1B
HTR2A
HTR2C
HTR7
IL11
RFK
SCL6A4
TSPAN5
CYP2C19CYP2C19*1; CYP2C19*2; CYP2C19*3; CYP2C19*17UM, LP, and PM: consideration of a clinically appropriate antidepressant not predominantly metabolized by CYP2C19 for CYP2C19
In case citalopram or escitalopram are clinically appropriate, dose alterations are recommended.
CPIC
AntidepressantsFluvoxamineABCB1
COMT
FGF2
HTR1A
MDGA2
SLC6A4
HTR2A
CYP2D6CYP2D6*1; CYP2D6*4; CYP2D6*5; CYP2D6*6; CYP2D6*10; CYP2D6*14PM: 25–50% reduction in recommended starting dose and slower titration schedule or use of an alternative drug not metabolized by CYP2D6 CPIC
AntidepressantsParoxetineABCB1
BDNF
COMT
CYP1A2
DRD3
FKBP5
GDNF
HTR1A
HTR1B
HTR2A
HTR7, RPP30
MDGA2
REEP5
SLC6A4
SRP19
HTR2A
HTR3B
TPH1
CYP2D6CYP2D6*1; CYP2D6*1xN; CYP2D6*2; CYP2D6*2xN; CYP2D6*3; CYP2D6*4; CYP2D6*5; CYP2D6*9; CYP2D6*10; CYP2D6*14; CYP2D6*41UM: alternative drug not predominantly metabolized by CYP2D6.
PM: 50% reduction in recommended starting dose, slower titration schedule, and a 50% lower maintenance dose
CPIC
AntidepressantsSertralineABCB1
ACE
CYP2D6
GNB3, P3H3
HTR1A
REEP5
SRP19
HTR2A
SLC6A4
CYP2C19, CYP2B6CYP2B6*1; CYP2B6*4; CYP2B6*6; CYP2B6*9
CYP2C19*1; CYP2C19*2; CYP2C19*3; CYP2C19*17
CYP2C19 PM: 50% reduction in recommended starting dose and titrate to response or selection of an alternative drug not predominantly metabolized by CYP2C19CPIC
AntidepressantsImipramine-CYP2C19, CYP2D6CYP2C19*1; CYP2C19*2; CYP2C19*3; CYP2C19*17 CYP2D6*1; CYP2D6*3; CYP2D6*4; CYP2D6*5; CYP2D6*6; CYP2D6*1xN; CYP2D6*2xNCYP2D6 IM: a 25% dose reduction should be considered.CPIC
AntidepressantsClomipramineABCB1
FKBP5
HTR1B
SLC6A4
CYP2C19, CYP2D6CYP2C19*1; CYP2C19*2; CYP2C19*3; CYP2C19*17 CYP2D6*1; CYP2D6*1xN; CYP2D6*2; CYP2D6*3; CYP2D6*4; CYP2D6*5; CYP2D6*6; CYP2D6*10; CYP2D6*41IM: 25% dose reductionCPIC
AntidepressantsNortriptyline-CYP2D6-IM: 25% dose reduction
UM/PM: alternative drug should be considered. If nortriptyline is warranted, consider a 50% dose reduction in CYP2D6-poor metabolizers.
CPIC
AntidepressantsAmitriptylineABCB1
CYP2D6 (CYP2D6*1, CYP2D6*87, CYP2D6*88, CYP2D6*89, CYP2D6*90,CYP2D6*91,CYP2D6*93, CYP2D6*94, CYP2D6*95,
CYP2D6*97, CYP2D6*98)
CYP2C19, CYP2D6CYP2C19*1; CYP2C19*2; CYP2C19*3; CYP2C19*17 CYP2D6*1; CYP2D6*1xN; CYP2D6*2; CYP2D6*3; CYP2D6*4; CYP2D6*5; CYP2D6*6; CYP2D6*10; CYP2D6*41CYP2D6 UM/PM or CYP2C19 UM/RM/PM: alternative drug
PM: If amitriptyline is warranted, consider a 50% dose reduction.
CYP2D6 IM: a 25% dose reduction should be considered.
CPIC
AntidepressantsVenlafaxineABCB1
COMT
CYP2C19 (CYP2C19*2)
CYP2D6 (CYP2D6*1, CYP2D6*87, CYP2D6*88, CYP2D6*89, CYP2D6*90, CYP2D6*91, CYP2D6*93,
CYP2D6*94, CYP2D6*95,
CYP2D6*97, CYP2D6*98)
FKBP5
GABRA6
GABRP
GABRQ
GRIA3
HTR2A
SLC6A2
TPH2
CYP2C19 (CYP2C19*1; CYP2C19*2)
HTR2A
SLC6A4
CYP2D6CYP2D6*1; CYP2D6*3; CYP2D6*4; CYP2D6*6; CYP2D6*81; CYP2D6*5; CYP2D6*10PM: alternative antidepressant not predominantly metabolized by CYP2D6CPIC
AntipsychoticsAripiprazoleABCB1
ANKK1
DRD2
FAAH
MC4R
RABEP1
SH2B1
TAAR6
CYP2D6CYP2D6*1; CYP2D6*4; CYP2D6*5; CYP2D6*6; CYP2D6*10; CYP2D6*41PM: reduce maximum doseDPWG
AntipsychoticsRisperidoneABCB1
ADRB2
AKT1
ANKK, DRD2
CCL2
CNR1
COMT
CYP1B1
CYP2D6 (CYP2D6*1, CYP2D6*87,
CYP2D6*88, CYP2D6*89, CYP2D6*90,CYP2D6*91, CYP2D6*93, CYP2D6*94, CYP2D6*95, CYP2D6*97, CYP2D6*98)
CYP2E1
DRD2
DRD3
EIF2AK4
EPM2A
FAAH
GRID2
GRIN2B
GRM3
GRM7
HRH3
HRH4
HTR1A
HTR2A
HTR2C
HTT, MSANTD1
LEP
MC4R
NR1I2
PPA2
RABEP1
RGS4
SH2B1
SLC6A4
TJP1
TNFRSF11A
UGT1A1, UGT1A10,UGT1A3, UGT1A4, UGT1A5, UGT1A6,
UGT1A7, UGT1A8, UGT1A9
ABCB1
ANKK1, DRD2
COMT
DRD2
CYP2D6CYP2D6*1; CYP2D6*1xN,
CYP2D6*3; CYP2D6*4; CYP2D6*5; CYP2D6*6; CYP2D6*10; CYP2D6*14
PM: decrease dose
UM: alternative drug or titration of the dose according to the maximum dose of the active metabolite
DPWG
AntipsychoticsZuclopenthixol-CYP2D6CYP2D6*1, CYP2D6*3, CYP2D6*4PM/IM: dose reduction
UM: increase dose (not exceeding 1.5x normal dose) if needed
DPWG
ADHDAtomoxetineCYP2D6 (CYP2D6*1, CYP2D6*87, CYP2D6*88, CYP2D6*89, CYP2D6*90, CYP2D6*91, CYP2D6*93, CYP2D6*94, CYP2D6*95, CYP2D6*97, CYP2D6*98, CYP2D6*4, CYP2D6*5)
SLC6A2
CYP2D6CYP2D6*1; CYP2D6*3; CYP2D6*4; CYP2D6*4xN; CYP2D6*5; CYP2D6*6; CYP2D6*10; CYP2D6*17; CYP2D6*92; CYP2D6*96UM: be alert to reduced efficacy or select alternative drug as precaution
PM: be alert to side effects
DPWG
AnticonvulsantsCarbamazepineSCN1A
ABCB1
ABCC2
BAG6, PRRC2A
CYP1A1
CYP1A2
CYP3A4
CYP3A5
EPHX1
GABRA1
HLA-A (HLA-A*24:02; HLA-A*11:01, HLA-A*02:01)
HLA-B (HLA-B*46:01, HLA-B*15:18, HLA-B*15:21, HLA-B*51:01, HLA-B*59:01, HLA-b*58:01, HLA-B*40:01)
HLA-C
HLA-DRB1
HSPA1A, HSPA1L
LTA, TNF
NR1I2
SCN2A
UGT2B7
HNF4A
SCN1A
SCN2A
HLA-A, HLA-BHLA-A*31:01
HLA-B*15:02; HLA-B*15:11
Alternative drug for carbamazepine-naive patients carrying at least one copy of either HLA-B*15:02 or HLA-A*31:01 CPIC
AnticonvulsantsOxcarbazepineABCB1
HLA-A
HLA-B (HLA-B*27:09, HLA-B*13:02, HLA-B*38:02, HLA-B*48:04, HLA-B*15:19, HLA-B*15:27, HLA-B*40:02, HLA-B*15:18, HLA-B*40:01, HLA-B*15:02)
HLA-DRB1
SCN2A
UGT1A10, UGT1A7, UGT1A8, UGT1A9
UGT2B7
ABCC2
SCN1A
HLA-BB*15:02Alternative drug for oxcarbazepine-naive patients carrying at least one copy of HLA-B*15:02CPIC
PharmGKB: Pharmacogenomics Knowledgebase; PGx: pharmacogenes; CYP: cytochrome P450; UM: ultra-rapid metabolizer; IM: intermediate metabolizer; LP: likely poor metabolizer; PM: poor metabolizer; CPIC: Clinical Pharmacogenetics Implementation Consortium; DPWG: Dutch Pharmacogenetic Working Group.
Table 2. Overview of key studies investigating DNA methylation patterns associated with response to psychiatric treatments.
Table 2. Overview of key studies investigating DNA methylation patterns associated with response to psychiatric treatments.
ReferenceParticipant Treatment(s)Duration (Weeks)Outcome MeasuresGenetic Marks AssessedSample TypeKey Findings
Hsieh et al. [197]MDD patients (Taiwan)SSRIs and benzodiazepines or hypnotics
(no mood stabilizers or antipsychotic drugs)
4HAM-D1714 CpG sites in BDNF exon IXPeripheral bloodHigher methylation level at CpG 24 and CpG 324 of BDNF exon IX associated with improved response
Schiele et al. [186]MDD patients (Caucasian)Antidepressants6HAM-D219 CpG sites in SLC6A4 promoter regionBlood cellsHigher methylation of CpG sites correlated with treatment response
Xu, Chen, et al. [181]MDD patients
(Chinese, Han)
Antidepressants 2HAM-D17 LES
CTQ
181 CpG sites in HTR1A and HTR1BPeripheral bloodMethylation of HTR1A-2-143 and HTR1B-3-61 associated with response; interaction with rs6298 genotype
Wang, Zhang, et al. [174]MDD patients (Chinese, Han)Escitalopram 8HAMD-17 LES
CTQ
90 CpG sites in BDNFPeripheral bloodMethylation at four BDNF amplicons associated with response (higher DNA methylation associated with better treatment response)
Takeuchi et al. [190] MDD patients (Japanese)Paroxetine6HAM-D 21 changeGenome-wide DNA methylation Peripheral bloodMethylation of PPFIA4 CpG cg00594917 and HS3ST1 CpG cg07260927 associated with response
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.

Share and Cite

MDPI and ACS Style

Dhieb, D.; Bastaki, K. Pharmaco-Multiomics: A New Frontier in Precision Psychiatry. Int. J. Mol. Sci. 2025, 26, 1082. https://doi.org/10.3390/ijms26031082

AMA Style

Dhieb D, Bastaki K. Pharmaco-Multiomics: A New Frontier in Precision Psychiatry. International Journal of Molecular Sciences. 2025; 26(3):1082. https://doi.org/10.3390/ijms26031082

Chicago/Turabian Style

Dhieb, Dhoha, and Kholoud Bastaki. 2025. "Pharmaco-Multiomics: A New Frontier in Precision Psychiatry" International Journal of Molecular Sciences 26, no. 3: 1082. https://doi.org/10.3390/ijms26031082

APA Style

Dhieb, D., & Bastaki, K. (2025). Pharmaco-Multiomics: A New Frontier in Precision Psychiatry. International Journal of Molecular Sciences, 26(3), 1082. https://doi.org/10.3390/ijms26031082

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

Article Metrics

Back to TopTop