We use cookies to improve your experience. By continuing to browse this site, you accept our cookie policy.×
Skip main navigation
Aging Health
Bioelectronics in Medicine
Biomarkers in Medicine
Breast Cancer Management
CNS Oncology
Colorectal Cancer
Concussion
Epigenomics
Future Cardiology
Future Medicine AI
Future Microbiology
Future Neurology
Future Oncology
Future Rare Diseases
Future Virology
Hepatic Oncology
HIV Therapy
Immunotherapy
International Journal of Endocrine Oncology
International Journal of Hematologic Oncology
Journal of 3D Printing in Medicine
Lung Cancer Management
Melanoma Management
Nanomedicine
Neurodegenerative Disease Management
Pain Management
Pediatric Health
Personalized Medicine
Pharmacogenomics
Regenerative Medicine

Pharmacogenomics in psychiatry – the challenge of cytochrome P450 enzyme phenoconversion and solutions to assist precision dosing

    Sam Mostafa

    *Author for correspondence: Tel.: +61 385 820 331;

    E-mail Address: sam.mostafa@monash.edu

    Centre for Medicine Use & Safety, Monash University, Parkville, Victoria, 3052, Australia

    MyDNA Life, Australia Limited, South Yarra, Victoria, Australia

    ,
    Thomas M Polasek

    Centre for Medicine Use & Safety, Monash University, Parkville, Victoria, 3052, Australia

    Certara, Princeton, NJ 08540, USA

    Department of Clinical Pharmacology, Royal Adelaide Hospital, Adelaide, South Australia, 5000, Australia

    ,
    Chad A Bousman

    Department of Psychiatry, Melbourne Neuropsychiatry Centre, University of Melbourne & Melbourne Health, Melbourne, Victoria, 3010, Australia

    The Cooperative Research Centre (CRC) for Mental Health, Carlton, Victoria, 3053, Australia

    Alberta Children's Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, T2N 1N4, Canada

    Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, T2N 1N4, Canada

    Departments of Medical Genetics, Psychiatry, & Physiology & Pharmacology, University of Calgary, Calgary, Alberta, T2N 1N4, Canada

    ,
    Daniel J Müeller

    Pharmacogenetics Research Clinic, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada

    Department of Psychiatry, University of Toronto, Toronto, Ontario, M5T 1R8, Canada

    ,
    Leslie J Sheffield

    MyDNA Life, Australia Limited, South Yarra, Victoria, Australia

    ,
    Joel Rembach

    MyDNA Life, Australia Limited, South Yarra, Victoria, Australia

    &
    Carl MJ Kirkpatrick

    Centre for Medicine Use & Safety, Monash University, Parkville, Victoria, 3052, Australia

    Published Online:https://doi.org/10.2217/pgs-2022-0104

    Abstract

    Pharmacogenomic (PGx) testing of cytochrome P450 (CYP) enzymes may improve the efficacy and/or safety of some medications. This is facilitated by increased availability and affordability of genotyping, the development of clinical practice PGx guidelines and regulatory support. However, the common occurrence of CYP phenoconversion, a mismatch between genotype-predicted CYP phenotype and the actual CYP phenotype, currently limits the application of PGx testing for precision dosing in psychiatry. This review proposes a stepwise approach to assist precision dosing in psychiatry via the introduction of PGx stewardship programs and innovative PGx education strategies. A future perspective on delivering precision dosing for psychiatrists is discussed that involves innovative clinical decision support systems powered by model-informed precision dosing.

    Pharmacogenomic (PGx) testing to assist precision dosing has increased significantly over the past decade. Facilitating factors for PGx include the increased availability and affordability of genotyping, the development of clinical practice PGx guidelines [1,2] and regulatory support [3]. Recommendations from PGx testing about drug and dose selection in psychiatry are primarily based on the genotype-predicted phenotypes of the major drug metabolizing cytochrome P450 (CYP) enzymes [4–8]. However, due to phenoconversion, a central topic of this review article, this does not always provide a reliable prediction of a patient's CYP phenotype [9].

    Indeed, prescribing medications in general – and in psychiatry in particular – is complex because it involves the simultaneous consideration of multiple patient (e.g., age and comorbidities) and nonpatient (e.g., cost) factors that influence the benefit versus risk of pharmacotherapy. Pharmacogenomics can sometimes be used for prescribing medications to improve the likelihood of safer and more effective treatment. Undeniably, PGx testing has advanced the field of precision dosing and increased awareness of its potential clinical and public health benefits [10]. However, PGx results are often presented to clinicians as static reports with only gene–drug guideline recommendations. This is acceptable when a single medication and a single gene are concerned. For example, a CYP genotype may help decide the choice of a particular antidepressant in a young adult with no other comorbidities who is taking no other medications. However, the simple addition of any other agent such as medications that inhibit or induce CYP enzymes may change the benefit versus risk of treatment with that antidepressant. Indeed, as the complexity of psychiatric patients increases with multiple medications metabolized by multiple CYPs with or without environmental effects, such as tobacco smoking, rational prescribing becomes increasingly difficult.

    In the first half of this article, the problem of genotype–phenotype mismatch with CYP enzymes, known as phenoconversion, is outlined, with an emphasis on the medications used in psychiatry. In the second half of the article, solutions to help precision dosing in patients at risk for phenoconversion are suggested. This includes future perspectives on using model-informed precision dosing (MIPD) tools embedded in dynamic clinical decision support systems (CDSS).

    The genotype–phenotype mismatch (phenoconversion)

    Current PGx reports provide static guidance on specific gene–drug pairs. This approach is most relevant when patients are not exposed to factors that significantly alter CYP activities post translation of the CYP genes. However, in some cases, there is a mismatch between the patient's genotype-predicted CYP phenotype and their actual CYP phenotype. This phenomenon is called phenoconversion [11,12]. It has been suggested that phenoconversion offers a plausible explanation for the lack of congruence between CYP genotype and phenotype in many PGx studies [9]. In addition, the combined interaction between patient demographic, environmental and PGx covariates is increasingly recognized as important for predicting drug response [11–14].

    Several factors can result in phenoconversion. These include the use of medications; inflammatory conditions, such as inflammatory bowel disease; cancer and environmental factors that can inhibit or induce CYP enzymes [9,13,14]. First, we consider the clinical impact of using concurrent medications that are capable of causing CYP phenoconversion. This type of drug-induced phenoconversion is more commonly encountered in practice and is dependent on the dose and washout period, determined by the half-life and inhibition/induction mechanism, of the phenoconverting medication and the substrate medication. For example, a patient predicted by genotype to be a normal metabolizer (NM) via CYP2D6 and CYP2C19 may in fact be phenoconverted to a CYP2D6 poor metabolizer (PM) and a CYP2C19 intermediate metabolizer (IM) phenotype by taking a strong CYP2D6 and moderate CYP2C19 inhibitor such as fluoxetine. The CYP2D6 inhibitory effect has been shown to occur as early as 7 days of continuous daily dosing with fluoxetine (20 mg) [15]. A static PGx report will only offer recommendations based on the CYP2D6 and CYP2C19 NM phenotype and will ignore the fluoxetine inhibition of CYP2D6 and CYP2C19. The clinical consequences of the CYP2D6 phenoconversion are:

    • For active drugs (e.g., aripiprazole, risperidone) – higher blood/central nervous system (CNS) concentrations and increased risk of adverse effects.

    • For prodrugs (e.g., codeine) – lower blood/CNS concentration of the active metabolite (morphine) and increased risk of therapeutic failure.

    Phenoconversion of CYP enzymes may also be caused by factors in the patient's lifestyle. For example, smoking induces the expression of CYP1A2, resulting in increased metabolic clearance and decreased concentrations of medications predominantly eliminated by CYP1A2 [16]. In this example, the time course of phenoconversion is dependent on the amount of cigarettes smoked, the turnover of the CYP1A2 enzyme and the half-life of the interacting medication. It is important to note that the turnover of the CYP1A2 enzyme is approximately 2 days [17] and is increased by smoking-induced phenoconversion over a 2- to 4-week period. The increased CYP1A2 enzyme abundance results in more efficient metabolism of the CYP1A2 substrate drug and a shorter half-life. The classic example is clozapine, where smoking-induced CYP1A2 phenoconversion results in increased clozapine dose requirements [18]. On the contrary, smoking cessation can increase clozapine concentrations and the risk of toxicity as the inducing effect of smoking on CYP1A2 is lost over a 2- to 4-week period [18]. Chronic alcohol consumption, a common comorbidity in patients with mental health diagnoses, can also contribute to the phenoconversion of CYP2C19 to a lower metabolic activity level; however, the exact mechanism causing this is not fully elucidated [19]. Regardless, the impact of this CYP2C19-phenoconversion is clinically significant as CYP2C19 is responsible for the metabolism of many psychotropics (Table 1).

    Table 1. Psychotropic medications with actionable pharmacogenomic recommendations in Clinical Pharmacogenetics Implementation Consortium, Dutch Pharmacogenetics Working Group guidelines and the US FDA table of pharmacogenomic associations.
    Psychotropics with actionable pharmacogenomic information
    Antipsychotics
    MedicationClassGene(s)Source(s)
    AripiprazoleAtypical APCYP2D6DPWG, FDA
    BrexpiprazoleAtypical APCYP2D6DPWG, FDA
    ClozapineAtypical APCYP2D6FDA
    HaloperidolTypical APCYP2D6DPWG
    IloperidoneAtypical APCYP2D6FDA
    PerphenazineTypical APCYP2D6FDA
    PimozideTypical APCYP2D6FDA, DPWG
    QuetiapineAtypical APCYP3A4DPWG
    RisperidoneAtypical APCYP2D6DPWG
    ThioridazineTypical APCYP2D6FDA
    ZuclopenthixolTypical APCYP2D6DPWG
    Antidepressants
    MedicationClassGene(s)Source(s)
    VenlafaxineSNRIsCYP2D6DPWG, FDA
    CitalopramSSRIsCYP2C19CPIC, DPWG, FDA
    EscitalopramSSRIsCYP2C19CPIC, DPWG, FDA
    FluvoxamineSSRIsCYP2D6CPIC, DPWG, FDA
    ParoxetineSSRIsCYP2D6CPIC, DPWG, FDA
    SertralineSSRIsCYP2C19CPIC, DPWG, FDA
    AmitriptylineTCAsCYP2C19, CYP2D6CPIC, DPWG, FDA
    AmoxapineTCAsCYP2D6FDA
    ClomipramineTCAsCYP2C19, CYP2D6CPIC, DPWG, FDA
    DesipramineTCAsCYP2D6CPIC, FDA
    DoxepinTCAsCYP2C19, CYP2D6CPIC, DPWG, FDA
    ImipramineTCAsCYP2C19, CYP2D6CPIC, DPWG, FDA
    NortriptylineTCAsCYP2D6CPIC, DPWG, FDA
    ProtriptylineTCAsCYP2D6FDA
    TrimipramineTCAsCYP2C19, CYP2D6CPIC, DPWG, FDA
    VortioxetineOther AntidepressantsCYP2D6FDA
    ADHD medications & anxiolytics
    MedicationClassGene(s)Source(s)
    AmphetaminePsychostimulantsCYP2D6FDA
    AtomoxetineNon-PsychostimulantsCYP2D6CPIC, DPWG, FDA
    DiazepamBenzodiazepinesCYP2C19FDA
    ClobazamBenzodiazepinesCYP2C19FDA

    ADHD: Attention-deficit/hyperactivity disorder; Atypical AP: Atypical antipsychotic; CPIC: Clinical Pharmacogenetics Implementation Consortium; DPWG: Dutch Pharmacogenetics Working Group; SNRI: Serotonin and Noradrenaline Reuptake Inhibitor; SSRI: Selective serotonin reuptake inhibitor; TCA: Tricyclic antidepressant; Typical AP: Typical antipsychotic.

    Changes in physiological or pathophysiological states also cause CYP phenoconversion [13,20]. Indeed, CYP2D6 is induced in pregnancy (e.g., potential phenoconversion of CYP2D6 from the NM phenotype to the ultrarapid metabolizer phenotype), which may lower concentrations of many psychotropics extensively metabolized by CYP2D6 (Table 1). Higher concentrations of cytokines in inflammatory conditions downregulate the expression of CYPs resulting in phenoconversion of genotype predicted NMs to lower metabolizer levels [21]. Patients with cancer classified as CYP2C19 NMs by genotype are more frequently phenoconverted into CYP2C19 PMs compared with non-cancer patients [20]. Although these lifestyle and disease-related phenoconversion factors are important, there are currently no expert consensus guidelines on how to test and quantify the clinical impact of these factors. Therefore, further research is required to guide the implementation of these phenoconversion factors in clinical practice.

    Precision dosing with pharmacogenomics is challenging in psychiatry

    Pharmacogenomics has particular appeal for precision dosing in psychiatry because therapeutic failure is common, adverse effects occur frequently and are sometimes severe, poor adherence and/or discontinuation of treatment is common, healthcare costs for psychiatric patients are high and many psychotropics are metabolized by the highly polymorphic CYP2D6 and CYP2C19 enzymes [22] (Table 1).

    Although PGx testing has particular appeal in psychiatry, the clinical implementation is faced with a number of practical challenges.

    i)

    Adherence: Poor adherence to medication regimens is a widespread problem in psychiatry and may impair the potential of PGx testing to improve treatment outcomes. For example, approximately one in two patients with schizophrenia are either nonadherent or partially adherent to antipsychotics, with adverse effects cited as the main reason [23]. For IMs and PMs (via genotype or phenoconversion) who may have higher drug concentrations on average compared with NMs, an increase in concentration-dependent adverse effects may result in poor adherence. For ultrarapid metabolizers (via genotype or phenoconversion) who may have lower drug concentrations on average compared with NMs, poor efficacy may result in patients ceasing medications due to lack of perceived benefits. In principle, PGx testing may improve adherence indirectly by increasing efficacy and lowering adverse effects, although evidence to support this in practice is needed.

    ii)

    Polypharmacy: Patients with psychiatric disorders often take five or more medications (polypharmacy), increasing their risk of CYP phenoconversion. In an Australian study of more than 2900 patients, predicted CYP phenoconversion increased the frequency of PMs for CYP2D6 (from 5.4 to 24.7%) and CYP2C19 (from 2.7 to 17%), with the majority of patients having PGx testing to guide the prescribing of psychotropics [24]. In a separate analysis of CYP phenoconversion in an acute, aged persons mental health setting [11], 75% of study patients were on polypharmacy. This study also demonstrated that the frequency of predicted CYP phenoconversion at admission and discharge increased for CYP2D6 IMs by 11.7 and 16.1% and for CYP2C19 IMs by 13.1 and 11.7%, respectively.

    iii)

    Poor documentation of medication histories: The fragmented nature of healthcare systems means that medication histories are often inaccurate and poorly communicated between healthcare providers. This makes prescribing and the implementation of pharmacogenomics difficult because it is unclear which medications have been taken previously and the therapeutic outcomes [25]. Take the following case as an example. A patient with a long and poorly documented medication history is prescribed escitalopram by their psychiatrist who reviews their recent PGx report. At the next appointment, the patient's mood has improved, but there is persistent nausea and diarrhea. Importantly, over-the-counter reflux treatment with esomeprazole was started by the patient but never documented because the patient thought it was harmless. Although the static PGx report indicates the patient as a CYP2C19 NM and recommends escitalopram, inhibition of CYP2C19 by esomeprazole can decrease escitalopram clearance and increase escitalopram exposure, resulting in significant gastrointestinal adverse effects.

    iv)

    Lifestyle: The prevalence of smoking in psychiatric patients is twice that of the general population [26]. Polycyclic aromatic hydrocarbons, produced by smoking tobacco, are potent inducers of CYP1A2 [27] and can cause clinically significant drug interactions in psychiatry. For example, CYP1A2 induction by smoking increases clozapine metabolism, resulting in subtherapeutic concentrations and the requirement for higher clozapine doses. Interestingly, this CYP phenoconversion appears to be CYP1A2 genotype specific, occurring mostly in patients with the CYP1A2*1F/*1F genotype [28–33]. The impact of other lifestyle factors that are important in psychiatry, such as diet, illicit drug use and alcohol abuse, is less well understood in terms of potential CYP phenoconversion.

    v)

    Lack of knowledge: There is developing academic research on how to apply phenoconversion to adjust certain genotype-predicted phenotypes for key CYPs. However, to predict the direction and degree of phenoconversion reliably in the presence of multiple phenoconverting drugs with opposing effects (inhibitor vs inducer) is challenging. For example, there is lack of guidelines and evidence in the literature to help reliably predict the phenoconversion of a CYP2C19 IM who is concomitantly taking a CYP2C19 moderate inhibitor (esomeprazole) and inducer (carbamazepine). In addition, more robust research and evaluation is required to also understand the effect of the inhibitor or inducer dose on the degree of phenoconversion because dosage will vary among patients in clinical practice.

    Prescribing tools that assist clinicians with CYP phenoconversion at the point of care are being developed. Sequence2Script, a free web-based tool, requires the CYP genotypes and current medications to be entered before it provides guidance on appropriate PGx-based recommendations for a particular patient [34]. Another example is The PROP Pharmacogenetics Calculator, produced by University of Florida Health [35], which requires the clinician to input the patient's CYP2D6 genotype result and any interacting medication to receive the phenoconverted CYP2D6 phenotype. However, the latter example does not provide the clinician with PGx guideline recommendations based on the phenoconverted CYP2D6 phenotype.

    Proposed solutions to assist precision dosing with pharmacogenomics in psychiatry

    Stepwise approach

    Figure 1 suggests a stepwise approach for implementing PGx testing while considering CYP phenoconversion as part of an advanced precision dosing approach in psychiatry. For a patient prescribed a psychotropic with no actionable PGx recommendations (Step 1), the standard prescribing recommendations based on the medication product label or current prescribing guidelines apply. In a drug-naive patient without any environmental factors causing phenoconversion (e.g., smoking) who is prescribed a psychotropic with actionable PGx recommendations (Step 2), a static PGx report with relevant interpretation and clinical prescribing recommendations could be effectively used. However, if this patient requires pharmacotherapy for other health conditions and one or more of the prescribed medications have actionable PGx recommendations (Step 3), then a clinical decision support system (CDSS) is typically required, supported by clinical review assessing phenoconversion, to provide prescribing guidance for multiple medications. Components required for this stepwise approach are described here and a later section describes Step 4 of Figure 1, in which MIPD could be used to guide dosing in complex patients.

    Figure 1. Improving the implementation of pharmacogenomics into precision dosing in psychiatry.

    CDSS: Clinical decision support system; MIPD: Model-informed precision dosing; PGx: Pharmacogenomic.

    Although Figure 1 may oversimplify the current state of PGx implementation, it is important to note that the PGx and pharmacology knowledge of the prescriber and the PGx readiness of their healthcare system must be considered.

    Improved CDSSs at the point of care

    Point-of-care solutions that efficiently deliver clinically actionable PGx recommendations to clinicians are needed. Recently, CDSSs have been implemented in practice to deliver PGx information [36]. These systems typically provide PGx-based prescribing information to the clinician and flag medications where PGx testing might be indicated.

    Although current CDSSs deliver PGx recommendations at the point of prescribing, they often do not have the ability to adjust recommendations based on phenoconversion. It is impractical for the busy psychiatrist to interrogate separate standalone CDSS and phenoconversion systems to determine which PGx guideline recommendation should be followed. As a first step, contemporary CDSSs should consider integration with validated phenoconversion tools to account for basic drug-induced phenoconversion. Therefore, when a patient's PGx result is available, the CDSS can facilitate a clinically actionable prescribing alert in real time for the psychiatrist.

    The past decade has seen a significant increase in newly discovered CYP alleles, notably CYP2D6 [22], enabled by more advanced genotyping technology with enhanced coverage. Retesting patient samples with more contemporary technologies may result in new CYP2D6 genotypes. Therefore, it is important that CDSSs allow patient PGx results to be updated over time to ensure up-to-date clinical utility of PGx results.

    Although there are examples of CDSSs used successfully to implement PGx testing in practice, a key limitation to their widespread adoption is the lack of standardization in the reporting of PGx data across service providers [37,38]. Ongoing efforts are needed by PGx guideline authorities such as CPIC and DPWG to establish a standardized system for CYP genotype to phenotype translation [39].

    Improved PGx education

    Many surveys show that doctors and pharmacists have positive attitudes toward PGx testing but feel inadequately trained to implement PGx in a clinically meaningful way [40]. While the inclusion of PGx education in the curriculum of tertiary institutions offering pharmacy and medical programs has improved, PGx education for practicing clinicians remains suboptimal. In psychiatry, many consultants lack an understanding about the high variability in CYP activities within phenotypes; prescribers take CYP genotype as an absolute – “you are a CYP2C19 NM and so should be OK to take the standard dose of citalopram”. This interpretation may be problematic if the patient is taking a medication that can inhibit CYP2C19 and result in phenoconversion to a lower level of activity. Professional bodies and continuing professional development accrediting organizations should develop strategies to integrate PGx education into practice via ‘just-in-time’ or ‘on-demand’ learning, which could be delivered via a CDSS. In addition, PGx education can also be embedded in stewardship programs (described next) in which prescribers are educated on the potential benefits and limitations of PGx testing for the medications they routinely prescribe.

    Rollout of PGx stewardship programs

    Pharmacogenomic stewardship programs that review PGx results and the potential for CYP phenoconversion could significantly improve prescribing for at-risk polypharmacy patients [11]. A PGx stewardship team should be multidisciplinary with expertise in medicine, with clinical pharmacology specialist training, pharmacy and PGx. The team would ensure correct diagnoses, that accurate medication histories are taken and maintained and that PGx information is used at the point-of-care. Such an approach would use PGx, medication and lifestyle factors to provide initial first doses, which could then be further adjusted using therapeutic drug monitoring, if required and available. A similar approach was recently endorsed by the Royal College of Physicians and British Pharmacological Society joint working party on personalized prescribing [41]. The Australian government has recently announced that funding will be provided to embed pharmacists in residential aged care facilities to help improve medication safety in older Australians [42]. Indeed, recent data from Australia show the use of psychotropic medicines in older Australians increases after their admission to residential aged care facilities. Embedding pharmacists in these facilities for timely medication reviews could support prescribers with the delivery of PGx, potentially unlocking healthcare benefits for these vulnerable patients. Such initiatives could provide a fertile landscape to implement PGx stewardship programs, offering best-practice clinical governance for the delivery of PGx within precision dosing initiatives.

    Future perspectives: CDSSs powered by MIPD

    MIPD is defined as “the use of computer modelling and simulation to predict a medication dose regimen that is most likely to yield a better benefit to harm balance than traditional dosing, based on individual characteristics” [43]. The combined interaction of patient demographic, environmental and PGx covariates is becoming more widely recognized as important in predicting medication response [43–46]. While catering for the interplay between all of these interaction terms in a CDSS is technically challenging, a CDSS powered by a MIPD platform might offer a pragmatic solution (Figure 2). In this design, the CDSS is responsible for the translation of patient genotype to phenotype, ingesting the MIPD dosage recommendation and delivering a complete PGx recommendation with precision dosing to the electronic medical record (EMR) for use by the psychiatrist. The MIPD platform will require the following data integrations:

    • Patient data from the EMR to provide demographic, disease and medication information.

    • CDSS information (patient genotypes and phenotypes).

    • Data from latest clinical studies and literature (e.g. to provide updated data on enzyme abundance).

    • Environmental and BioSensor information from various devices (e.g. wearables).

    Figure 2. Foundational architecture of a clinical decision support system powered by model-informed precision dosing for delivery of pharmacogenomic information into an electronic medical record.

    CDSS: Clinical decision support system; EMR: Electronic medical record; IoT: Internet of things; MIPD: Model-informed precision dosing.

    The precision dosing recommendation predicted by the modelling can be sent back to the CDSS, which then renders this data into a Fast Healthcare Interoperability Resources (FHIR) file for ingestion by an EMR that is FHIR aware and then integrated into the patient management system for use at the point of prescribing (Figure 2).

    In psychiatry, the proposed CDSS powered by MIPD would apply precision dosing of psychotropics by accounting for the influence of genetic variations in CYPs; the presence of CYP phenoconversion; and coexisting lifestyle (smoking), pregnancy or disease (cancer) factors. Indeed, a recent study of clozapine showed that MIPD using a physiologically based pharmacokinetic model improved the prediction of clozapine's PK compared with population-based statistics [47]. In this study, clozapine concentrations were better predicted by MIPD accounting for the CYP1A2 inducing effect in smokers homozygous for the CYP1A2*1F allele. This is an example of where environmental (smoking) and PGx (CYP1A2 genotype) factors were used to optimize the MIPD model, resulting in improved predictions of clozapine plasma concentrations. In principle, this approach can be applied across other psychotropics, especially those with a high risk of toxicity in overdose (e.g., tricyclic antidepressants). Although there are many technical challenges (some examples are given in Table 2), these preliminary data warrant further investigation with other psychotropics to understand how incorporating gene–environment effects may improve MIPD predictions of pharmacokinetics and whether this translates into meaningful predictions of medication responses. The exact regulatory framework to ensure adequate validation of such models before clinical use is currently being addressed [43].

    Table 2. Model-informed precision dosing technical challenges and proposed solutions.
    ChallengeProposed solutionRef.
    MIPD platforms have complex designs and are targeted for research and academic centersEnable clinician friendly MIPD platforms that are interoperable with existing primary and secondary care patient management systems 
    MIPD platforms have limited validated inhibitor and inducer profilesIncrease the number of validated inhibitor and inducer profiles that are clinically relevant, and include moderate and weak CYP inhibitors and inducers 
    Limited CYP enzyme abundance data based on genotypesUtilize advances in technology (e.g., liquid biopsy) to enrich enzyme abundance data for key CYPs[48]
    CYP: Cytochrome P450; MIPD: Model-informed precision dosing.

    Conclusion

     The potential of PGx testing to assist medication prescribing in psychiatry is limited by CYP phenoconversion. Several factors may cause phenoconversion, but inhibition or induction of CYP2C19 and/or CYP2D6 by concurrently prescribed medications is the main mechanism of CYP phenoconversion in psychiatry. As described in the executive summary, there are mitigation strategies that clinicians and healthcare workers can implement to optimize the choice and dose of medicines. As such, realizing the full potential of PGx testing requires innovative education strategies, particularly at the postgraduate level, PGx stewardship programs embedded in clinical practice, user-friendly CDSS and, in the future, more advanced technologies such as MIPD that aid precision dosing in complex patients.

    Executive summary
    • Adherence, poor documentation of medication histories, polypharmacy and lifestyle factors are key challenges for the implementation of pharmacogenomic (PGx) testing in psychiatry.

    • In addition, phenoconversion can mitigate the interpretation of PGx results.

    • Phenoconversion of drug metabolizing cytochrome P450 (CYP) enzymes can be drug-induced (CYP inhibition or induction), disease-induced (e.g., inflammation, cancer) or caused by environmental factors (e.g., smoking).

    • A stepwise approach to assist precision dosing with PGx testing is suggested, with increasing levels of technical sophistication as patient treatment complexities increases.

    • Rollout of PGx stewardship programs can support practicing clinicians deal with the complex interactions among patient, medication, environment and PGx.

    • PGx continuing professional development programs for psychiatrists can help bridge the current PGx education gap and empower clinicians to realize the benefits of PGx testing.

    • In the future, CDSS powered by MIPD could offer superior medication and dosing options for psychiatrists and their patients compared with current practice.

    Author Contributions

    S Mostafa, TM Polasek, CMJ Kirkpatrick and CA Bousman contributed to the design and conception and review of the manuscript. S Mostafa, TM Polasek and CMJ Kirkpatrick wrote the first draft of the manuscript. J Rembach contributed to the content and design of the CDSS and MIPD section of the manuscript. LJ Sheffield, CA Bousman and DJ Müeller wrote sections of the manuscript. All authors contributed to the manuscript revision, read and approved the submitted version.

    Open access

    This work is licensed under the Creative Commons Attribution 4.0 License. To view a copy of this license, visit http://creativecommons.org/licenses/by/4.0/

    Financial & competing interests disclosure

    S Mostafa, L Sheffield and J Rembach are employees and shareholders of myDNA Inc, a pharmacogenomic testing and interpretation company. TM Polasek provides a consultancy service to Sonic Genetics for the interpretation of pharmacogenomic test results. TM Polasek is an employee of Certara, a company that provides modeling and simulation software and services to the pharmaceutical industry, including a population-based PBPK simulator (Simcyp). CMJ Kirkpatrick was the academic lead on the Certara-Monash Fellowship program funded by MTPConnect. CA Bousman is founder and equity holder of Sequence2Script Inc. and a member of the Clinical Pharmacogenetics Implementation Consortium and the Genetic Testing Committee of the International Society of Psychiatric Genetics. He has also received material support from Assurex, CNSDose, Genomind and AB-Biotics for research purposes and has ongoing research collaborations with MyDNA but does not have equity, stocks, or options in these companies or any other pharmacogenetic companies. The authors have no other relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript apart from those disclosed.

    No writing assistance was utilized in the production of this manuscript.

    References

    • 1. The Dutch Pharmacogenetics Working Group.www.knmp.nl/downloads/farmacogenetica-engels-recommendation-tekst.pdf
    • 2. Clinical Pharmacogenetics Implementation Consortium. https://cpicpgx.org/guidelines/
    • 3. US FDA. Table of pharmacogenetic associations. www.fda.gov/medical-devices/precision-medicine/table-pharmacogenetic-associations
    • 4. Hicks JK, Bishop JR, Sangkuhl K et al. Clinical Pharmacogenetics Implementation Consortium (CPIC) guideline for CYP2D6 and CYP2C19 genotypes and dosing of selective serotonin reuptake inhibitors. Clin. Pharmacol. Ther. 98(2), 127–34 (2015).
    • 5. Hicks JK, Sangkuhl K, Swen JJ et al. Clinical pharmacogenetics implementation consortium guideline (CPIC) for CYP2D6 and CYP2C19 genotypes and dosing of tricyclic antidepressants: 2016 update. Clin. Pharmacol. Ther. 102(1), 37–44 (2016).
    • 6. Crews KR, Monte AA, Huddart R et al. Clinical Pharmacogenetics Implementation Consortium guideline for CYP2D6, OPRM1, and COMT genotypes and select opioid therapy. Clin. Pharmacol. Ther. 110(4), 888–896 (2021).
    • 7. Lima JJ, Thomas CD, Barbarino J et al. Clinical Pharmacogenetics Implementation Consortium (CPIC) guideline for CYP2C19 and proton pump inhibitor dosing. Clin. Pharmacol. Ther. 109(6), 1417–1423 (2020).
    • 8. Theken KN, Lee CR, Gong L et al. Clinical Pharmacogenetics Implementation Consortium (CPIC) guideline for CYP2C9 and nonsteroidal anti-inflammatory drugs. Clin. Pharmacol. Ther. 108(2), 191–200 (2020).
    • 9. Klomp SD, Manson ML, Guchelaar HJ, Swen JJ. Phenoconversion of cytochrome P450 metabolism: a systematic review. J. Clin. Med. 9(9), (2020).
    • 10. Polasek TM, Shakib S, Rostami-Hodjegan A. Precision dosing in clinical medicine: present and future. Expert Rev. Clin. Pharmacol. 11(8), 743–746 (2018).
    • 11. Mostafa S, Polasek TM, Sheffield LJ et al. Quantifying the impact of phenoconversion on medications with actionable pharmacogenomic guideline recommendations in an acute aged persons mental health setting. Front Psychiatry 12, 724170 (2021).
    • 12. Shah RR, Smith RL. Addressing phenoconversion: the Achilles' heel of personalized medicine. Br. J. Clin. Pharmacol. 79(2), 222–240 (2015).
    • 13. Shah RR, Smith RL. Inflammation-induced phenoconversion of polymorphic drug metabolizing enzymes: hypothesis with implications for personalized medicine. Drug Metab. Dispos. 43(3), 400–410 (2015).
    • 14. Storelli F, Matthey A, Lenglet S et al. Impact of CYP2D6 functional allelic variations on phenoconversion and drug–drug interactions. Clin. Pharmacol. Ther. 104(1), 148–157 (2018).
    • 15. Amchin J, Ereshefsky L, Zarycranski W et al. Effect of venlafaxine versus fluoxetine on metabolism of dextromethorphan, a CYP2D6 probe. J. Clin. Pharmacol. 41(4), 443–451 (2001).
    • 16. Kroon LA. Drug interactions with smoking. Am. J. Health Syst. Pharm. 64(18), 1917–1921 (2007).
    • 17. Anderson GD, Chan LN. Pharmacokinetic drug interactions with tobacco, cannabinoids and smoking cessation products. Clin. Pharmacokinet. 55(11), 1353–1368 (2016).
    • 18. de Leon J, Schoretsanitis G, Smith RL et al. An international adult guideline for making clozapine titration safer by using six ancestry-based personalized dosing titrations, CRP, and clozapine levels. Pharmacopsychiatry 55(2), 73–86 (2021).
    • 19. Kiss AF, Vasko D, Deri MT et al. Combination of CYP2C19 genotype with non-genetic factors evoking phenoconversion improves phenotype prediction. Pharmacol. Rep. 70(3), 525–532 (2018).
    • 20. Helsby NA, Lo WY, Sharples K et al. CYP2C19 pharmacogenetics in advanced cancer: compromised function independent of genotype. Br. J. Cancer 99(8), 1251–1255 (2008).
    • 21. Lenoir C, Daali Y, Rollason V et al. Impact of acute inflammation on cytochromes P450 activity assessed by the geneva cocktail. Clin. Pharmacol. Ther. 109(6), 1668–1676 (2021).
    • 22. Pharmacogene Variation Consortium (PharmVar) at (Gaedigk et al. 2018, CPT 103:399; Gaedigk et al. 2019, CPT 105:29). www.PharmVar.org/gene/CYP2D6
    • 23. Kulkarni J, Reeve-Parker K. Psychiatrists' awareness of partial- and non-adherence to antipsychotic medication in schizophrenia: results from the Australian ADHES survey. Australas. Psychiatry 23(3), 258–264 (2015).
    • 24. Mostafa S, Kirkpatrick CMJ, Byron K, Sheffield L. An analysis of allele, genotype and phenotype frequencies, actionable pharmacogenomic (PGx) variants and phenoconversion in 5408 Australian patients genotyped for CYP2D6, CYP2C19, CYP2C9 and VKORC1 genes. J. Neural. Transm. 126(1), 5–18 (2019).
    • 25. Tam VC, Knowles SR, Cornish PL et al. Frequency, type and clinical importance of medication history errors at admission to hospital: a systematic review. CMAJ 173(5), 510–515 (2005).
    • 26. AIHW – Alcohol, tobacco & other drugs in Australia. www.aihw.gov.au/reports/phe/221/alcohol-tobacco-other-drugs-australia/contents/population-groups-of-interest/people-with-mental-health-conditions
    • 27. Zevin S, Benowitz NL. Drug interactions with tobacco smoking. An update. Clin. Pharmacokinet. 36(6), 425–438 (1999).
    • 28. Sanguesa E, Cirujeda C, Concha J et al. Exploring the usefulness of plasma level determination and pharmacogenetics for patients treated with clozapine. Per. Med. 19(3), 181–192 (2022).
    • 29. Ozdemir V, Kalow W, Okey AB et al. Treatment-resistance to clozapine in association with ultrarapid CYP1A2 activity and the C–>A polymorphism in intron 1 of the CYP1A2 gene: effect of grapefruit juice and low-dose fluvoxamine. J. Clin. Psychopharmacol. 21(6), 603–607 (2001).
    • 30. Huang HC, Lua AC, Wu LS et al. Cigarette smoking has a differential effect on the plasma level of clozapine in Taiwanese schizophrenic patients associated with the CYP1A2 gene-163A/C single nucleotide polymorphism. Psychiatr. Genet. 26(4), 172–177 (2016).
    • 31. Sachse C, Brockmoller J, Bauer S, Roots I. Functional significance of a C–>A polymorphism in intron 1 of the cytochrome P450 CYP1A2 gene tested with caffeine. Br. J. Clin. Pharmacol. 47(4), 445–449 (1999).
    • 32. Thorn CF, Muller DJ, Altman RB, Klein TE. PharmGKB summary: clozapine pathway, pharmacokinetics. Pharmacogenet. Genomics 28(9), 214–222 (2018).
    • 33. Kootstra-Ros JE, Smallegoor W, van der Weide J. The cytochrome P450 CYP1A2 genetic polymorphisms *1F and *1D do not affect clozapine clearance in a group of schizophrenic patients. Ann. Clin. Biochem. 42(Pt 3), 216–219 (2005).
    • 34. Bousman CA, Wu P, Aitchison KJ, Cheng T. Sequence2Script: a web-based tool for translation of pharmacogenetic data into evidence-based prescribing recommendations. Front. Pharmacol. 12, 636650 (2021).
    • 35. Cicali EJ, Elchynski AL, Cook KJ et al. How to Integrate CYP2D6 phenoconversion into clinical pharmacogenetics: a tutorial. Clin. Pharmacol. Ther. 110(3), 677–687 (2021).
    • 36. Hinderer M, Boeker M, Wagner SA et al. Integrating clinical decision support systems for pharmacogenomic testing into clinical routine – a scoping review of designs of user-system interactions in recent system development. BMC Med. Inform. Decis. Mak. 17(1), 81 (2017).
    • 37. Caraballo PJ, Bielinski SJ, St Sauver JL, Weinshilboum RM. Electronic medical record-integrated pharmacogenomics and related clinical decision support concepts. Clin. Pharmacol. Ther. 102(2), 254–264 (2017).
    • 38. Blagec K, Swen JJ, Koopmann R et al. Pharmacogenomics decision support in the U-PGx project: results and advice from clinical implementation across seven European countries. PLOS ONE 17(6), e0268534 (2022).
    • 39. Caudle KE, Dunnenberger HM, Freimuth RR et al. Standardizing terms for clinical pharmacogenetic test results: consensus terms from the Clinical Pharmacogenetics Implementation Consortium (CPIC). Genet. Med. 19(2), 215–223 (2017).
    • 40. Karas Kuzelicki N, Prodan Zitnik I, Gurwitz D et al. Pharmacogenomics education in medical and pharmacy schools: conclusions of a global survey. Pharmacogenomics 20(9), 643–657 (2019).
    • 41. Society RCoPaBP. Personalised prescribing. Using pharmacogenomics to improve patient outcomes. A report from the Royal College of Physicians and British Pharmacological Society joint working party (2022).
    • 42. Australian Government, Department of Health. Budget 2022-23. www.health.gov.au/sites/default/files/documents/2022/03/budget-2022-23-residential-aged-care-quality-and-safety-assuring-access-to-multidisciplinary-care-and-maintaining-effective-quality-audits.docx
    • 43. Darwich AS, Polasek TM, Aronson JK et al. Model-informed precision dosing: background, requirements, validation, implementation, and forward trajectory of individualizing drug therapy. Annu. Rev. Pharmacol. Toxicol. 61, 225–245 (2021).
    • 44. Polasek TM, Rostami-Hodjegan A. Virtual twins: Understanding the data required for model-informed precision dosing. Clin. Pharmacol. Ther. 107(4), 742–745 (2020).
    • 45. Polasek TM, Tucker GT, Sorich MJ et al. Prediction of olanzapine exposure in individual patients using physiologically based pharmacokinetic modelling and simulation. Br. J. Clin. Pharmacol. 84(3), 462–476 (2018).
    • 46. Rowland A, van Dyk M, Hopkins AM et al. Physiologically based pharmacokinetic modeling to identify physiological and molecular characteristics driving variability in drug exposure. Clin. Pharmacol. Ther. 104(6), 1219–1228 (2018).
    • 47. Mostafa S, Polasek TM, Bousman CA et al. Delineating gene-environment effects using virtual twins of patients treated with clozapine. Manuscript revision re-submitted to CPT Pharmacometrics Syst Pharmacol on September 8, 2022; copy available on request.
    • 48. Ladumor MK, Thakur A, Sharma S et al. A repository of protein abundance data of drug metabolizing enzymes and transporters for applications in physiologically based pharmacokinetic (PBPK) modelling and simulation. Sci. Rep. 9(1), 9709 (2019).