1
|
Santenna C, Shubham A, Ratinder J, Abhijit R, Tamonud M, Jitendra S, Shamim MA, Balakrishnan S. Drug metabolizing enzymes pharmacogenetic variation-informed antidepressant therapy approach for common mental disorders: A systematic review and meta-analysis. J Affect Disord 2024; 367:832-844. [PMID: 39265864 DOI: 10.1016/j.jad.2024.09.041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2024] [Revised: 08/22/2024] [Accepted: 09/08/2024] [Indexed: 09/14/2024]
Abstract
IMPORTANCE Currently, 30-50 % of individuals with depression and 40 % with anxiety-collectively referred to as common mental disorders (CMDs), exhibit inadequate responses to antidepressant treatments. OBJECTIVE To assess the effectiveness and safety of drug-metabolizing enzyme pharmacogenetic variation informed treatment (PGxIT) versus usual antidepressant treatment (UT) in patients with CMDs. DATA SOURCES A literature search was conducted in the MEDLINE, Scopus, and Cochrane Library databases from inception until January 30, 2024. STUDY SELECTION Studies were selected based on CMD diagnoses, reporting on the genetic variations of drug-metabolizing enzyme (DME) genes in relation to antidepressants, involving PGxIT and UT groups with human subjects, and published in English. DATA EXTRACTION AND SYNTHESIS Data extraction and quality assessment were performed independently by two authors. A pooled risk ratio (RR) with 95 % CI was estimated using both random and fixed-effect models, and heterogeneity was assessed using Cochran's Q test and the I2 statistic. The publication bias of eligible studies was assessed using post hoc Doi plots and the LFK index. RESULTS This systematic review included 18 studies (n = 7021). The PGxIT demonstrated greater efficacy in the remission of symptoms of depressive disorder at 8 weeks (RR 1.523 [95 % CI: 1.255-1.843]; I2 = 48 %) and 12 weeks (RR 1.631 [95 % CI: 1.001-2.657]; I2 = 86 %; p < 0.01), and symptoms of anxiety disorder compared to UT. Additionally, the risk of adverse drug events (ADEs) was significantly lower in the PGxIT group (RR = 0.65 [95 % CI: 0.52-0.82]; I2 = 0 %) than in the UT group. The certainty of evidence for both outcomes was moderate. CONCLUSIONS AND RELEVANCE This systematic review and meta-analysis suggest that pharmacogenetically guided antidepressant treatment, based on genetic variation in drug-metabolizing enzymes, is associated with superior efficacy in the remission of symptoms for patients with depressive disorders and a reduction in ADEs compared to usual treatment and the findings of the systematic review for remission in anxiety disorders indicate that, PGx guided treatment is also associated with increased remission of symptoms in anxiety disorders compared to usual treatment.
Collapse
Affiliation(s)
- Chenchula Santenna
- Department of Pharmacology, All India Institute of Medical Sciences-Bhopal, Madhya Pradesh 462020, India.
| | - Atal Shubham
- Department of Pharmacology, All India Institute of Medical Sciences-Bhopal, Madhya Pradesh 462020, India
| | - Jhaj Ratinder
- Department of Pharmacology, All India Institute of Medical Sciences-Bhopal, Madhya Pradesh 462020, India
| | - Rozatkar Abhijit
- Department of Psychiatry, All India Institute of Medical Sciences-Bhopal, Bhopal 462020, India
| | - Modak Tamonud
- Department of Psychiatry, All India Institute of Medical Sciences-Bhopal, Bhopal 462020, India
| | - Singh Jitendra
- Department of Translational Medicine(,) All India Institute of Medical Sciences-Bhopal, Bhopal 462020, India
| | - Muhammad Aaqib Shamim
- Department of Pharmacology, All India Institute of Medical Sciences - Jodhpur, Jodhpur 342005, India
| | - S Balakrishnan
- Department of Pharmacology, All India Institute of Medical Sciences-Bhopal, Madhya Pradesh 462020, India
| |
Collapse
|
2
|
Lopez-Saavedra J, Abad-Santos F. Cost-effectiveness of pharmacogenetic screening in the management of major depressive disorder in the Spanish Healthcare System. J Affect Disord 2024; 365:597-605. [PMID: 39187185 DOI: 10.1016/j.jad.2024.08.154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 08/15/2024] [Accepted: 08/23/2024] [Indexed: 08/28/2024]
Abstract
BACKGROUND Major depressive disorder (MDD) represents a sizable economic burden in Spain. Pharmacogenetic (PGx) screening to guide the choice of antidepressant medication (ADM) in MDD patients yields higher response and remission rates, which could reduce both healthcare and indirect costs. METHODS We built a cost-effectiveness probabilistic Markov model with microsimulation using Tree Age Pro 2022, simulating a patient cohort from the SNHS starting ADM for MDD, and comparing PGx screening before starting ADM versus no screening (No PGx). We carried out a probabilistic sensitivity analysis using the Monte Carlo simulation with microsimulation, set for 1000 iterations and 1000 microsimulation trials, both from societal and healthcare provider perspectives, for a time horizon of 3 years. RESULTS From a societal perspective, the model estimated a mean cost of 3172.85€ and effectiveness of 2.64 quality-adjusted life years (QALYs) for the No PGx strategy, and a mean cost of 1687.02€ and effectiveness of 2.84 QALYs for the PGx strategy. The mean ICER was -7820.56 €/QALY. From a healthcare provider perspective (no indirect costs considered), the mean cost was 662.62€ for the No PGx strategy, and 446.60€ for the PGx strategy. The mean ICER was -1130.16 €/QALY. LIMITATIONS The heterogeneity of input data from the literature, the need for assumptions of homogeneous distribution of variables and events across population and time, and the inherent limitations of cost-effectiveness analysis should be considered. The model omits combined therapies (ADMs with mood stabilizers, antipsychotics, cognitive behavioral therapy…). CONCLUSIONS PGx screening in MDD prior to ADM start is a dominant strategy in the SNHS.
Collapse
Affiliation(s)
- Javier Lopez-Saavedra
- Clinical Pharmacology Department, Hospital Universitario de La Princesa, Instituto de Investigación Sanitaria La Princesa (IIS-Princesa), Faculty of Medicine, Universidad Autónoma de Madrid (UAM), 28029 Madrid, Spain
| | - Francisco Abad-Santos
- Clinical Pharmacology Department, Hospital Universitario de La Princesa, Instituto de Investigación Sanitaria La Princesa (IIS-Princesa), Faculty of Medicine, Universidad Autónoma de Madrid (UAM), 28029 Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, 28029 Madrid, Spain.
| |
Collapse
|
3
|
Patel JN, Morris SA, Torres R, Rhead B, Vlangos C, Mueller DJ, Brown LC, Lefkofsky H, Ali M, De La Vega FM, Barnes KC, Zoghbi A, Stanton JD, Badgeley MA. Pharmacogenomic insights in psychiatric care: uncovering novel actionability, allele-specific CYP2D6 copy number variation, and phenoconversion in 15,000 patients. Mol Psychiatry 2024; 29:3495-3502. [PMID: 38783055 PMCID: PMC11541190 DOI: 10.1038/s41380-024-02588-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 04/19/2024] [Accepted: 04/30/2024] [Indexed: 05/25/2024]
Abstract
Pharmacogenomic testing has emerged as an aid in clinical decision making for psychiatric providers, but more data is needed regarding its utility in clinical practice and potential impact on patient care. In this cross-sectional study, we determined the real-world prevalence of pharmacogenomic actionability in patients receiving psychiatric care. Potential actionability was based on the prevalence of CYP2C19 and CYP2D6 phenotypes, including CYP2D6 allele-specific copy number variations (CNVs). Combined actionability additionally incorporated CYP2D6 phenoconversion and the novel CYP2C-TG haplotype in patients with available medication data. Across 15,000 patients receiving clinical pharmacogenomic testing, 65% had potentially actionable CYP2D6 and CYP2C19 phenotypes, and phenotype assignment was impacted by CYP2D6 allele-specific CNVs in 2% of all patients. Of 4114 patients with medication data, 42% had CYP2D6 phenoconversion from drug interactions and 20% carried a novel CYP2C haplotype potentially altering actionability. A total of 87% had some form of potential actionability from genetic findings and/or phenoconversion. Genetic variation detected via next-generation sequencing led to phenotype reassignment in 22% of individuals overall (2% in CYP2D6 and 20% in CYP2C19). Ultimately, pharmacogenomic testing using next-generation sequencing identified potential actionability in most patients receiving psychiatric care. Early pharmacogenomic testing may provide actionable insights to aid clinicians in drug prescribing to optimize psychiatric care.
Collapse
Affiliation(s)
- Jai N Patel
- Department of Cancer Pharmacology & Pharmacogenomics, Levine Cancer Institute, Atrium Health, Charlotte, NC, USA
| | - Sarah A Morris
- Department of Cancer Pharmacology & Pharmacogenomics, Levine Cancer Institute, Atrium Health, Charlotte, NC, USA
| | | | | | | | - Daniel J Mueller
- Pharmacogenetics Research Clinic, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON, Canada
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada
| | | | | | | | | | | | - Anthony Zoghbi
- Department of Psychiatry and Behavioral Sciences, Baylor College of Medicine, Houston, TX, USA
| | | | | |
Collapse
|
4
|
Norris ML. Exploring biologically oriented precision mental health initiatives for the care of patients with eating disorders: A narrative review. EUROPEAN EATING DISORDERS REVIEW 2024; 32:1117-1137. [PMID: 38867415 DOI: 10.1002/erv.3114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 05/08/2024] [Accepted: 05/30/2024] [Indexed: 06/14/2024]
Abstract
OBJECTIVE Eating disorders (EDs) represent a major public health burden. Increasingly, studies suggest mental health (MH) fields are failing to improve the effectiveness of treatments and that alternative models of care must be considered. Precision mental health (PMH) seeks to tailor treatment to individual needs and relies on a comprehensive understanding of the neurobiological and physiological underpinnings of mental illness. METHODS In this narrative review, published literature with focus on biological application of PMH strategies for EDs is reviewed and summarised. RESULTS A total of 39 articles were retained for the review covering a variety of themes with relevance to PMH. Many studies of biological markers with PMH applicability focused on anorexia nervosa. Although a variety of potential PMH research applications were identified, the review failed to identify any evidence of implementation into routine ED practice. CONCLUSIONS Despite the theoretical merit of biological application of PMH in ED treatment, clinical applications for standard practice are lacking. There is a need to invest further in studies that seek to identify biological markers and investigate neurobiological underpinnings of disease in hopes of targeting and developing treatments that can be better tailored to the individualised needs of patients.
Collapse
Affiliation(s)
- Mark L Norris
- Division of Adolescent Medicine, Children's Hospital of Eastern Ontario, University of Ottawa, Ottawa, Ontario, Canada
- Children's Hospital of Eastern Ontario Research Institute, Ottawa, Ontario, Canada
| |
Collapse
|
5
|
Medwid S, Schwarz UI, Choi YH, Keller D, Ross C, Kim RB. Solanidine Metabolites as Diet-Derived Biomarkers of CYP2D6-Mediated Tamoxifen Metabolism in Breast Cancer Patients. Clin Pharmacol Ther 2024; 116:1269-1277. [PMID: 39039708 DOI: 10.1002/cpt.3380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 07/08/2024] [Indexed: 07/24/2024]
Abstract
Tamoxifen is an important antiestrogen for the treatment of hormone receptor-positive breast cancer and undergoes bioactivation by CYP2D6 to its active metabolite endoxifen. Genetic variation in CYP2D6 has been linked to endoxifen levels during tamoxifen therapy. Recent studies have suggested solanidine, a glycoalkaloid phytochemical in potatoes, undergoes CYP2D6-mediated metabolism to 4-OH-solanidine (m/z 414) and 3,4-seco-solanidine-3,4-dioic acid (SSDA; m/z 444). Using a retrospective cohort of 1,032 breast cancer patients on tamoxifen therapy, we examined the association of solanidine metabolites with CYP2D6 activity and its correlation with tamoxifen metabolism. Solanidine, 4-OH-solanidine, or SSDA was detected in 99.7% (N = 1,029) of plasma samples. Decreased solanidine metabolite ratios were found in CYP2D6 intermediate and poor metabolizers (P < 0.0001). Patients on CYP2D6 strong inhibitors had a 77.6% and 94.2% decrease in 4-OH-solandine/solanidine (P < 0.0001) and SSDA/solanidine (P < 0.0001), respectively. The ratio of endoxifen to tamoxifen was highly correlated with both 4-OH-solandine/solanidine (ρ = 0.3207, P < 0.0001) and SSDA/solanidine (ρ = 0.5022, P < 0.0001) ratios. Logistic regression modeling was used to determine that 4-OH-solanidine/solanidine and SSDA/solanidine ratios below 2.1 and 0.8, respectively, predicted endoxifen concentrations of <16 nM. In conclusion, solanidine, 4-OH-solanidine, and SSDA are diet-derived biomarkers of CYP2D6 activity. Moreover, in patients on tamoxifen therapy, 4-OH-solanidine/solanidine and SSDA/solanidine predicted endoxifen levels including the inhibitory effects of concomitantly prescribed CYP2D6-interacting medications. Accordingly, 4-OH-solanidine/solanidine or SSDA/solanidine ratio has the potential to be particularly useful prior to initiation of tamoxifen or for determining the impact of CYP2D6 drug interactions, as well as prior to switching from an aromatase inhibitor to tamoxifen.
Collapse
Affiliation(s)
- Samantha Medwid
- Department of Medicine, Western University, London, Ontario, Canada
| | - Ute I Schwarz
- Department of Medicine, Western University, London, Ontario, Canada
- Department of Physiology and Pharmacology, Western University, London, Ontario, Canada
| | - Yun-Hee Choi
- Department of Epidemiology and Biostatistics, Western University, London, Ontario, Canada
| | - Denise Keller
- London Health Sciences Centre, London, Ontario, Canada
| | - Cameron Ross
- Department of Medicine, Western University, London, Ontario, Canada
| | - Richard B Kim
- Department of Medicine, Western University, London, Ontario, Canada
- Department of Physiology and Pharmacology, Western University, London, Ontario, Canada
- London Health Sciences Centre, London, Ontario, Canada
- Lawson Health Research Institute, London, Ontario, Canada
| |
Collapse
|
6
|
Stimpfl JN, Walkup JT, Robb AS, Alford AE, Stahl SM, McCracken JT, Stancil SL, Ramsey LB, Emslie GJ, Strawn JR. Deprescribing Antidepressants in Children and Adolescents: A Systematic Review of Discontinuation Approaches, Cross-Titration, and Withdrawal Symptoms. J Child Adolesc Psychopharmacol 2024. [PMID: 39469761 DOI: 10.1089/cap.2024.0099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/30/2024]
Abstract
Background: Antidepressant medications, including selective serotonin reuptake inhibitors (SSRIs) and serotonin-norepinephrine reuptake inhibitors (SNRIs), are commonly used to treat depressive, anxiety, and obsessive-compulsive disorders in youth. Yet, data on discontinuing these medications, withdrawal symptoms, and strategies to switch between them are limited. Methods: We searched PubMed and ClinicalTrials.gov through June 1, 2024, to identify randomized controlled trials assessing antidepressant discontinuation in youth. We summarized pediatric pharmacokinetic data to inform tapering and cross-titration strategies for antidepressants and synthesized these data with reports of antidepressant withdrawal. Results: Our search identified 528 published articles, of which 28 were included. In addition, 19 records were obtained through other methods, with 14 included. The corpus of records included 13 randomized, double-blind, placebo-controlled trials (3026 patients), including SSRIs (K = 10), SNRIs (K = 4), and TCAs (K = 1), ranging from 4 to 35 weeks. Deprescribing antidepressants requires considering clinical status, treatment response, and, in cross-titration cases, the pharmacokinetics and pharmacodynamics of both medications. Antidepressant withdrawal symptoms are related to the pharmacokinetics of the medication, which vary across antidepressants and may include irritability, palpitations, anxiety, nausea, sweating, headaches, insomnia, paresthesia, and dizziness. These symptoms putatively involve changes in serotonin transporter expression and receptor sensitivity, impacting the serotonin, dopamine, and norepinephrine pathways. Conclusions: Although approaches to deprescribing antidepressants in pediatric patients are frequently empirically guided, accumulating data related to the course of relapse and withdrawal symptoms, as well as the pharmacokinetic and pharmacodynamic properties of medications, should inform these approaches. Recommendations within this review support data-informed discussions of deprescribing-including when and how-that are critically important in the clinician-family-patient relationship.
Collapse
Affiliation(s)
- Julia N Stimpfl
- Department of Psychiatry & Behavioral Neuroscience, College of Medicine, University of Cincinnati, Cincinnati, Ohio, USA
| | - John T Walkup
- Pritzker Department of Psychiatry and Behavioral Health, Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois, USA
| | - Adelaide S Robb
- Department of Psychiatry and Behavioral Sciences, Children's National Hospital, George Washington University School of Medicine, Washington DC, USA
| | - Alexandra E Alford
- Department of Psychiatry & Behavioral Neuroscience, College of Medicine, University of Cincinnati, Cincinnati, Ohio, USA
| | - Stephen M Stahl
- Department of Psychiatry, University of California, San Diego, California, USA
| | - James T McCracken
- Department of Psychiatry, University of California, San Francisco, California, USA
| | - Stephani L Stancil
- Department of Pediatrics, Schools of Medicine, University of Missouri-Kansas City, Kansas City, Missouri, USA
- Division of Clinical Pharmacology, Toxicology & Therapeutic Innovation, Children's Mercy Kansas City, Kansas City, Missouri, USA
| | - Laura B Ramsey
- Department of Psychiatry & Behavioral Neuroscience, College of Medicine, University of Cincinnati, Cincinnati, Ohio, USA
- Department of Pediatrics, Schools of Medicine, University of Missouri-Kansas City, Kansas City, Missouri, USA
- Division of Clinical Pharmacology, Toxicology & Therapeutic Innovation, Children's Mercy Kansas City, Kansas City, Missouri, USA
| | - Graham J Emslie
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Jeffrey R Strawn
- Department of Psychiatry & Behavioral Neuroscience, College of Medicine, University of Cincinnati, Cincinnati, Ohio, USA
| |
Collapse
|
7
|
Eckert AP, Hahn M. [Pharmacogenetic testing to optimize psychopharmacotherapy: Case study of a patient with severe depression and lack of treatment success due to genetic polymorphisms for CYP2D6 and CYP2C19]. FORTSCHRITTE DER NEUROLOGIE-PSYCHIATRIE 2024. [PMID: 39406367 DOI: 10.1055/a-2405-5037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/01/2024]
Affiliation(s)
- Andreas Philipp Eckert
- Klinik für Psychiatrie, Psychosomatik und Psychotherapie, Universitätsklinikum Frankfurt Klinik für Psychiatrie, Frankfurt am Main, Germany
| | - Martina Hahn
- Klinik für Psychiatrie, Psychosomatik und Psychotherapie, Universitätsklinikum Frankfurt Klinik für Psychiatrie, Frankfurt am Main, Germany
- Klinik für psychische Gesundheit, varisano Klinikum Frankfurt Hoechst, Frankfurt, Germany
| |
Collapse
|
8
|
da Rocha Zurchimitten G, Camerini L, Izídio GS, Ghisleni G. Identifying genetic variants associated with side effects of antidepressant treatment: A systematic review. Prog Neuropsychopharmacol Biol Psychiatry 2024; 136:111154. [PMID: 39369809 DOI: 10.1016/j.pnpbp.2024.111154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Revised: 09/17/2024] [Accepted: 09/24/2024] [Indexed: 10/08/2024]
Abstract
Major Depressive Disorder (MDD) is one of the most prevalent neurobiological disorders globally. Antidepressant medications are the first-line treatment for managing symptoms. However, over time, pharmacotherapy has been linked to several challenges, primarily due to the wide array of side effects that often reduce patient adherence to treatment. The literature suggests that these side effects may be influenced by polymorphisms in genes related to the pharmacokinetics and pharmacodynamics of antidepressants. Thus, this systematic review aimed to identify studies that investigated the association between genetic variants and side effects resulting from antidepressant treatment in individuals with MDD. Original articles indexed in the electronic databases Cochrane Library, EMBASE, MEDLINE via PubMed, and Scopus were identified. A total of 55 studies were included in the review, and data regarding the outcomes of interest were extracted. Due to the exploratory nature of the review, a narrative/descriptive synthesis of the results was performed. The risk of bias was evaluated using the Joanna Briggs Institute's tools, tailored to the design of each study. Polymorphisms in 35 genes were statistically associated with the development of side effects. A subsequent Protein-Protein Interaction Network analysis helped elucidate the key biological pathways involved in antidepressant side effects, with a view toward exploring the potential application of pharmacogenetic markers in clinical practice.
Collapse
Affiliation(s)
| | - Laísa Camerini
- Postgraduate Program in Epidemiology, Federal University of Pelotas, Rio Grande do Sul, Brazil
| | - Geison Souza Izídio
- Postgraduate Program in Pharmacology, Federal University of Santa Catarina, Santa Catarina, Brazil
| | - Gabriele Ghisleni
- Postgraduate Program in Health and Behavior, Catholic University of Pelotas, Rio Grande do Sul, Brazil.
| |
Collapse
|
9
|
Litonius K, Kulla N, Falkenbach P, Kristiansson K, Tarkiainen EK, Ukkola-Vuoti L, Cajanus K, Korhonen M, Khan S, Sistonen J, Orpana A, Lindstedt M, Nyrönen T, Perola M, Turpeinen M, Kytö V, Tornio A, Niemi M. Value of Pharmacogenetic Testing Assessed with Real-World Drug Utilization and Genotype Data. Clin Pharmacol Ther 2024. [PMID: 39365028 DOI: 10.1002/cpt.3458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2024] [Accepted: 09/03/2024] [Indexed: 10/05/2024]
Abstract
Implementation of pharmacogenetic testing in clinical care has been slow and with few exceptions is hindered by the lack of real-world evidence on how to best target testing. In this retrospective register-based study, we analyzed a nationwide cohort of 1,425,000 patients discharged from internal medicine or surgical wards and a cohort of 2,178 university hospital patients for purchases and prescriptions of pharmacogenetically actionable drugs. Pharmacogenetic variants were obtained from whole genome genotype data for a subset (n = 930) of the university hospital patients. We investigated factors associated with receiving pharmacogenetically actionable drugs and developed a literature-based cost-benefit model for pre-emptive pharmacogenetic panel testing. In a 2-year follow-up, 60.4% of the patients in the nationwide cohort purchased at least one pharmacogenetically actionable drug, most commonly ibuprofen (25.0%) and codeine (19.4%). Of the genotyped subset, 98.8% carried at least one actionable pharmacogenetic genotype and 23.3% had at least one actionable gene-drug pair. Patients suffering from musculoskeletal or cardiovascular diseases were more prone to receive pharmacogenetically actionable drugs during inpatient episode. The cost-benefit model included frequently dispensed drugs in the university hospital cohort, comprising ondansetron (19.4%), simvastatin (7.4%), clopidogrel (5.0%), warfarin (5.1%), (es)citalopram (5.3%), and azathioprine (0.5%). For untargeted pre-emptive pharmacogenetic testing of all university hospital patients, the model indicated saving €17.49 in direct healthcare system costs per patient in 2 years without accounting for the cost of the test itself. Therefore, it might be reasonable to target pre-emptive pharmacogenetic testing to patient groups most likely to receive pharmacogenetically actionable drugs.
Collapse
Affiliation(s)
- Kaisa Litonius
- Department of Clinical Pharmacology, University of Helsinki, Helsinki, Finland
- Department of Clinical Pharmacology, HUS Diagnostic Center, Helsinki University Hospital, Helsinki, Finland
- Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Noora Kulla
- Integrative Physiology and Pharmacology, Institute of Biomedicine, University of Turku, Turku, Finland
- Unit of Clinical Pharmacology, Turku University Hospital, Turku, Finland
| | - Petra Falkenbach
- Finnish Coordinating Center for Health Technology Assessment, Oulu University Hospital, University of Oulu, Oulu, Finland
| | | | - E Katriina Tarkiainen
- Department of Clinical Pharmacology, University of Helsinki, Helsinki, Finland
- Department of Clinical Pharmacology, HUS Diagnostic Center, Helsinki University Hospital, Helsinki, Finland
- Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | | | - Kristiina Cajanus
- Integrative Physiology and Pharmacology, Institute of Biomedicine, University of Turku, Turku, Finland
- Unit of Clinical Pharmacology, Turku University Hospital, Turku, Finland
| | - Mari Korhonen
- Genome Unit, HUS Diagnostic Center, Helsinki University Hospital, Helsinki, Finland
| | - Sofia Khan
- Genome Unit, HUS Diagnostic Center, Helsinki University Hospital, Helsinki, Finland
| | - Johanna Sistonen
- Genome Unit, HUS Diagnostic Center, Helsinki University Hospital, Helsinki, Finland
| | - Arto Orpana
- Genome Unit, HUS Diagnostic Center, Helsinki University Hospital, Helsinki, Finland
| | | | | | - Markus Perola
- Finnish Institute for Health and Welfare, Helsinki, Finland
- Clinical and Molecular Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Miia Turpeinen
- Finnish Coordinating Center for Health Technology Assessment, Oulu University Hospital, University of Oulu, Oulu, Finland
- Medical Research Center Oulu, Oulu University Hospital, University of Oulu, Oulu, Finland
| | - Ville Kytö
- Heart Center, Turku University Hospital, University of Turku, Turku, Finland
- Clinical Research Center, Turku University Hospital, Turku, Finland
| | - Aleksi Tornio
- Integrative Physiology and Pharmacology, Institute of Biomedicine, University of Turku, Turku, Finland
- Unit of Clinical Pharmacology, Turku University Hospital, Turku, Finland
| | - Mikko Niemi
- Department of Clinical Pharmacology, University of Helsinki, Helsinki, Finland
- Department of Clinical Pharmacology, HUS Diagnostic Center, Helsinki University Hospital, Helsinki, Finland
- Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| |
Collapse
|
10
|
Shugg T, Tillman EM, Breman AM, Hodge JC, McDonald CA, Ly RC, Rowe EJ, Osei W, Smith TB, Schwartz PH, Callaghan JT, Pratt VM, Lynch S, Eadon MT, Skaar TC. Development of a Multifaceted Program for Pharmacogenetics Adoption at an Academic Medical Center: Practical Considerations and Lessons Learned. Clin Pharmacol Ther 2024; 116:914-931. [PMID: 39169556 PMCID: PMC11452286 DOI: 10.1002/cpt.3402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Accepted: 07/25/2024] [Indexed: 08/23/2024]
Abstract
In 2019, Indiana University launched the Precision Health Initiative to enhance the institutional adoption of precision medicine, including pharmacogenetics (PGx) implementation, at university-affiliated practice sites across Indiana. The overarching goal of this PGx implementation program was to facilitate the sustainable adoption of genotype-guided prescribing into routine clinical care. To accomplish this goal, we pursued the following specific objectives: (i) to integrate PGx testing into existing healthcare system processes; (ii) to implement drug-gene pairs with high-level evidence and educate providers and pharmacists on established clinical management recommendations; (iii) to engage key stakeholders, including patients to optimize the return of results for PGx testing; (iv) to reduce health disparities through the targeted inclusion of underrepresented populations; (v) and to track third-party reimbursement. This tutorial details our multifaceted PGx implementation program, including descriptions of our interventions, the critical challenges faced, and the major program successes. By describing our experience, we aim to assist other clinical teams in achieving sustainable PGx implementation in their health systems.
Collapse
Affiliation(s)
- Tyler Shugg
- Division of Clinical Pharmacology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Emma M. Tillman
- Division of Clinical Pharmacology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Amy M. Breman
- Division of Diagnostic Genomics, Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Jennelle C. Hodge
- Division of Diagnostic Genomics, Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Christine A. McDonald
- Division of Clinical Pharmacology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Reynold C. Ly
- Division of Diagnostic Genomics, Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Elizabeth J. Rowe
- Division of Clinical Pharmacology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Wilberforce Osei
- Division of Clinical Pharmacology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Tayler B. Smith
- Division of Clinical Pharmacology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Peter H. Schwartz
- Division of General Internal Medicine and Geriatrics, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - John T. Callaghan
- Division of Clinical Pharmacology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Victoria M. Pratt
- Division of Diagnostic Genomics, Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Sheryl Lynch
- Division of Clinical Pharmacology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Michael T. Eadon
- Division of Clinical Pharmacology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Division of Nephrology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Todd C. Skaar
- Division of Clinical Pharmacology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, Indiana, USA
| |
Collapse
|
11
|
Sakon CM, Sales C, Mertami S, Raibulet A, Schulte RR, Slaven JE, Tillman EM. Utilization of supportive care medications and opportunities for pre-emptive pharmacogenomic testing in pediatric and young adults with leukemia. Pediatr Hematol Oncol 2024; 41:480-488. [PMID: 38904214 PMCID: PMC11410515 DOI: 10.1080/08880018.2024.2368007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 05/21/2024] [Accepted: 06/10/2024] [Indexed: 06/22/2024]
Abstract
This study aimed to evaluate the utilization of drugs with pharmacogenomic guidelines (PGx-drugs) for personalized dosing in pediatric leukemia. A retrospective observational study of pediatric leukemia patients admitted between 2009-2019 at a single-center academic children's hospital was conducted to determine PGx-drug exposure within 3 years of diagnosis. Along with baseline demographic and clinical characteristics of these patients, data regarding dates of diagnosis, relapse, death were collected. During the study period, inclusion criteria were met by 714 patients. The most frequently given medications were ondansetron (96.1%), morphine (92.2%), and allopurinol (85.3%) during the study period. In this cohort, 82% of patients received five or more PGx-drugs. Patients diagnosed with acute myeloid leukemia and leukemia unspecified were prescribed more PGx-drugs than other types of leukemia. There was a significant relationship between age at diagnosis and the number of PGx-drugs prescribed. Adolescents and adults both received a median of 10 PGx-drugs, children received a median of 6 PGx-drugs, and infants received a median of 7 PGx-drugs (p < 0.001). Patients with recurrent leukemia had significantly more PGx-drugs prescribed compared to those without recurrent disease, 10 drugs and 6 drugs, respectively (p < 0.001). Patients diagnosed with childhood leukemia are high utilizers of PGx-drugs. There is a vital need to understand how PGx testing may be utilized to optimize treatment and enhance quality of life. Preemptive PGx testing is a tool that aids in optimization of drug therapy and decreases the need for later treatment modifications. This can result in financial savings from decreased health-care encounters.
Collapse
Affiliation(s)
| | - Carmina Sales
- Purdue University College of Pharmacy, Purdue University
| | - Selsbiel Mertami
- Division of Clinical Pharmacology, Indiana University School of Medicine (IUSM)
| | - Andra Raibulet
- College of Pharmacy and Health Sciences, Butler University
| | - Rachael R Schulte
- Department of Pediatrics, Division of Pediatric Hematology/Oncology/Stem Cell Transplant, IUSM
| | | | - Emma M. Tillman
- Division of Clinical Pharmacology, Indiana University School of Medicine (IUSM)
| |
Collapse
|
12
|
Halman A, Conyers R, Moore C, Khatri D, Sarris J, Perkins D. Harnessing Pharmacogenomics in Clinical Research on Psychedelic-Assisted Therapy. Clin Pharmacol Ther 2024. [PMID: 39345195 DOI: 10.1002/cpt.3459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Accepted: 09/18/2024] [Indexed: 10/01/2024]
Abstract
Psychedelics have recently re-emerged as potential treatments for various psychiatric conditions that impose major public health costs and for which current treatment options have limited efficacy. At the same time, personalized medicine is increasingly being implemented in psychiatry to provide individualized drug dosing recommendations based on genetics. This review brings together these topics to explore the utility of pharmacogenomics (a key component of personalized medicine) in psychedelic-assisted therapies. We summarized the literature and explored the potential implications of genetic variability on the pharmacodynamics and pharmacokinetics of psychedelic drugs including lysergic acid diethylamide (LSD), psilocybin, N,N-dimethyltryptamine (DMT), 5-methoxy-N,N-dimethyltryptamine (5-MeO-DMT), ibogaine and 3,4-methylenedioxymethamphetamine (MDMA). Although existing evidence is limited, particularly concerning pharmacodynamics, studies investigating pharmacokinetics indicate that genetic variants in drug-metabolizing enzymes, such as cytochrome P450, impact the intensity of acute psychedelic effects for LSD and ibogaine, and that a dose reduction for CYP2D6 poor metabolizers may be appropriate. Furthermore, based on the preclinical evidence, it can be hypothesized that CYP2D6 metabolizer status might contribute to altered acute psychedelic experiences with 5-MeO-DMT and psilocybin when combined with monoamine oxidase inhibitors. In conclusion, considering early evidence that genetic factors can influence the effects of certain psychedelics, we suggest that pharmacogenomic testing should be further investigated in clinical research. This is necessary to evaluate its utility in improving the safety and therapeutic profile of psychedelic therapies and a potential future role in personalizing psychedelic-assisted therapies, should these treatments become available.
Collapse
Affiliation(s)
- Andreas Halman
- Psychae Therapeutics, Melbourne, Victoria, Australia
- Cancer Therapies, Stem Cell Medicine, Murdoch Children's Research Institute, Parkville, Victoria, Australia
- School of Population and Global Health, The University of Melbourne, Melbourne, Victoria, Australia
| | - Rachel Conyers
- Cancer Therapies, Stem Cell Medicine, Murdoch Children's Research Institute, Parkville, Victoria, Australia
- Department of Paediatrics, The University of Melbourne, Melbourne, Victoria, Australia
| | - Claire Moore
- Cancer Therapies, Stem Cell Medicine, Murdoch Children's Research Institute, Parkville, Victoria, Australia
- Department of Paediatrics, The University of Melbourne, Melbourne, Victoria, Australia
| | - Dhrita Khatri
- Cancer Therapies, Stem Cell Medicine, Murdoch Children's Research Institute, Parkville, Victoria, Australia
| | - Jerome Sarris
- Psychae Therapeutics, Melbourne, Victoria, Australia
- Centre for Mental Health, Swinburne University, Melbourne, Victoria, Australia
- NICM Health Research Institute, Western Sydney University, Westmead, New South Wales, Australia
- The Florey Institute of Neuroscience and Mental Health & The Department of Psychiatry, Melbourne University, Melbourne, Victoria, Australia
| | - Daniel Perkins
- Psychae Therapeutics, Melbourne, Victoria, Australia
- School of Population and Global Health, The University of Melbourne, Melbourne, Victoria, Australia
- Centre for Mental Health, Swinburne University, Melbourne, Victoria, Australia
| |
Collapse
|
13
|
Kaptsis D, Lewis M, Sorich M, Battersby M. Long-read sequencing of CYP2D6 may improve psychotropic prescribing and treatment outcomes: A systematic review and meta-analysis. J Psychopharmacol 2024; 38:771-783. [PMID: 39262167 PMCID: PMC11447996 DOI: 10.1177/02698811241268899] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/13/2024]
Abstract
BACKGROUND The enzyme expression (i.e. phenotype) of the Cytochrome P450 2D6 (CYP2D6) gene is highly relevant to the metabolism of psychotropic medications, and therefore to precision medicine (i.e. personalised prescribing). AIMS This review aims to assess the improvement in CYP2D6 phenotyping sensitivity (IPS) and accuracy (IPA) offered by long-read sequencing (LRS), a new genetic testing technology. METHODS Human DNA samples that underwent LRS genotyping of CYP2D6 in published, peer-reviewed clinical research were eligible for inclusion. A systematic literature search was conducted until 30 September 2023. CYP2D6 genotypes were translated into phenotypes using the international consensus method. IPS was the percentage of non-normal LRS CYP2D6 phenotypes undetectable with FDA-approved testing (AmpliChip). IPA was the percentage of LRS CYP2D6 phenotypes mischaracterised by non-LRS genetic tests (for samples with LRS and non-LRS data). RESULTS Six studies and 1411 samples were included. In a meta-analysis of four studies, IPS was 10% overall (95% CI = (2, 18); n = 1385), 20% amongst Oceanians (95% CI = (17, 23); n = 582) and 2% amongst Europeans (95% CI = (1, 4); n = 803). IPA was 4% in a large European cohort (95% CI = (2, 7); n = 567). When LRS was used selectively (e.g. for novel or complex CYP2D6 genotypes), very high figures were observed for IPS (e.g. 88%; 95% CI = (72, 100); n = 17; country = Japan) and IPA (e.g. 76%; 95% CI = (55, 98); n = 17; country = Japan). CONCLUSIONS LRS improves CYP2D6 phenotyping compared to established genetic tests, particularly amongst Oceanian and Japanese individuals, and those with novel or complex genotypes. LRS may therefore assist in optimising personalised prescribing of psychotropic medications. Further research is needed to determine associated clinical benefits, such as increased medication safety and efficacy.
Collapse
Affiliation(s)
- Dean Kaptsis
- College of Medicine and Public Health, Flinders University, Bedford Park, SA, Australia
| | - Martin Lewis
- Neuropsychiatric Laboratory, South Australian Health and Medical Research Institute, Adelaide, SA, Australia
- School of Biological Sciences, The University of Adelaide, Adelaide, SA, Australia
| | - Michael Sorich
- College of Medicine and Public Health, Flinders University, Bedford Park, SA, Australia
| | - Malcolm Battersby
- College of Medicine and Public Health, Flinders University, Bedford Park, SA, Australia
| |
Collapse
|
14
|
Abbes H, Zubiaur P, Soria-Chacartegui P, de la Torre T, Villapalos-García G, Candau C, Rodríguez-Lopez A, González-Iglesias E, Aldama M, Navares-Gomez M, Omezzine A, Ochoa D, Abad-Santos F. SLCO1B1 and ABCG2 genotype-informed phenotypes are related to variation in ramipril exposure. Basic Clin Pharmacol Toxicol 2024; 135:295-307. [PMID: 39011815 DOI: 10.1111/bcpt.14046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 05/13/2024] [Accepted: 06/17/2024] [Indexed: 07/17/2024]
Abstract
Ramipril is an angiotensin-converting enzyme inhibitor used for hypertension and heart failure management. To date, scarce literature is available on pharmacogenetic associations affecting ramipril. The goal of this study was to investigate the effect of 120 genetic variants in 34 pharmacogenes (i.e., genes encoding for enzymes like CYPs or UGTs and transporters like ABC or SLC) on ramipril pharmacokinetic variability and adverse drug reaction (ADR) incidence. Twenty-nine healthy volunteers who had participated in a single-dose bioequivalence clinical trial of two formulations of ramipril were recruited. A univariate and multivariate analysis searching for associations between genetic variants and ramipril pharmacokinetics was performed. SLCO1B1 and ABCG2 genotype-informed phenotypes strongly predicted ramipril exposure. Volunteers with the SLCO1B1 decreased function (DF) phenotype presented around 1.7-fold higher dose/weight-corrected area under the curve (AUC/DW) than volunteers with the normal function (NF) phenotype (univariate p-value [puv] < 0.001, multivariate p-value [pmv] < 0.001, β = 0.533, R2 = 0.648). Similarly, volunteers with ABCG2 DF + poor function (PF) phenotypes presented around 1.6-fold higher AUC/DW than those with the NF phenotype (puv = 0.011, pmv < 0.001, β = 0.259, R2 = 0.648). Our results suggest that SLCO1B1 and ABCG2 are important transporters to ramipril pharmacokinetics, and their genetic variation strongly alters its pharmacokinetics. Further studies are required to confirm these associations and their clinical relevance.
Collapse
Affiliation(s)
- Houwaida Abbes
- Clinical Pharmacology Department, Hospital Universitario de La Princesa, Universidad Autónoma de Madrid (UAM), Instituto de Investigación Sanitaria La Princesa (IP), Madrid, Spain
- Biochemistry Department, LR12SP11, Sahloul University Hospital, Sousse, Tunisia
- Faculty of Pharmacy of Monastir, University of Monastir, Monastir, Tunisia
| | - Pablo Zubiaur
- Clinical Pharmacology Department, Hospital Universitario de La Princesa, Universidad Autónoma de Madrid (UAM), Instituto de Investigación Sanitaria La Princesa (IP), Madrid, Spain
| | - Paula Soria-Chacartegui
- Clinical Pharmacology Department, Hospital Universitario de La Princesa, Universidad Autónoma de Madrid (UAM), Instituto de Investigación Sanitaria La Princesa (IP), Madrid, Spain
| | - Tamara de la Torre
- Clinical Pharmacology Department, Hospital Universitario de La Princesa, Universidad Autónoma de Madrid (UAM), Instituto de Investigación Sanitaria La Princesa (IP), Madrid, Spain
| | - Gonzalo Villapalos-García
- Clinical Pharmacology Department, Hospital Universitario de La Princesa, Universidad Autónoma de Madrid (UAM), Instituto de Investigación Sanitaria La Princesa (IP), Madrid, Spain
| | - Carmen Candau
- Clinical Pharmacology Department, Hospital Universitario de La Princesa, Universidad Autónoma de Madrid (UAM), Instituto de Investigación Sanitaria La Princesa (IP), Madrid, Spain
| | - Andrea Rodríguez-Lopez
- Clinical Pharmacology Department, Hospital Universitario de La Princesa, Universidad Autónoma de Madrid (UAM), Instituto de Investigación Sanitaria La Princesa (IP), Madrid, Spain
| | - Eva González-Iglesias
- Clinical Pharmacology Department, Hospital Universitario de La Princesa, Universidad Autónoma de Madrid (UAM), Instituto de Investigación Sanitaria La Princesa (IP), Madrid, Spain
| | - Marina Aldama
- Clinical Pharmacology Department, Hospital Universitario de La Princesa, Universidad Autónoma de Madrid (UAM), Instituto de Investigación Sanitaria La Princesa (IP), Madrid, Spain
| | - Marcos Navares-Gomez
- Clinical Pharmacology Department, Hospital Universitario de La Princesa, Universidad Autónoma de Madrid (UAM), Instituto de Investigación Sanitaria La Princesa (IP), Madrid, Spain
| | - Asma Omezzine
- Biochemistry Department, LR12SP11, Sahloul University Hospital, Sousse, Tunisia
- Faculty of Pharmacy of Monastir, University of Monastir, Monastir, Tunisia
| | - Dolores Ochoa
- Clinical Pharmacology Department, Hospital Universitario de La Princesa, Universidad Autónoma de Madrid (UAM), Instituto de Investigación Sanitaria La Princesa (IP), Madrid, Spain
| | - Francisco Abad-Santos
- Clinical Pharmacology Department, Hospital Universitario de La Princesa, Universidad Autónoma de Madrid (UAM), Instituto de Investigación Sanitaria La Princesa (IP), Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
15
|
Johnson L, Youssef E, O'Shea J, Thornley T, Gallagher J, Ledwidge M, Ryan C. Estimating the prevalence of potential and actionable drug-gene interactions in Irish primary care: A cross-sectional study. Br J Clin Pharmacol 2024; 90:2280-2298. [PMID: 38864275 DOI: 10.1111/bcp.16122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 04/03/2024] [Accepted: 04/13/2024] [Indexed: 06/13/2024] Open
Abstract
AIMS Pharmacogenetics (PGx) is increasingly recognized as a strategy for medicines optimisation and prevention of adverse drug reactions. According to guidelines produced by the Clinical Pharmacogenetics Implementation Consortium (CPIC) and the Dutch Pharmacogenetic Working Group (DPWG), most medicines with drug-gene interactions (DGIs) are prescribed in primary care. This study aimed to estimate the prevalence of potential and actionable DGIs involving all medicines dispensed in Irish primary care. METHODS Dispensings of 46 drugs to General Medical Services (GMS) patients in the Health Service Executive Primary Care Reimbursement Service Irish pharmacy claims database from 01 January 2021 to 31 December 2021 were analysed to estimate the national prevalence of total dispensings and incidence of first-time dispensings of drugs with potential DGIs according to the CPIC and/or DPWG guidelines. Phenotype frequency data from the UK Biobank and the CPIC were used to estimate the incidence of actionable DGIs. RESULTS One in five dispensings (12 443 637 of 62 754 498, 19.8%) were medicines with potential DGIs, 1 878 255 of these dispensed for the first time. On application of phenotype frequencies and linked guideline based therapeutic recommendations, 2 349 055 potential DGIs (18.9%) required action, such as monitoring and guarding against maximum dose, drug or dose change. One in five (369 700, 19.7%) first-time dispensings required action, with 139 169 (7.4%) requiring a change in prescribing. Antidepressants, weak opioids and statins were most commonly identified as having actionable DGIs. CONCLUSIONS This study estimated a high prevalence of DGIs in primary care in Ireland, identifying the need and opportunity to optimize drug therapy through PGx testing.
Collapse
Affiliation(s)
- L Johnson
- School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, Dublin, Ireland
| | - E Youssef
- Faculty of Health, Science, Social Care & Education, Kingston University, London, UK
| | - J O'Shea
- School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, Dublin, Ireland
| | - T Thornley
- School of Pharmacy, University of Nottingham, Nottingham, UK
| | - J Gallagher
- School of Medicine, University College Dublin, Dublin, Ireland
| | - M Ledwidge
- School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, Dublin, Ireland
- School of Medicine, University College Dublin, Dublin, Ireland
| | - C Ryan
- School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
16
|
Viviani R, Berres J, Stingl JC. Phenotypic Models of Drug-Drug-Gene Interactions Mediated by Cytochrome Drug-Metabolizing Enzymes. Clin Pharmacol Ther 2024; 116:592-601. [PMID: 38318716 DOI: 10.1002/cpt.3188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Accepted: 01/15/2024] [Indexed: 02/07/2024]
Abstract
Genetic polymorphisms in drug metabolizing enzymes and drug-drug interactions are major sources of inadequate drug exposure and ensuing adverse effects or insufficient responses. The current challenge in assessing drug-drug gene interactions (DDGIs) for the development of precise dose adjustment recommendation systems is to take into account both simultaneously. Here, we analyze the static models of DDGI from in vivo data and focus on the concept of phenoconversion to model inhibition and genetic polymorphisms jointly. These models are applicable to datasets where pharmacokinetic information is missing and are being used in clinical support systems and consensus dose adjustment guidelines. We show that all such models can be handled by the same formal framework, and that models that differ at first sight are all versions of the same linear phenoconversion model. This model includes the linear pharmacogenetic and inhibition models as special cases. We highlight present challenges in this endeavor and the open issues for future research in developing DDGI models for recommendation systems.
Collapse
Affiliation(s)
- Roberto Viviani
- Institute of Psychology, University of Innsbruck, Innsbruck, Austria
- Department of Psychiatry and Psychotherapy III, University of Ulm, Ulm, Germany
| | - Judith Berres
- Institute of Clinical Pharmacology, University Hospital RWTH Aachen, Aachen, Germany
| | - Julia C Stingl
- Institute of Clinical Pharmacology, University Hospital RWTH Aachen, Aachen, Germany
| |
Collapse
|
17
|
Okpete UE, Byeon H. Challenges and prospects in bridging precision medicine and artificial intelligence in genomic psychiatric treatment. World J Psychiatry 2024; 14:1148-1164. [PMID: 39165556 PMCID: PMC11331387 DOI: 10.5498/wjp.v14.i8.1148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 06/13/2024] [Accepted: 07/09/2024] [Indexed: 08/12/2024] Open
Abstract
Precision medicine is transforming psychiatric treatment by tailoring personalized healthcare interventions based on clinical, genetic, environmental, and lifestyle factors to optimize medication management. This study investigates how artificial intelligence (AI) and machine learning (ML) can address key challenges in integrating pharmacogenomics (PGx) into psychiatric care. In this integration, AI analyzes vast genomic datasets to identify genetic markers linked to psychiatric conditions. AI-driven models integrating genomic, clinical, and demographic data demonstrated high accuracy in predicting treatment outcomes for major depressive disorder and bipolar disorder. This study also examines the pressing challenges and provides strategic directions for integrating AI and ML in genomic psychiatry, highlighting the importance of ethical considerations and the need for personalized treatment. Effective implementation of AI-driven clinical decision support systems within electronic health records is crucial for translating PGx into routine psychiatric care. Future research should focus on developing enhanced AI-driven predictive models, privacy-preserving data exchange, and robust informatics systems to optimize patient outcomes and advance precision medicine in psychiatry.
Collapse
Affiliation(s)
- Uchenna Esther Okpete
- Department of Digital Anti-aging Healthcare (BK21), Inje University, Gimhae 50834, South Korea
| | - Haewon Byeon
- Department of Digital Anti-aging Healthcare (BK21), Inje University, Gimhae 50834, South Korea
- Department of Medical Big Data, Inje University, Gimhae 50834, South Korea
| |
Collapse
|
18
|
Wang WY, Lin L, Boone EC, Stevens J, Gaedigk A. CYP2D6 copy number determination using digital PCR. Front Pharmacol 2024; 15:1429286. [PMID: 39206265 PMCID: PMC11349684 DOI: 10.3389/fphar.2024.1429286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 07/22/2024] [Indexed: 09/04/2024] Open
Abstract
Background CYP2D6 testing is increasingly used to guide drug therapy and thus, reliable methods are needed to test this complex and polymorphic gene locus. A particular challenge arises from the detection and interpretation of structural variants (SVs) including gene deletions, duplications, and hybrids with the CYP2D7 pseudogene. This study validated the Absolute Q™ platform for digital PCR-based CYP2D6 copy number variation (CNV) determination by comparing results to those obtained with a previously established method using the QX200 platform. In addition, protocols for streamlining CYP2D6 CNV testing were established and validated including the "One-pot" single-step restriction enzyme digestion and a multiplex assay simultaneously targeting the CYP2D6 5'UTR, intron 6, and exon 9 regions. Methods Genomic DNA (gDNA) samples from Coriell (n = 13) and from blood, saliva, and liver tissue (n = 17) representing 0-6 copies were tested on the Absolute Q and QX200 platforms. Custom TaqMan™ copy number (CN) assays targeting CYP2D6 the 5'UTR, intron 6, and exon 9 regions and a reference gene assay (TERT or RNaseP) were combined for multiplexing by optical channel. In addition, two digestion methods (One-pot digestion and traditional) were assessed. Inconclusive CN values on the Absolute Q were resolved using an alternate reference gene and/or diluting gDNA. Results Overall, results between the two platforms and digestions methods were consistent. The "One-pot" digestion method and optically multiplexing up to three CYP2D6 regions yielded consistent result across DNA sample types and diverse SVs, reliably detecting up to 6 gene copies. Rare variation in reference genes were found to interfere with results and interpretation, which were resolved by using a different reference. Conclusion The Absolute Q produced accurate and reliable CYP2D6 copy number results allowing for a streamlined and economical protocol using One-pot digestion and multiplexing three target regions. Protocols are currently being expanded to other pharmacogenes presenting with SVs/CNVs.
Collapse
Affiliation(s)
- Wendy Y. Wang
- Division of Clinical Pharmacology, Toxicology and Therapeutic Innovation, Children’s Mercy Research Institute (CMRI), Kansas City, MO, United States
| | - Lancy Lin
- Genetic Sciences Division, Thermo Fisher Scientific, Waltham, CA, United States
| | - Erin C. Boone
- Division of Clinical Pharmacology, Toxicology and Therapeutic Innovation, Children’s Mercy Research Institute (CMRI), Kansas City, MO, United States
| | - Junko Stevens
- Genetic Sciences Division, Thermo Fisher Scientific, Waltham, CA, United States
| | - Andrea Gaedigk
- Division of Clinical Pharmacology, Toxicology and Therapeutic Innovation, Children’s Mercy Research Institute (CMRI), Kansas City, MO, United States
- School of Medicine, University of Missouri-Kansas City, Kansas City, MO, United States
| |
Collapse
|
19
|
Pedersen KW, Andersen JD, Hansen J, Børsting C, Banner J, Hasselstrøm JB, Jornil JR. Investigating the Correlation between Genotypes and Hepatic Protein Expression of CYP2C9, CYP2C19, CYP2D6, and CYP3A5 Using Postmortem Tissue from a Danish Population. Drug Metab Dispos 2024; 52:975-980. [PMID: 38906700 DOI: 10.1124/dmd.124.001692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 05/03/2024] [Accepted: 06/06/2024] [Indexed: 06/23/2024] Open
Abstract
The cytochrome P450 (CYP) family of enzymes plays a central role in the metabolism of many drugs. CYP genes are highly polymorphic, which is known to affect protein levels, but for some low frequent CYP genotypes the correlation between genotype and CYP protein expression is less established. In this study, we determined the CYP2C9, CYP2C19, CYP2D6, and CYP3A5 genotypes of 250 Danish individuals included in a postmortem study. For 116 of the individuals, the hepatic CYP protein levels were investigated by a proteomics approach. Overall, we found the postmortem genetic and proteomic data to be in agreement with those of other studies performed on fresh hepatic tissue, showing the usability of postmortem hepatic tissue for this type of investigation. For less investigated genotypes, we could corroborate previously found results: 1) statistically significantly lower levels of hepatic CYP2C9 protein in individuals carrying the CYP2C9*3 variant compared with individuals with two wild type (wt) alleles; 2) comparable levels of CYP2C19 in CYP2C19*2/*17 and CYP2C19*1/*2 individuals; 3) reduced CYP2D6 protein levels in heterozygous individuals with the CYP2D6*3, CYP2D6*4, and CYP2D6*5 gene deletion variants; and 4) significantly lower levels of CYP3A5 protein in CYP3A5*3 homozygous individuals compared with individuals who were heterozygous for the CYP3A5*3 allele or homozygous individuals for the wt alleles. In conclusion, the use of postmortem tissue significantly increases the access to human specimens for research purposes, and postmortem proteomics can be used to investigate the link between CYP genotypes and hepatic protein expression. SIGNIFICANCE STATEMENT: In tissue samples from a large postmortem cohort (n = 250) we determined the CYP2C9, CYP2C19, CYP2D6, and CYP3A5 genotypes. Hepatic CYP protein levels were investigated in 116 individuals using a proteomics approach. For common genotypes, we found results similar to previous knowledge, pointing toward the usability of postmortem tissue. For the less investigated genotypes, we were able to corroborate genotype/protein expression correlations. It is a novel approach to use a large postmortem cohort to investigate genetic/protein expression correlations.
Collapse
Affiliation(s)
- Kata W Pedersen
- Department of Forensic Medicine, Aarhus University, Aarhus, Denmark (K.W.P., J.H., J.B.H., J.R.J.); and Department of Forensic Medicine, University of Copenhagen, Copenhagen, Denmark (J.D.A., C.B., J.B.)
| | - Jeppe D Andersen
- Department of Forensic Medicine, Aarhus University, Aarhus, Denmark (K.W.P., J.H., J.B.H., J.R.J.); and Department of Forensic Medicine, University of Copenhagen, Copenhagen, Denmark (J.D.A., C.B., J.B.)
| | - Jakob Hansen
- Department of Forensic Medicine, Aarhus University, Aarhus, Denmark (K.W.P., J.H., J.B.H., J.R.J.); and Department of Forensic Medicine, University of Copenhagen, Copenhagen, Denmark (J.D.A., C.B., J.B.)
| | - Claus Børsting
- Department of Forensic Medicine, Aarhus University, Aarhus, Denmark (K.W.P., J.H., J.B.H., J.R.J.); and Department of Forensic Medicine, University of Copenhagen, Copenhagen, Denmark (J.D.A., C.B., J.B.)
| | - Jytte Banner
- Department of Forensic Medicine, Aarhus University, Aarhus, Denmark (K.W.P., J.H., J.B.H., J.R.J.); and Department of Forensic Medicine, University of Copenhagen, Copenhagen, Denmark (J.D.A., C.B., J.B.)
| | - Jørgen B Hasselstrøm
- Department of Forensic Medicine, Aarhus University, Aarhus, Denmark (K.W.P., J.H., J.B.H., J.R.J.); and Department of Forensic Medicine, University of Copenhagen, Copenhagen, Denmark (J.D.A., C.B., J.B.)
| | - Jakob R Jornil
- Department of Forensic Medicine, Aarhus University, Aarhus, Denmark (K.W.P., J.H., J.B.H., J.R.J.); and Department of Forensic Medicine, University of Copenhagen, Copenhagen, Denmark (J.D.A., C.B., J.B.)
| |
Collapse
|
20
|
Medwid S, Kim RB. Implementation of pharmacogenomics: Where are we now? Br J Clin Pharmacol 2024; 90:1763-1781. [PMID: 36366858 DOI: 10.1111/bcp.15591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 11/01/2022] [Accepted: 11/07/2022] [Indexed: 11/13/2022] Open
Abstract
Pharmacogenomics (PGx), examining the effect of genetic variation on interpatient variation in drug disposition and response, has been widely studied for several decades. However, as cost, as well as turnaround time associated with PGx testing, has significantly improved, the use of PGx in the clinical setting has been gaining momentum. Nevertheless, challenges have emerged in the broader clinical implementation of PGx. In this review, we will outline current models of PGx delivery and methodologies of evaluation, and discuss clinically relevant PGx tests and associated medications. Additionally, we will describe our approach for the broad implementation of pre-emptive DPYD genotyping in patients taking fluoropyrimidines in Ontario, Canada, as an example of clinically actionable PGx testing with sufficient clinical evidence of patient benefit that can become a new standard of patient care. We will highlight challenges associated with PGx testing, including a lack of diversity in PGx studies as well as general limitations that impact the broad adoption of PGx testing. Lastly, we examine the future of PGx, discussing new clinical targets, methodologies and analysis approaches.
Collapse
Affiliation(s)
- Samantha Medwid
- Department of Medicine, University of Western Ontario, London, Ontario, Canada
- Lawson Health Research Institute, London, Ontario, Canada
- London Health Sciences Centre, London, Ontario, Canada
| | - Richard B Kim
- Department of Medicine, University of Western Ontario, London, Ontario, Canada
- Lawson Health Research Institute, London, Ontario, Canada
- London Health Sciences Centre, London, Ontario, Canada
| |
Collapse
|
21
|
Narayanan S, Yuile A, Venkatesh B, McKay M, Itchins M, Pavlakis N, Wheeler H, Gray L, Wei J, Miller S, Kirwin B, Molloy MP, Clarke S. Therapeutic drug monitoring of osimertinib in EGFR mutant non-small cell lung cancer by dried blood spot and plasma collection: A pilot study. Br J Clin Pharmacol 2024; 90:1942-1951. [PMID: 38706157 DOI: 10.1111/bcp.16070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 03/19/2024] [Accepted: 03/21/2024] [Indexed: 05/07/2024] Open
Abstract
AIMS Therapeutic drug monitoring (TDM) has led to significant improvements in individualized medical care, although its implementation in oncology has been limited to date. Tyrosine kinase inhibitors (TKIs) are a group of therapies for which TDM has been suggested. Osimertinib is one such therapy used in the treatment of epidermal growth factor receptor (EGFR) mutation-driven lung cancer. Herein, we describe a prospective pilot study involving 21 patients on osimertinib primarily as a preliminary evaluation of drug levels in a real-world setting. METHODS Concentrations of the drug and its primary metabolites were measured with a validated liquid chromatography-mass spectrometry (LC-MS) assay across serial timepoints. As part of this study, inter-individual variability by dose and ethnicity as well as intra-individual variability across timepoints are explored. Furthermore, we attempted to validate dried blood spot (DBS)-based quantitation as an accurate alternative to plasma quantitation. RESULTS Successful quantitation of osimertinib and primary metabolites was achieved for our subjects. Compound plasma levels were highly correlated to DBS levels. There was no significant difference in concentrations with ethnicity or dosing or intra-individual variability across timepoints. CONCLUSIONS As such, we demonstrate that TDM for osimertinib is practical for future trials. We also validated the use of DBS as an alternative to conventional quantitation for exploration of TDM for osimertinib in larger trials and for other targeted therapies.
Collapse
Affiliation(s)
- Sathya Narayanan
- Department of Medical Oncology, Royal North Shore Hospital, Sydney, NSW, Australia
- Macquarie University Clinical Trials Unit, Macquarie University, Sydney, NSW, Australia
| | - Alexander Yuile
- Department of Medical Oncology, Royal North Shore Hospital, Sydney, NSW, Australia
- School of Medicine, University of Sydney, Sydney, NSW, Australia
| | - Bharat Venkatesh
- Kolling Institute of Medical Research, Sydney, NSW, Australia
- School of Medical Sciences, University of Sydney, Sydney, NSW, Australia
| | - Matthew McKay
- Kolling Institute of Medical Research, Sydney, NSW, Australia
- School of Medical Sciences, University of Sydney, Sydney, NSW, Australia
| | - Malinda Itchins
- Department of Medical Oncology, Royal North Shore Hospital, Sydney, NSW, Australia
- School of Medicine, University of Sydney, Sydney, NSW, Australia
- Chris O'Brien Lifehouse, Camperdown, NSW, Australia
| | - Nick Pavlakis
- Department of Medical Oncology, Royal North Shore Hospital, Sydney, NSW, Australia
- School of Medicine, University of Sydney, Sydney, NSW, Australia
| | - Helen Wheeler
- Department of Medical Oncology, Royal North Shore Hospital, Sydney, NSW, Australia
- School of Medicine, University of Sydney, Sydney, NSW, Australia
| | - Lauren Gray
- Department of Medical Oncology, Royal North Shore Hospital, Sydney, NSW, Australia
| | - Joe Wei
- Department of Medical Oncology, Royal North Shore Hospital, Sydney, NSW, Australia
| | - Samuel Miller
- Department of Medical Oncology, Royal North Shore Hospital, Sydney, NSW, Australia
| | - Brendan Kirwin
- Department of Medical Oncology, Royal North Shore Hospital, Sydney, NSW, Australia
| | - Mark P Molloy
- Kolling Institute of Medical Research, Sydney, NSW, Australia
- School of Medical Sciences, University of Sydney, Sydney, NSW, Australia
| | - Stephen Clarke
- Department of Medical Oncology, Royal North Shore Hospital, Sydney, NSW, Australia
- School of Medicine, University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
22
|
Dołoto A, Bąk E, Batóg G, Piątkowska-Chmiel I, Herbet M. Interactions of antidepressants with concomitant medications-safety of complex therapies in multimorbidities. Pharmacol Rep 2024; 76:714-739. [PMID: 39012418 PMCID: PMC11294384 DOI: 10.1007/s43440-024-00611-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 06/06/2024] [Accepted: 06/07/2024] [Indexed: 07/17/2024]
Abstract
Depression is the fourth most serious disease in the world. Left untreated, it is a cause of suicide attempts, emergence or exacerbation worsening of serious diseases, bodily and mental disorders, as well as increased risk of cardiovascular diseases, stroke, diabetes, and obesity, as well as endocrine and neurological diseases. Frequent coexistence of depression and other diseases requires the simultaneous use of several drugs from different therapeutic groups, which very often interact and intensify comorbidities, sometimes unrelated mechanisms. Sufficient awareness of potential drug interactions is critical in clinical practice, as it allows both to avoid disruption of proper pharmacotherapy and achieve substantive results. Therefore, this review aims to analyze the interactions of antidepressants with other concomitant medications. Against the backdrop of experimental research and a thorough analysis of the up-to-date literature, the authors discuss in detail the mechanisms and effects of action of individual drug interactions and adaptogens, including the latest antidepressants.
Collapse
Affiliation(s)
- Anna Dołoto
- Chair and Department of Toxicology, Faculty of Pharmacy, Medical University of Lublin, Jaczewskiego 8B Street, 20-090, Lublin, Poland
| | - Ewelina Bąk
- Chair and Department of Toxicology, Faculty of Pharmacy, Medical University of Lublin, Jaczewskiego 8B Street, 20-090, Lublin, Poland
| | - Gabriela Batóg
- Chair and Department of Toxicology, Faculty of Pharmacy, Medical University of Lublin, Jaczewskiego 8B Street, 20-090, Lublin, Poland
| | - Iwona Piątkowska-Chmiel
- Chair and Department of Toxicology, Faculty of Pharmacy, Medical University of Lublin, Jaczewskiego 8B Street, 20-090, Lublin, Poland
| | - Mariola Herbet
- Chair and Department of Toxicology, Faculty of Pharmacy, Medical University of Lublin, Jaczewskiego 8B Street, 20-090, Lublin, Poland.
| |
Collapse
|
23
|
Li D, Pain O, Fabbri C, Wong WLE, Lo CWH, Ripke S, Cattaneo A, Souery D, Dernovsek MZ, Henigsberg N, Hauser J, Lewis G, Mors O, Perroud N, Rietschel M, Uher R, Maier W, Baune BT, Biernacka JM, Bondolfi G, Domschke K, Kato M, Liu YL, Serretti A, Tsai SJ, Weinshilboum R, McIntosh AM, Lewis CM. Metabolic activity of CYP2C19 and CYP2D6 on antidepressant response from 13 clinical studies using genotype imputation: a meta-analysis. Transl Psychiatry 2024; 14:296. [PMID: 39025838 PMCID: PMC11258238 DOI: 10.1038/s41398-024-02981-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 06/10/2024] [Accepted: 06/18/2024] [Indexed: 07/20/2024] Open
Abstract
Cytochrome P450 enzymes including CYP2C19 and CYP2D6 are important for antidepressant metabolism and polymorphisms of these genes have been determined to predict metabolite levels. Nonetheless, more evidence is needed to understand the impact of genetic variations on antidepressant response. In this study, individual clinical and genetic data from 13 studies of European and East Asian ancestry populations were collected. The antidepressant response was clinically assessed as remission and percentage improvement. Imputed genotype was used to translate genetic polymorphisms to metabolic phenotypes (poor, intermediate, normal, and rapid+ultrarapid) of CYP2C19 and CYP2D6. CYP2D6 structural variants cannot be imputed from genotype data, limiting the determination of metabolic phenotypes, and precluding testing for association with response. The association of CYP2C19 metabolic phenotypes with treatment response was examined using normal metabolizers as the reference. Among 5843 depression patients, a higher remission rate was found in CYP2C19 poor metabolizers compared to normal metabolizers at nominal significance but did not survive after multiple testing correction (OR = 1.46, 95% CI [1.03, 2.06], p = 0.033, heterogeneity I2 = 0%, subgroup difference p = 0.72). No metabolic phenotype was associated with percentage improvement from baseline. After stratifying by antidepressants primarily metabolized by CYP2C19, no association was found between metabolic phenotypes and antidepressant response. Metabolic phenotypes showed differences in frequency, but not effect, between European- and East Asian-ancestry studies. In conclusion, metabolic phenotypes imputed from genetic variants using genotype were not associated with antidepressant response. CYP2C19 poor metabolizers could potentially contribute to antidepressant efficacy with more evidence needed. Sequencing and targeted pharmacogenetic testing, alongside information on side effects, antidepressant dosage, depression measures, and diverse ancestry studies, would more fully capture the influence of metabolic phenotypes.
Collapse
Affiliation(s)
- Danyang Li
- Social, Genetic and Developmental Psychiatry Centre, King's College London, London, GB, UK
- Cancer Centre, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, CN, China
| | - Oliver Pain
- Maurice Wohl Clinical Neuroscience Institute, Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, GB, UK
| | - Chiara Fabbri
- Social, Genetic and Developmental Psychiatry Centre, King's College London, London, GB, UK
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Win Lee Edwin Wong
- Social, Genetic and Developmental Psychiatry Centre, King's College London, London, GB, UK
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Chris Wai Hang Lo
- Social, Genetic and Developmental Psychiatry Centre, King's College London, London, GB, UK
| | - Stephan Ripke
- Department of Psychiatry and Psychotherapy, Universitätsmedizin Berlin Campus Charité Mitte, Berlin, DE, Germany
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
| | - Annamaria Cattaneo
- Biological Psychiatry Laboratory, IRCCS Fatebenefratelli, Brescia, Italy
- Department of Pharmacological and Biomedical Sciences, University of Milan, Milan, Italy
| | - Daniel Souery
- Laboratoire de Psychologie Medicale, Universitè Libre de Bruxelles and Psy Pluriel, Centre Européen de Psychologie Medicale, Brussels, Italy
| | - Mojca Z Dernovsek
- University Psychiatric Clinic, University of Ljubliana, Ljubljana, Slovenia
| | - Neven Henigsberg
- Department of Psychiatry, Croatian Institute for Brain Research, University of Zagreb Medical School, Zagreb, HR, Croatia
| | - Joanna Hauser
- Psychiatric Genetic Unit, Poznan University of Medical Sciences, Poznan, Poland
| | - Glyn Lewis
- Division of Psychiatry, University College London, London, GB, UK
| | - Ole Mors
- Psychosis Research Unit, Aarhus University Hospital - Psychiatry, Aarhus, Denmark
| | - Nader Perroud
- Department of Psychiatry, Geneva University Hospitals, Geneva, CH, Switzerland
| | - Marcella Rietschel
- Department of Genetic Epidemiology in Psychiatry, Medical Faculty Mannheim, University of Heidelberg, Central Institute of Mental Health, Mannheim, Denmark
| | - Rudolf Uher
- Department of Psychiatry, Dalhousie University, Halifax, NS, Canada
| | - Wolfgang Maier
- Department of Psychiatry and Psychotherapy, University of Bonn, Bonn, Denmark
| | - Bernhard T Baune
- Department of Psychiatry, University of Münster, Münster, Denmark
- Florey Institute for Neuroscience and Mental Health, University of Melbourne, Melbourne, Australia
- Department of Psychiatry, Melbourne Medical School, University of Melbourne, Melbourne, Australia
| | - Joanna M Biernacka
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, MN, USA
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | - Guido Bondolfi
- Department of Psychiatry, Geneva University Hospitals, Geneva, CH, Switzerland
| | - Katharina Domschke
- Department of Psychiatry and Psychotherapy, Medical Center, University of Freiburg, Freiburg, Denmark
| | - Masaki Kato
- Department of Neuropsychiatry, Kansai Medical University, Osaka, Japan
| | - Yu-Li Liu
- Center for Neuropsychiatric Research, National Health Research Institutes, Miaoli, Taiwan
| | | | - Shih-Jen Tsai
- Department of Psychiatry, Taipei Veterans General Hospital, Taipei, Taiwan
- Division of Psychiatry, School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Richard Weinshilboum
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
| | | | - Cathryn M Lewis
- Social, Genetic and Developmental Psychiatry Centre, King's College London, London, GB, UK.
- Department of Medical & Molecular Genetics, King's College London, London, GB, UK.
| |
Collapse
|
24
|
Floris M, Moschella A, Alcalay M, Montella A, Tirelli M, Fontana L, Idda ML, Guarnieri P, Capasso M, Mammì C, Nicoletti P, Miozzo M. Pharmacogenetics in Italy: current landscape and future prospects. Hum Genomics 2024; 18:78. [PMID: 38987819 PMCID: PMC11234611 DOI: 10.1186/s40246-024-00612-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2024] [Accepted: 04/30/2024] [Indexed: 07/12/2024] Open
Abstract
Pharmacogenetics investigates sequence of genes that affect drug response, enabling personalized medication. This approach reduces drug-induced adverse reactions and improves clinical effectiveness, making it a crucial consideration for personalized medical care. Numerous guidelines, drawn by global consortia and scientific organizations, codify genotype-driven administration for over 120 active substances. As the scientific community acknowledges the benefits of genotype-tailored therapy over traditionally agnostic drug administration, the push for its implementation into Italian healthcare system is gaining momentum. This evolution is influenced by several factors, including the improved access to patient genotypes, the sequencing costs decrease, the growing of large-scale genetic studies, the rising popularity of direct-to-consumer pharmacogenetic tests, and the continuous improvement of pharmacogenetic guidelines. Since EMA (European Medicines Agency) and AIFA (Italian Medicines Agency) provide genotype information on drug leaflet without clear and explicit clinical indications for gene testing, the regulation of pharmacogenetic testing is a pressing matter in Italy. In this manuscript, we have reviewed how to overcome the obstacles in implementing pharmacogenetic testing in the clinical practice of the Italian healthcare system. Our particular emphasis has been on germline testing, given the absence of well-defined national directives in contrast to somatic pharmacogenetics.
Collapse
Affiliation(s)
- Matteo Floris
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy.
| | - Antonino Moschella
- Unit of Medical Genetics, Grande Ospedale Metropolitano Bianchi-Melacrino-Morelli, Reggio Calabria, Italy
| | - Myriam Alcalay
- Department of Experimental Oncology, European Institute of Oncology IRCCS, Milano, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milano, Italy
| | - Annalaura Montella
- CEINGE Biotecnologie Avanzate, Napoli, Italy
- Department of Molecular Medicine and Medical Biotechnology (DMMBM), Università degli Studi di Napoli "Federico II", Napoli, Italy
| | - Matilde Tirelli
- CEINGE Biotecnologie Avanzate, Napoli, Italy
- Department of Molecular Medicine and Medical Biotechnology (DMMBM), Università degli Studi di Napoli "Federico II", Napoli, Italy
| | - Laura Fontana
- Medical Genetics Unit, Department of Health Sciences, ASST Santi Paolo e Carlo, Università degli Studi di Milano, Milan, Italy
| | - Maria Laura Idda
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
| | | | - Mario Capasso
- CEINGE Biotecnologie Avanzate, Napoli, Italy
- Department of Molecular Medicine and Medical Biotechnology (DMMBM), Università degli Studi di Napoli "Federico II", Napoli, Italy
| | - Corrado Mammì
- Unit of Medical Genetics, Grande Ospedale Metropolitano Bianchi-Melacrino-Morelli, Reggio Calabria, Italy
| | - Paola Nicoletti
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Monica Miozzo
- Medical Genetics Unit, Department of Health Sciences, ASST Santi Paolo e Carlo, Università degli Studi di Milano, Milan, Italy.
| |
Collapse
|
25
|
Wiss FM, Jakober D, Lampert ML, Allemann SS. Overcoming Barriers: Strategies for Implementing Pharmacist-Led Pharmacogenetic Services in Swiss Clinical Practice. Genes (Basel) 2024; 15:862. [PMID: 39062642 PMCID: PMC11276441 DOI: 10.3390/genes15070862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 06/27/2024] [Accepted: 06/28/2024] [Indexed: 07/28/2024] Open
Abstract
There is growing evidence that pharmacogenetic analysis can improve drug therapy for individual patients. In Switzerland, pharmacists are legally authorized to initiate pharmacogenetic tests. However, pharmacogenetic tests are rarely conducted in Swiss pharmacies. Therefore, we aimed to identify implementation strategies that facilitate the integration of a pharmacist-led pharmacogenetic service into clinical practice. To achieve this, we conducted semi-structured interviews with pharmacists and physicians regarding the implementation process of a pharmacist-led pharmacogenetic service. We utilized the Consolidated Framework for Implementation Research (CFIR) to identify potential facilitators and barriers in the implementation process. Additionally, we employed Expert Recommendations for Implementing Change (ERIC) to identify strategies mentioned in the interviews and used the CFIR-ERIC matching tool to identify additional strategies. We obtained interview responses from nine pharmacists and nine physicians. From these responses, we identified 7 CFIR constructs as facilitators and 12 as barriers. Some of the most commonly mentioned barriers included unclear procedures, lack of cost coverage by health care insurance, insufficient pharmacogenetics knowledge, lack of interprofessional collaboration, communication with the patient, and inadequate e-health technologies. Additionally, we identified 23 implementation strategies mentioned by interviewees using ERIC and 45 potential strategies using the CFIR-ERIC matching tool. In summary, we found that significant barriers hinder the implementation process of this new service. We hope that by highlighting potential implementation strategies, we can advance the integration of a pharmacist-led pharmacogenetic service in Switzerland.
Collapse
Affiliation(s)
- Florine M. Wiss
- Pharmaceutical Care, Department of Pharmaceutical Sciences, University of Basel, 4056 Basel, Switzerland; (D.J.); (M.L.L.)
- Institute of Hospital Pharmacy, Solothurner Spitäler, 4600 Olten, Switzerland
| | - Deborah Jakober
- Pharmaceutical Care, Department of Pharmaceutical Sciences, University of Basel, 4056 Basel, Switzerland; (D.J.); (M.L.L.)
| | - Markus L. Lampert
- Pharmaceutical Care, Department of Pharmaceutical Sciences, University of Basel, 4056 Basel, Switzerland; (D.J.); (M.L.L.)
- Institute of Hospital Pharmacy, Solothurner Spitäler, 4600 Olten, Switzerland
| | - Samuel S. Allemann
- Pharmaceutical Care, Department of Pharmaceutical Sciences, University of Basel, 4056 Basel, Switzerland; (D.J.); (M.L.L.)
| |
Collapse
|
26
|
Frear S, Sherman A, Rule D, Marcath LA. Prevalence of CYP2D6 structural variation in large retrospective study. Pharmacogenet Genomics 2024; 34:135-138. [PMID: 38372405 DOI: 10.1097/fpc.0000000000000525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/20/2024]
Abstract
CYP2D6 is a highly polymorphic gene with clinically important structural variations. Commonly, only exon 9 is assayed on clinical pharmacogenomics panels, as it allows for accurate functional characterization even in the presence of a CYP2D6::CYP2D7 conversion. However, this method does not capture CYP2D7::CYP2D6 (CYP2D6*13) conversions, possibly leading to inaccurate phenotype assignment. The study's purpose was to determine the frequency of structural variations in CYP2D6 utilizing multiple copy number variation (CNV) assay locations to quantify the potential impact on clinical phenotype classification. A retrospective analysis was conducted of de-identified pharmacogenomics data submitted through the Translational Software, Inc. platform. Samples with CYP2D6 CNV data for exon 9 and at least one additional CNV location (5'UTR, exon 1, intron 2, exon 5 or intron 6) were included. CYP2D7::CYP2D6 and CYP2D6::CYP2D7 conversions were classified according to PharmVar nomenclature. The CYP2D6 copies were capped at four total copies to account for assay limitations in detecting more than four copies. A total of 106,474 samples were included for analysis. CYP2D7::CYP2D6 conversions were present in approximately 2.44% of samples, and 5.84% of samples had CYP2D6::CYP2D7 conversions. Many samples did not have a CYP2D7 conversion detected (91.5%; 97,462/106,474). A full gene deletion was detected in 0.15%, and 5.98% had a duplication or multiplication present. This retrospective study underscores the importance of testing more than one CNV site for CYP2D6 . Over 2% of patients were found to have a CYP2D7::CYP2D6 conversion. This translates into potentially misclassified phenotype classification and incongruent clinical recommendations.
Collapse
Affiliation(s)
- Samantha Frear
- Translational Software, Inc. Mercer Island, Washington, USA
| | | | | | | |
Collapse
|
27
|
Hahn M, Frantz AM, Eckert A, Reif A. [Barriers for Implementation of PGx Testing in Psychiatric Hospitals in Germany: Results of the FACT-PGx Study]. FORTSCHRITTE DER NEUROLOGIE-PSYCHIATRIE 2024; 92:221-229. [PMID: 37130546 DOI: 10.1055/a-2060-0694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
OBJECTIVE The FACT-PGx study was conducted to analyze barriers to implementation of pharmacogenetic testing in psychiatric hospitals in Germany and to propose solutions for its faster and easier implementation in all hospitals. METHODS 104 patients (50% female) were genotyped and participated in the study. 67 completed a survey. To analyze the correlation between continuous data (age) of the survey, the wilcoxon rank test and for categorial data (education level, history of treatment and episodes), t-test was used. RESULTS No patient declined to be genotyped. 99% believed that genotyping could help to shorten their hospital stay. Patients >40 years of age and with higher educational levels were willing to pay for the PGx (p=0.009). On average, patients were willing to pay 117.42€ +/-140.49€ and to wait 15.83+/- 8.92 days for the results. Processes differed significantly between routine laboratory screening and PGx testing which could be a barrier for implementation. CONCLUSION Patients are not barriers to but enablers of an implementation of PGx. New process flows can be barriers, but can be overcome by optimization.
Collapse
Affiliation(s)
- Martina Hahn
- Klinik für Psychiatrie, Psychosomatik und Psychotherapie, Universitätsklinikum Frankfurt, Frankfurt am Main, Germany
- Klinik für psychische Gesundheit, varisano Klinikum Frankfurt Hoechst, Frankfurt, Germany
| | - Amelie Merle Frantz
- Klinik für Psychiatrie, Psychosomatik und Psychotherapie, Universitätsklinikum Frankfurt, Frankfurt am Main, Germany
| | - Andreas Eckert
- Klinik für Psychiatrie, Psychosomatik und Psychotherapie, Universitätsklinikum Frankfurt, Frankfurt am Main, Germany
| | - Andreas Reif
- Klinik für Psychiatrie, Psychosomatik und Psychotherapie, Universitätsklinikum Frankfurt, Frankfurt am Main, Germany
| |
Collapse
|
28
|
Nakad Z, Saab Y. Precision dosing for patients on tricyclic antidepressants. Pharmacogenet Genomics 2024; 34:117-125. [PMID: 38465522 DOI: 10.1097/fpc.0000000000000527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
OBJECTIVE We aim to develop a personalized dosing tool for tricyclic antidepressants (TCAs) that integrates CYP2D6 and CYP2C19 gene variants and their effects while also considering the polypharmacy effect. METHODS The study first adopted a scoring system that assigns weights to each genetic variant. A formula was then developed to compute the effect of both genes' variants on drug dosing. The output of the formula was assessed by a comparison with the clinical pharmacogenetics implementation consortium recommendation. The study also accounts for the effect of the co-administration of inhibitors and inducers on drug metabolism. Accordingly, a user-friendly tool, Clinical Dosing Tool ver.2, was created to assist clinicians in dosing patients on TCAs. RESULTS The study provides a comprehensive list of all alleles with corresponding activity values and phenotypes for both enzymes. The tool calculated an updated area under the curve ratio that utilizes the effects of both enzymes' variants for dose adjustment. The tool provided a more accurate individualized dosing that also integrates the polypharmacy effect. CONCLUSION To the best of our knowledge, the literature misses such a tool that provides a numerical adjusted dose based on continuous numerical activity scores for the considered patients' alleles and phenoconversion.
Collapse
Affiliation(s)
- Zahi Nakad
- Electrical and Computer Engineering Department, School of Engineering, Lebanese American University, Lebanon
| | - Yolande Saab
- Pharmaceutical Sciences Department, School of Pharmacy, Lebanese American University, Lebanon
| |
Collapse
|
29
|
Brouwer JMJL, Wardenaar KJ, Nolte IM, Liemburg EJ, Bet PM, Snieder H, Mulder H, Cath DC, Penninx BWJH. Association of CYP2D6 and CYP2C19 metabolizer status with switching and discontinuing antidepressant drugs: an exploratory study. BMC Psychiatry 2024; 24:394. [PMID: 38797832 PMCID: PMC11129450 DOI: 10.1186/s12888-024-05764-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Accepted: 04/15/2024] [Indexed: 05/29/2024] Open
Abstract
BACKGROUND Tailoring antidepressant drugs (AD) to patients' genetic drug-metabolism profile is promising. However, literature regarding associations of ADs' treatment effect and/or side effects with drug metabolizing genes CYP2D6 and CYP2C19 has yielded inconsistent results. Therefore, our aim was to longitudinally investigate associations between CYP2D6 (poor, intermediate, and normal) and CYP2C19 (poor, intermediate, normal, and ultrarapid) metabolizer-status, and switching/discontinuing of ADs. Next, we investigated whether the number of perceived side effects differed between metabolizer statuses. METHODS Data came from the multi-site naturalistic longitudinal cohort Netherlands Study of Depression and Anxiety (NESDA). We selected depression- and/or anxiety patients, who used AD at some point in the course of the 9 years follow-up period (n = 928). Medication use was followed to assess patterns of AD switching/discontinuation over time. CYP2D6 and CYP2C19 alleles were derived using genome-wide data of the NESDA samples and haplotype data from the PharmGKB database. Logistic regression analyses were conducted to investigate the association of metabolizer status with switching/discontinuing ADs. Mann-Whitney U-tests were conducted to compare the number of patient-perceived side effects between metabolizer statuses. RESULTS No significant associations were observed of CYP metabolizer status with switching/discontinuing ADs, nor with the number of perceived side effects. CONCLUSIONS We found no evidence for associations between CYP metabolizer statuses and switching/discontinuing AD, nor with side effects of ADs, suggesting that metabolizer status only plays a limited role in switching/discontinuing ADs. Additional studies with larger numbers of PM and UM patients are needed to further determine the potential added value of pharmacogenetics to guide pharmacotherapy.
Collapse
Affiliation(s)
- Jurriaan M J L Brouwer
- Research School of Behavioral and Cognitive Neurosciences, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.
- GGZ Drenthe Mental Health Center Drenthe, Assen, The Netherlands.
- Department of Clinical Pharmacy, Wilhelmina Hospital Assen, Assen, The Netherlands.
- Department of Clinical Pharmacy, Martini Hospital Groningen, Van Swietenlaan 1, Groningen, 9728 NT, The Netherlands.
| | - Klaas J Wardenaar
- GGZ Drenthe Mental Health Center Drenthe, Assen, The Netherlands
- Department of Psychiatry, University Medical Center Groningen, Interdisciplinary Center Psychopathology and Emotion Regulation, University of Groningen, Groningen, The Netherlands
- Faculty of Behavioural and Social Sciences, University of Groningen, Groningen, The Netherlands
| | - Ilja M Nolte
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Edith J Liemburg
- GGZ Drenthe Mental Health Center Drenthe, Assen, The Netherlands
- Rob Giel Research Center, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Pierre M Bet
- Department of Clinical Pharmacology and Pharmacy, Amsterdam University Medical Center, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Harold Snieder
- Rob Giel Research Center, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Hans Mulder
- Department of Clinical Pharmacy, Wilhelmina Hospital Assen, Assen, The Netherlands
| | - Danielle C Cath
- Research School of Behavioral and Cognitive Neurosciences, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- GGZ Drenthe Mental Health Center Drenthe, Assen, The Netherlands
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Brenda W J H Penninx
- Department of Psychiatry, Amsterdam University Medical Center, Vrije Universiteit Amsterdam, Amsterdam Public Health, Amsterdam, The Netherlands
| |
Collapse
|
30
|
Ragia G, Pallikarou M, Manolopoulou Y, Vorvolakos T, Manolopoulos VG. Genetic diversity of cytochrome P450 in patients receiving psychiatric care in Greece: a step towards clinical implementation. Pharmacogenomics 2024; 25:217-229. [PMID: 38884939 PMCID: PMC11388136 DOI: 10.1080/14622416.2024.2346072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Accepted: 04/12/2024] [Indexed: 06/18/2024] Open
Abstract
Aim: We herein inferred the genetic diversity of CYP450 isoenzymes to predict the percentage of patients who need dose adjustment in drugs used in psychiatry.Materials & methods: Data of 784 Greek patients receiving psychiatric care who were genotyped for CYP2D6, CYP2C19, CYP1A2, CYP3A5 and CYP2C9 isoenzymes were inferred to gene-drug pairs according to the US FDA, Clinical Pharmacogenetics Implementation Consortium and Dutch Pharmacogenetics Working Group annotations and published literature.Results: Atypical metabolism was found for 36.8% of patients in CYP2D6, 49.2% in CYP2C19, 45% in CYP1A2, 16.7% in CYP3A5 and 41.8% in CYP2C9. Dosage adjustment need was estimated for 10.2% of venlafaxine, 10.0% of paroxetine, 6.4% of sertraline, 30.8% of citalopram, 52.1% of escitalopram, 18.2% of fluvoxamine, 54.1% of tricyclic antidepressants, 16.7% of zuclopenthixol, 10.6% of haloperidol and 13.3% of risperidone treated patients.Conclusion: Clinical psychiatric pharmacogenomic implementation holds promise to improve drug effectiveness and safety.
Collapse
Affiliation(s)
- Georgia Ragia
- Laboratory of Pharmacology, Medical School, Democritus University of Thrace, Dragana Campus, Alexandroupolis, 68100, Greece
- Individualised Medicine & Pharmacological Research Solutions (IMPReS) Center, Dragana Campus, Alexandroupolis, 68100, Greece
- DNALEX S.A., Leontaridou 2, Alexandroupolis, 68100, Greece
| | - Myria Pallikarou
- Laboratory of Pharmacology, Medical School, Democritus University of Thrace, Dragana Campus, Alexandroupolis, 68100, Greece
- Individualised Medicine & Pharmacological Research Solutions (IMPReS) Center, Dragana Campus, Alexandroupolis, 68100, Greece
| | | | - Theofanis Vorvolakos
- Department of Psychiatry, Medical School, Academic General Hospital of Alexandroupolis, Democritus University of Thrace, Dragana Campus, Alexandroupolis, 68100, Greece
| | - Vangelis G Manolopoulos
- Laboratory of Pharmacology, Medical School, Democritus University of Thrace, Dragana Campus, Alexandroupolis, 68100, Greece
- Individualised Medicine & Pharmacological Research Solutions (IMPReS) Center, Dragana Campus, Alexandroupolis, 68100, Greece
- Clinical Pharmacology Unit, Academic General Hospital of Alexandroupolis, Dragana Campus, Alexandroupolis, 68100, Greece
| |
Collapse
|
31
|
Ghanbarian S, Wong GWK, Bunka M, Edwards L, Cressman S, Conte T, Peterson S, Vijh R, Price M, Schuetz C, Erickson D, Riches L, Landry G, McGrail K, Austin J, Bryan S. A Canadian Simulation Model for Major Depressive Disorder: Study Protocol. PHARMACOECONOMICS - OPEN 2024; 8:493-505. [PMID: 38528312 PMCID: PMC11058136 DOI: 10.1007/s41669-024-00481-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 02/25/2024] [Indexed: 03/27/2024]
Abstract
BACKGROUND Major depressive disorder (MDD) is a common, often recurrent condition and a significant driver of healthcare costs. People with MDD often receive pharmacological therapy as the first-line treatment, but the majority of people require more than one medication trial to find one that relieves symptoms without causing intolerable side effects. There is an acute need for more effective interventions to improve patients' remission and quality of life and reduce the condition's economic burden on the healthcare system. Pharmacogenomic (PGx) testing could deliver these objectives, using genomic information to guide prescribing decisions. With an already complex and multifaceted care pathway for MDD, future evaluations of new treatment options require a flexible analytic infrastructure encompassing the entire care pathway. Individual-level simulation models are ideally suited for this purpose. We sought to develop an economic simulation model to assess the effectiveness and cost effectiveness of PGx testing for individuals with major depression. Additionally, the model serves as an analytic infrastructure, simulating the entire patient pathway for those with MDD. METHODS AND ANALYSIS Key stakeholders, including patient partners, clinical experts, researchers, and modelers, designed and developed a discrete-time microsimulation model of the clinical pathways of adults with MDD in British Columbia (BC), including all publicly-funded treatment options and multiple treatment steps. The Simulation Model of Major Depression (SiMMDep) was coded with a modular approach to enhance flexibility. The model was populated using multiple original data analyses conducted with BC administrative data, a systematic review, and an expert panel. The model accommodates newly diagnosed and prevalent adult patients with MDD in BC, with and without PGx-guided treatment. SiMMDep comprises over 1500 parameters in eight modules: entry cohort, demographics, disease progression, treatment, adverse events, hospitalization, costs and quality-adjusted life-years (payoff), and mortality. The model predicts health outcomes and estimates costs from a health system perspective. In addition, the model can incorporate interactive decision nodes to address different implementation strategies for PGx testing (or other interventions) along the clinical pathway. We conducted various forms of model validation (face, internal, and cross-validity) to ensure the correct functioning and expected results of SiMMDep. CONCLUSION SiMMDep is Canada's first medication-specific, discrete-time microsimulation model for the treatment of MDD. With patient partner collaboration guiding its development, it incorporates realistic care journeys. SiMMDep synthesizes existing information and incorporates provincially-specific data to predict the benefits and costs associated with PGx testing. These predictions estimate the effectiveness, cost-effectiveness, resource utilization, and health gains of PGx testing compared with the current standard of care. However, the flexible analytic infrastructure can be adapted to support other policy questions and facilitate the rapid synthesis of new data for a broader search for efficiency improvements in the clinical field of depression.
Collapse
Affiliation(s)
- Shahzad Ghanbarian
- School for Population and Public Health, University of British Columbia, Vancouver, BC, Canada.
- Centre for Clinical Epidemiology and Evaluation, Vancouver Coastal Health Research Institute, Vancouver, BC, Canada.
| | - Gavin W K Wong
- School for Population and Public Health, University of British Columbia, Vancouver, BC, Canada
- Centre for Clinical Epidemiology and Evaluation, Vancouver Coastal Health Research Institute, Vancouver, BC, Canada
| | - Mary Bunka
- School for Population and Public Health, University of British Columbia, Vancouver, BC, Canada
- Centre for Clinical Epidemiology and Evaluation, Vancouver Coastal Health Research Institute, Vancouver, BC, Canada
| | - Louisa Edwards
- School for Population and Public Health, University of British Columbia, Vancouver, BC, Canada
- Centre for Clinical Epidemiology and Evaluation, Vancouver Coastal Health Research Institute, Vancouver, BC, Canada
| | - Sonya Cressman
- Department of Emergency Medicine, University of British Columbia, Vancouver, BC, Canada
- Faculty of Health Sciences, Simon Fraser University, Vancouver, Canada
| | - Tania Conte
- School for Population and Public Health, University of British Columbia, Vancouver, BC, Canada
| | - Sandra Peterson
- Centre for Health Services and Policy Research, University of British Columbia, Vancouver, BC, Canada
| | - Rohit Vijh
- School for Population and Public Health, University of British Columbia, Vancouver, BC, Canada
- Department of Family Practice, University of British Columbia, Vancouver, BC, Canada
| | - Morgan Price
- Department of Family Practice, University of British Columbia, Vancouver, BC, Canada
| | - Christian Schuetz
- Department of Psychiatry, University of British Columbia, Vancouver, BC, Canada
| | - David Erickson
- Department of Psychiatry, University of British Columbia, Vancouver, BC, Canada
- Psychology Department, Fraser Health, Vancouver, BC, Canada
| | | | | | - Kim McGrail
- Centre for Health Services and Policy Research, University of British Columbia, Vancouver, BC, Canada
| | - Jehannine Austin
- Department of Psychiatry, University of British Columbia, Vancouver, BC, Canada
- Department of Medical Genetics, University of British Columbia, Vancouver, Canada
| | - Stirling Bryan
- School for Population and Public Health, University of British Columbia, Vancouver, BC, Canada
- Centre for Clinical Epidemiology and Evaluation, Vancouver Coastal Health Research Institute, Vancouver, BC, Canada
| |
Collapse
|
32
|
Platona RI, Voiță-Mekeres F, Tudoran C, Tudoran M, Enătescu VR. The Contribution of Genetic Testing in Optimizing Therapy for Patients with Recurrent Depressive Disorder. Clin Pract 2024; 14:703-717. [PMID: 38804388 PMCID: PMC11130888 DOI: 10.3390/clinpract14030056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 04/16/2024] [Accepted: 04/19/2024] [Indexed: 05/29/2024] Open
Abstract
(1) Background: The aim of this study was to analyze the impact of pharmacogenetic-guided antidepressant therapy on the 12-month evolution of the intensity of depressive symptoms in patients with recurrent depressive disorder (RDD) in comparison to a control group of depressive subjects who were treated conventionally. (2) Methods: This prospective longitudinal study was conducted between 2019 and 2022, and the patients were evaluated by employing the Hamilton Depression Rating Scale (HAM-D), Hamilton Anxiety Rating Scale (HAM-A) and the Clinical Global Impressions Scale: Severity and Improvement. We followed them up at 1, 3, 6, and 12 months. (3) Results: Of the 76 patients with RDD, 37 were tested genetically (Group A) and 39 were not (Group B). Although the patients from Group A had statistically significantly more severe MDD at baseline than those from Group B (p < 0.001), by adjusting their therapy according to the genetic testing, they had a progressive and more substantial reduction in the severity of RDD symptoms [F = 74.334; η2 = 0.674; p < 0.001], indicating a substantial association with the results provided by the genetic testing (67.4%). (4) Conclusions: In patients with RDD and a poor response to antidepressant therapy, pharmacogenetic testing allows for treatment adjustment, resulting in a constant and superior reduction in the intensity of depression and anxiety symptoms.
Collapse
Affiliation(s)
- Rita Ioana Platona
- Doctoral School, University of Medicine and Pharmacy “Victor Babes” Timisoara, 300041 Timisoara, Romania;
- Psychiatry Department, County Clinical Emergency Hospital of Oradea, 410169 Oradea, Romania
| | - Florica Voiță-Mekeres
- Psychiatry Department, County Clinical Emergency Hospital of Oradea, 410169 Oradea, Romania
- Morphological Disciplines Department, Faculty of Medicine and Pharmacy, University of Oradea, 410087 Oradea, Romania
| | - Cristina Tudoran
- Department VII, Internal Medicine II, Discipline of Cardiology, University of Medicine and Pharmacy “Victor Babes” Timisoara, E. Murgu Square, No. 2, 300041 Timisoara, Romania; (C.T.); (M.T.)
- Center of Molecular Research in Nephrology and Vascular Disease, University of Medicine and Pharmacy “Victor Babes” Timisoara, E. Murgu Square, No. 2, 300041 Timisoara, Romania
- Cardiology Clinic, County Emergency Hospital “Pius Brinzeu”, L. Rebreanu, No. 156, 300723 Timisoara, Romania;
| | - Mariana Tudoran
- Department VII, Internal Medicine II, Discipline of Cardiology, University of Medicine and Pharmacy “Victor Babes” Timisoara, E. Murgu Square, No. 2, 300041 Timisoara, Romania; (C.T.); (M.T.)
- Center of Molecular Research in Nephrology and Vascular Disease, University of Medicine and Pharmacy “Victor Babes” Timisoara, E. Murgu Square, No. 2, 300041 Timisoara, Romania
- Cardiology Clinic, County Emergency Hospital “Pius Brinzeu”, L. Rebreanu, No. 156, 300723 Timisoara, Romania;
| | - Virgil Radu Enătescu
- Cardiology Clinic, County Emergency Hospital “Pius Brinzeu”, L. Rebreanu, No. 156, 300723 Timisoara, Romania;
- Discipline of Psychiatry, Department of Neurosciences, University of Medicine and Pharmacy “Victor Babes” Timisoara, 300041 Timişoara, Romania
| |
Collapse
|
33
|
Qian J, Xu T, Pan P, Sun W, Hu G, Cai J. Study on genotype and phenotype of novel CYP2D6 variants using pharmacokinetic and pharmacodynamic models with metoprolol as a substrate drug. THE PHARMACOGENOMICS JOURNAL 2024; 24:13. [PMID: 38637522 DOI: 10.1038/s41397-024-00332-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 03/31/2024] [Accepted: 04/09/2024] [Indexed: 04/20/2024]
Abstract
To investigate the pharmacokinetic and pharmacodynamic profiles of volunteers carrying CYP2D6 genotypes with unknow metabolic phenotypes, a total of 22 volunteers were recruited based on the sequencing results. Peripheral blood and urine samples were collected at specific time points after oral administration of metoprolol. A validated high-performance liquid chromatography (HPLC) method was used to determine the concentrations of metoprolol and α-hydroxymetoprolol. Blood pressure and electrocardiogram were also monitored. The results showed that the main pharmacokinetic parameters of metoprolol in CYP2D6*1/*34 carriers are similar to those in CYP2D6*1/*1 carriers. However, in individuals carrying the CYP2D6*10/*87, CYP2D6*10/*95, and CYP2D6*97/*97 genotypes, the area under the curve (AUC) and half-life (t1/2) of metoprolol increased by 2-3 times compared to wild type. The urinary metabolic ratio of metoprolol in these genotypes is consistent with the trends observed in plasma samples. Therefore, CYP2D6*1/*34 can be considered as normal metabolizers, while CYP2D6*10/*87, CYP2D6*10/*95, and CYP2D6*97/*97 are intermediate metabolizers. Although the blood concentration of metoprolol has been found to correlate with CYP2D6 genotype, its blood pressure-lowering effect reaches maximum effectiveness at a reduction of 25 mmHg. Furthermore, P-Q interval prolongation and heart rate reduction are not positively correlated with metoprolol blood exposure. Based on the pharmacokinetic-pharmacodynamic model, this study clarified the properties of metoprolol in subjects with novel CYP2D6 genotypes and provided important fundamental data for the translational medicine of this substrate drug.
Collapse
Affiliation(s)
- Jianchang Qian
- Institute of Molecular Toxicology and Pharmacology, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China.
| | - Tao Xu
- Institute of Molecular Toxicology and Pharmacology, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Peipei Pan
- Taizhou Central Hospital (Taizhou University Hospital), Taizhou, Zhejiang, China
| | - Wei Sun
- Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Guoxin Hu
- Institute of Molecular Toxicology and Pharmacology, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China.
| | - Jianping Cai
- Institute of Molecular Toxicology and Pharmacology, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China.
- The Key Laboratory of Geriatrics, Beijing Hospital & Beijing Institute of Geriatrics, Ministry of Health, Beijing, 100005, China.
| |
Collapse
|
34
|
Kharasch ED, Lenze EJ. Pharmacogenetic Influence on Stereoselective Steady-State Disposition of Bupropion. Drug Metab Dispos 2024; 52:455-466. [PMID: 38467432 PMCID: PMC11023817 DOI: 10.1124/dmd.124.001697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2024] [Revised: 03/02/2024] [Accepted: 03/07/2024] [Indexed: 03/13/2024] Open
Abstract
Bupropion is used for treating depression, obesity, and seasonal affective disorder, and for smoking cessation. Bupropion is commonly prescribed, but has complex pharmacokinetics and interindividual variability in metabolism and bioactivation may influence therapeutic response, tolerability, and safety. Bupropion is extensively and stereoselectively metabolized, the metabolites are pharmacologically active, and allelic variation in cytochrome P450 (CYP) 2B6 affects clinical hydroxylation of single-dose bupropion. Genetic effects on stereoselective disposition of steady-state bupropion are not known. In this preplanned secondary analysis of a prospective, randomized, double-blinded, crossover study which compared brand and generic bupropion XL 300 mg drug products, we measured steady-state enantiomeric plasma and urine parent bupropion and primary and secondary metabolite concentrations. This investigation evaluated the influence of genetic polymorphisms in CYP2B6, CYP2C19, and P450 oxidoreductase on the disposition of Valeant Pharmaceuticals Wellbutrin brand bupropion in 67 participants with major depressive disorder. We found that hydroxylation of both bupropion enantiomers was lower in carriers of the CYP2B6*6 allele and in carriers of the CYP2B6 516G>T variant, with correspondingly greater bupropion and lesser hydroxybupropion plasma concentrations. Hydroxylation was 25-50% lower in CYP2B6*6 carriers and one-third to one-half less in 516T carriers. Hydroxylation of the bupropion enantiomers was comparably affected by CYP2B6 variants. CYP2C19 polymorphisms did not influence bupropion plasma concentrations or hydroxybupropion formation but did influence the minor pathway of 4'-hydroxylation of bupropion and primary metabolites. P450 oxidoreductase variants did not influence bupropion disposition. Results show that CYP2B6 genetic variants affect steady-state metabolism and bioactivation of Valeant brand bupropion, which may influence therapeutic outcomes. SIGNIFICANCE STATEMENT: Bupropion, used for depression, obesity, and smoking cessation, undergoes metabolic bioactivation, with incompletely elucidated interindividual variability. We evaluated cytochrome P450 (CYP) 2B6, CYP2C19 and P450 oxidoreductase genetic variants and steady-state bupropion and metabolite enantiomers disposition. Both enantiomers hydroxylation was lower in CYP2B6*6 and CYP2B6 516G>T carriers, with greater bupropion and lesser hydroxybupropion plasma concentrations. CYP2C19 polymorphisms did not affect bupropion or hydroxybupropion but did influence minor 4'-hydroxylation of bupropion and primary metabolites. CYP2B6 variants affect steady-state bupropion bioactivation, which may influence therapeutic outcomes.
Collapse
Affiliation(s)
- Evan D Kharasch
- Department of Anesthesiology, Duke University, Durham, North Carolina (E.D.K.); Bermaride, LLC (E.D.K.); and Department of Psychiatry, Washington University in St. Louis, St. Louis, Missouri (E.J.L.)
| | - Eric J Lenze
- Department of Anesthesiology, Duke University, Durham, North Carolina (E.D.K.); Bermaride, LLC (E.D.K.); and Department of Psychiatry, Washington University in St. Louis, St. Louis, Missouri (E.J.L.)
| |
Collapse
|
35
|
Chen YC, Wang H, Mandrekar JN, Robertson CE, Starling AJ, Cutrer FM, Chiang CC. Pharmacogenomic study-A pilot study of the effect of pharmacogenomic phenotypes on the adequate dosing of verapamil for migraine prevention. THE PHARMACOGENOMICS JOURNAL 2024; 24:11. [PMID: 38594235 DOI: 10.1038/s41397-024-00331-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/10/2022] [Revised: 03/29/2024] [Accepted: 04/02/2024] [Indexed: 04/11/2024]
Abstract
OBJECTIVE To investigate factors affecting the efficacy and tolerability of verapamil for migraine prevention using individual pharmacogenomic phenotypes. BACKGROUND Verapamil has a wide range of dosing in headache disorders without reliable tools to predict the optimal doses for an individual. METHODS This is a retrospective chart review examining adults with existing pharmacogenomic reports at Mayo Clinic who had used verapamil for migraine. Effects of six cytochrome P450 phenotypes on the doses of verapamil for migraine prevention were assessed. RESULTS Our final analysis included 33 migraine patients (82% with aura). The mean minimum effective and maximum tolerable doses of verapamil were 178.2(20-320) mg and 227.9(20-480) mg. A variety of CYP2C9, CYP2D6, and CYP3A5 phenotypes were found, without significant association with the verapamil doses after adjusting for age, sex, body mass index, and smoking status. CONCLUSIONS We demonstrated a wide range of effective and tolerable verapamil doses used for migraine in a cohort with various pharmacogenomic phenotypes.
Collapse
Affiliation(s)
- Yi-Chieh Chen
- Department of Pharmacy, Mayo Clinic, Rochester, MN, USA.
| | - Han Wang
- Department of Neurology, Mayo Clinic Health System, Mankato, MN, USA
| | | | | | | | - Fred M Cutrer
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
| | | |
Collapse
|
36
|
Scherf-Clavel M, Weber H, Unterecker S, Müller DJ, Deckert J. Frequencies of CYP2C19 and CYP2D6 gene variants in a German inpatient sample with mood and anxiety disorders. World J Biol Psychiatry 2024; 25:214-221. [PMID: 38493365 DOI: 10.1080/15622975.2024.2321553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 02/12/2024] [Indexed: 03/18/2024]
Abstract
OBJECTIVES Previous results demonstrated that CYP2D6 and CYP2C19 gene variants affect serum concentrations of antidepressants. We implemented a PGx service determining gene variants in CYP2D6 and CYP2C19 in our clinical routine care and report on our first patient cohort. METHODS We analysed CYP2D6 and CYP2C19 allele, genotype, and phenotype frequencies, and actionable pharmacogenetic variants in this German psychiatric inpatient cohort. Two-tailed z-test was used to investigate for differences in CYP2D6 and CYP2C19 phenotypes and actionable/non-actionable genetic variant frequencies between our cohort and reference cohorts. RESULTS Out of the 154 patients included, 44.8% of patients were classified as CYP2D6 normal metabolizer, 38.3% as intermediate metabolizers, 8.4% as poor metabolizers, and 2.6% as ultrarapid metabolizers. As for CYP2C19, 40.9% of patients were classified as normal metabolizers, 19.5% as intermediate metabolizers, 2.6% as poor metabolizers, 31.2% as rapid metabolizers, and 5.8% as ultrarapid metabolizers. Approximately, 80% of patients had at least one actionable PGx variant. CONCLUSION There is a high prevalence of actionable PGx variants in psychiatric inpatients which may affect treatment response. Physicians should refer to PGx-informed dosing guidelines in carriers of these variants. Pre-emptive PGx testing in general may facilitate precision medicine also for other drugs metabolised by CYP2D6 and/or CYP2C19.
Collapse
Affiliation(s)
- Maike Scherf-Clavel
- Department of Psychiatry, Psychosomatics and Psychotherapy, Center of Mental Health, University Hospital of Würzburg, Würzburg, Germany
| | - Heike Weber
- Department of Psychiatry, Psychosomatics and Psychotherapy, Center of Mental Health, University Hospital of Würzburg, Würzburg, Germany
| | - Stefan Unterecker
- Department of Psychiatry, Psychosomatics and Psychotherapy, Center of Mental Health, University Hospital of Würzburg, Würzburg, Germany
| | - Daniel J Müller
- Department of Psychiatry, Psychosomatics and Psychotherapy, Center of Mental Health, University Hospital of Würzburg, Würzburg, Germany
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
- Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
| | - Jürgen Deckert
- Department of Psychiatry, Psychosomatics and Psychotherapy, Center of Mental Health, University Hospital of Würzburg, Würzburg, Germany
| |
Collapse
|
37
|
Loftus J, Levy HP, Stevenson JM. Documentation of results and medication prescribing after combinatorial psychiatric pharmacogenetic testing: A case for discrete results. Genet Med 2024; 26:101056. [PMID: 38153010 DOI: 10.1016/j.gim.2023.101056] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 12/18/2023] [Accepted: 12/20/2023] [Indexed: 12/29/2023] Open
Abstract
PURPOSE Combinatorial pharmacogenetic (PGx) panels intended to aid psychiatric prescribing are available to clinicians. Here, we evaluated the documentation of PGx panel results and subsequent prescribing patterns within a tertiary health care system. METHODS We performed a query of psychiatry service note text in our electronic health record using 71 predefined PGx terms. Patients who underwent combinatorial PGx testing were identified, and documentation of test results was analyzed. Prescription data following testing were examined for the frequency of prescriptions influenced by genes on the panel along with the medical specialties involved. RESULTS A total of 341 patients received combinatorial PGx testing, and documentation of results was found to be absent or incomplete for 198 patients (58%). The predominant method of documentation was through portable document formats uploaded to the electronic health record's "Media" section. Among patients with at least 1 year of follow-up, a large majority (194/228, 85%) received orders for medications affected by the tested genes, including 132 of 228 (58%) patients receiving at least 1 non-psychiatric medication influenced by the test results. CONCLUSION Results from combinatorial PGx testing were poorly documented. Medications affected by these results were often prescribed after testing, highlighting the need for discrete results and clinical decision support.
Collapse
Affiliation(s)
- John Loftus
- Johns Hopkins University School of Medicine, Baltimore, MD
| | - Howard P Levy
- Maryland Primary Care Physicians, Hanover, MD; Division of General Internal Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD; McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD
| | - James M Stevenson
- Division of Clinical Pharmacology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD; Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD.
| |
Collapse
|
38
|
Shen C, Yang H, Shao W, Zheng L, Zhang W, Xie H, Jiang X, Wang L. Physiologically Based Pharmacokinetic Modeling to Unravel the Drug-gene Interactions of Venlafaxine: Based on Activity Score-dependent Metabolism by CYP2D6 and CYP2C19 Polymorphisms. Pharm Res 2024; 41:731-749. [PMID: 38443631 DOI: 10.1007/s11095-024-03680-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Accepted: 02/19/2024] [Indexed: 03/07/2024]
Abstract
BACKGROUND Venlafaxine (VEN) is a commonly utilized medication for alleviating depression and anxiety disorders. The presence of genetic polymorphisms gives rise to considerable variations in plasma concentrations across different phenotypes. This divergence in phenotypic responses leads to notable differences in both the efficacy and tolerance of the drug. PURPOSE A physiologically based pharmacokinetic (PBPK) model for VEN and its metabolite O-desmethylvenlafaxine (ODV) to predict the impact of CYP2D6 and CYP2C19 gene polymorphisms on VEN pharmacokinetics (PK). METHODS The parent-metabolite PBPK models for VEN and ODV were developed using PK-Sim® and MoBi®. Leveraging prior research, derived and implemented CYP2D6 and CYP2C19 activity score (AS)-dependent metabolism to simulate exposure in the drug-gene interactions (DGIs) scenarios. The model's performance was evaluated by comparing predicted and observed values of plasma concentration-time (PCT) curves and PK parameters values. RESULTS In the base models, 91.1%, 94.8%, and 94.6% of the predicted plasma concentrations for VEN, ODV, and VEN + ODV, respectively, fell within a twofold error range of the corresponding observed concentrations. For DGI scenarios, these values were 81.4% and 85% for VEN and ODV, respectively. Comparing CYP2D6 AS = 2 (normal metabolizers, NM) populations to AS = 0 (poor metabolizers, PM), 0.25, 0.5, 0.75, 1.0 (intermediate metabolizers, IM), 1.25, 1.5 (NM), and 3.0 (ultrarapid metabolizers, UM) populations in CYP2C19 AS = 2.0 group, the predicted DGI AUC0-96 h ratios for VEN were 3.65, 3.09, 2.60, 2.18, 1.84, 1.56, 1.34, 0.61, and for ODV, they were 0.17, 0.35, 0.51, 0.64, 0.75, 0.83, 0.90, 1.11, and the results were similar in other CYP2C19 groups. It should be noted that PK differences in CYP2C19 phenotypes were not similar across different CYP2D6 groups. CONCLUSIONS In clinical practice, the impact of genotyping on the in vivo disposition process of VEN should be considered to ensure the safety and efficacy of treatment.
Collapse
Affiliation(s)
- Chaozhuang Shen
- Department of Clinical Pharmacy and Pharmacy Administration, School of Pharmacy, Sichuan University, Chengdu, 610064, West China, China
| | - Hongyi Yang
- Department of Clinical Pharmacy and Pharmacy Administration, School of Pharmacy, Sichuan University, Chengdu, 610064, West China, China
| | - Wenxin Shao
- Anhui Provincial Center for Drug Clinical Evaluation, Yijishan Hospital of Wannan Medical College, Wuhu, 241001, Anhui, China
| | - Liang Zheng
- Department of Clinical Pharmacology, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, Anhui, China
| | - Wei Zhang
- Department of Clinical Pharmacology, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, Anhui, China
| | - Haitang Xie
- Anhui Provincial Center for Drug Clinical Evaluation, Yijishan Hospital of Wannan Medical College, Wuhu, 241001, Anhui, China
| | - Xuehua Jiang
- Department of Clinical Pharmacy and Pharmacy Administration, School of Pharmacy, Sichuan University, Chengdu, 610064, West China, China
| | - Ling Wang
- Department of Clinical Pharmacy and Pharmacy Administration, School of Pharmacy, Sichuan University, Chengdu, 610064, West China, China.
| |
Collapse
|
39
|
Nguyen TK, Vu GM, Duong VC, Pham TL, Nguyen NT, Tran TTH, Tran MH, Nguyen DT, Vo NS, Phung HT, Hoang TH. The therapeutic landscape for COVID-19 and post-COVID-19 medications from genetic profiling of the Vietnamese population and a predictive model of drug-drug interaction for comorbid COVID-19 patients. Heliyon 2024; 10:e27043. [PMID: 38509882 PMCID: PMC10950508 DOI: 10.1016/j.heliyon.2024.e27043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 12/13/2023] [Accepted: 02/22/2024] [Indexed: 03/22/2024] Open
Abstract
Despite the raised awareness of the role of pharmacogenomic (PGx) in personalized medicines for COVID-19, data for COVID-19 drugs is extremely scarce and not even a publication on this topic for post-COVID-19 medications to date. In the current study, we investigated the genetic variations associated with COVID-19 and post-COVID-19 therapies by using whole genome sequencing data of the 1000 Vietnamese Genomes Project (1KVG) in comparison with other populations retrieved from the 1000 Genomes Project Phase 3 (1KGP3) and the Genome Aggregation Database (gnomAD). Moreover, we also evaluated the risk of drug interactions in comorbid COVID-19 and post-COVID-19 patients based on pharmacogenomic profiles of drugs using a computational approach. For COVID-19 therapies, variants related to the response of two causal treatment agents (tolicizumab and ritonavir) and antithrombotic drugs are common in the Vietnamese cohort. Regarding post-COVID-19, drugs for mental manipulations possess the highest number of clinical annotated variants carried by Vietnamese individuals. Among the superpopulations, East Asian populations shared the most similar genetic structure with the Vietnamese population, whereas the African population showed the most difference. Comorbid patients are at an increased drug-drug interaction (DDI) risk when suffering from COVID-19 and after recovering as well due to a large number of potential DDIs which have been identified. Our results presented the population-specific understanding of the pharmacogenomic aspect of COVID-19 and post-COVID-19 therapy to optimize therapeutic outcomes and promote personalized medicine strategy. We also partly clarified the higher risk in COVID-19 patients with underlying conditions by assessing the potential drug interactions.
Collapse
Affiliation(s)
| | - Giang Minh Vu
- Center for Biomedical Informatics, Vingroup Big Data Institute, Hanoi, Viet Nam
- GeneStory JSC, Hanoi, Viet Nam
| | - Vinh Chi Duong
- Center for Biomedical Informatics, Vingroup Big Data Institute, Hanoi, Viet Nam
- GeneStory JSC, Hanoi, Viet Nam
| | | | | | - Trang Thi Ha Tran
- Center for Biomedical Informatics, Vingroup Big Data Institute, Hanoi, Viet Nam
- GeneStory JSC, Hanoi, Viet Nam
| | - Mai Hoang Tran
- Center for Biomedical Informatics, Vingroup Big Data Institute, Hanoi, Viet Nam
- GeneStory JSC, Hanoi, Viet Nam
| | - Duong Thuy Nguyen
- Center for Biomedical Informatics, Vingroup Big Data Institute, Hanoi, Viet Nam
- GeneStory JSC, Hanoi, Viet Nam
| | - Nam S. Vo
- Center for Biomedical Informatics, Vingroup Big Data Institute, Hanoi, Viet Nam
- GeneStory JSC, Hanoi, Viet Nam
| | - Huong Thanh Phung
- Faculty of Biotechnology, Hanoi University of Pharmacy, Hanoi, Viet Nam
| | - Tham Hong Hoang
- Center for Biomedical Informatics, Vingroup Big Data Institute, Hanoi, Viet Nam
- GeneStory JSC, Hanoi, Viet Nam
| |
Collapse
|
40
|
Polasek TM. Pharmacogenomics - a minor rather than major force in clinical medicine. Expert Rev Clin Pharmacol 2024; 17:203-212. [PMID: 38307498 DOI: 10.1080/17512433.2024.2314726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 02/01/2024] [Indexed: 02/04/2024]
Abstract
INTRODUCTION Pharmacogenomics (PGx) is touted as essential for the future of precision medicine. But the opportunity cost of PGx from the prescribers' perspective is rarely considered. The aim of this article is to critique PGx-guided prescribing using clinical pharmacology principles so that important cases for PGx testing are not missed by doctors responsible for therapeutic decision making. AREAS COVERED Three categories of PGx and their limitations are outlined - exposure PGx, response PGx, and immune-mediated safety PGx. Clinical pharmacology reasons are given for the narrow scope of PGx-guided prescribing apart from a few medical specialties. Clinical problems for doctors that may arise from PGx are then explained, including mismatch between patients' expectations of PGx testing and the benefits or answers it provides. EXPERT OPINION Contrary to popular opinion, PGx is unlikely to become the cornerstone of precision medicine. Sound clinical pharmacology reasons explain why PGx-guided prescribing is unnecessary for most drugs. Pharmacogenomics is important for niche areas of prescribing but has limited clinical utility more broadly. The opportunity cost of PGx-guided prescribing is currently too great for most doctors.
Collapse
Affiliation(s)
- Thomas M Polasek
- Centre for Medicine Use and Safety, Monash University, Melbourne, Australia
- CMAX Clinical Research, Adelaide, Australia
| |
Collapse
|
41
|
Scherf-Clavel M, Weber H, Unterecker S, Frantz A, Eckert A, Reif A, Deckert J, Hahn M. The Relevance of Integrating CYP2C19 Phenoconversion Effects into Clinical Pharmacogenetics. PHARMACOPSYCHIATRY 2024; 57:69-77. [PMID: 38354747 PMCID: PMC10948286 DOI: 10.1055/a-2248-6924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 12/25/2023] [Indexed: 02/16/2024]
Abstract
INTRODUCTION CYP2D6 and CYP2C19 functional status as defined by genotype is modulated by phenoconversion (PC) due to pharmacokinetic interactions. As of today, there is no data on the effect size of PC for CYP2C19 functional status. The primary aim of this study was to investigate the impact of PC on CYP2C19 functional status. METHODS Two patient cohorts (total n=316; 44.2±15.4 years) were investigated for the functional enzyme status of CYP2C19 applying two different correction methods (PCBousman, PCHahn&Roll) as well as serum concentration and metabolite-to-parent ratio of venlafaxine, amitriptyline, mirtazapine, sertraline, escitalopram, risperidone, and quetiapine. RESULTS There was a decrease in the number of normal metabolizers of CYP2C19 and an increase in the number of poor metabolizers. When controlled for age, sex, and, in the case of amitriptyline, venlafaxine, and risperidone, CYP2D6 functional enzyme status, an association was observed between the CYP2C19 phenotype/functional enzyme status and serum concentration of amitriptyline, sertraline, and escitalopram. DISCUSSION PC of CYP2C19 changes phenotypes but does not improve correlations with serum concentrations. However, only a limited number of patients received perturbators of CYP2C19. Studies with large numbers of patients are still lacking, and thus, it cannot be decided if there are minor differences and which method of correction to use. For the time being, PC is relevant in individual patients treated with CYP2C19-affecting drugs, for example, esomeprazole. To ensure adequate serum concentrations in these patients, this study suggests the use of therapeutic drug monitoring.
Collapse
Affiliation(s)
- Maike Scherf-Clavel
- Department of Psychiatry, Psychosomatics and Psychotherapy, Center of
Mental Health, University Hospital of Würzburg, 97080 Würzburg,
Germany
| | - Heike Weber
- Department of Psychiatry, Psychosomatics and Psychotherapy, Center of
Mental Health, University Hospital of Würzburg, 97080 Würzburg,
Germany
- Department of Psychiatry, Psychosomatics and Psychotherapy, University
Hospital Frankfurt, 60528 Frankfurt, Germany
| | - Stefan Unterecker
- Department of Psychiatry, Psychosomatics and Psychotherapy, Center of
Mental Health, University Hospital of Würzburg, 97080 Würzburg,
Germany
| | - Amelie Frantz
- Department of Psychiatry, Psychosomatics and Psychotherapy, University
Hospital Frankfurt, 60528 Frankfurt, Germany
| | - Andreas Eckert
- Department of Psychiatry, Psychosomatics and Psychotherapy, University
Hospital Frankfurt, 60528 Frankfurt, Germany
| | - Andreas Reif
- Department of Psychiatry, Psychosomatics and Psychotherapy, University
Hospital Frankfurt, 60528 Frankfurt, Germany
| | - Jürgen Deckert
- Department of Psychiatry, Psychosomatics and Psychotherapy, Center of
Mental Health, University Hospital of Würzburg, 97080 Würzburg,
Germany
| | - Martina Hahn
- Department of Psychiatry, Psychosomatics and Psychotherapy, University
Hospital Frankfurt, 60528 Frankfurt, Germany
- Department of Mental Health, Varisano Hospital Frankfurt Hoechst, 65929
Frankfurt, Germany
| |
Collapse
|
42
|
Stern S, Hyland PL, Pacanowski M, Schuck RN. Leveraging in Vitro Models for Clinically Relevant Rare CYP2D6 Variants in Pharmacogenomics. Drug Metab Dispos 2024; 52:159-170. [PMID: 38167410 PMCID: PMC10877705 DOI: 10.1124/dmd.123.001512] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 11/09/2023] [Accepted: 12/19/2023] [Indexed: 01/05/2024] Open
Abstract
Cytochrome P450 2D6 (CYP2D6) is responsible for the metabolism of up to 20% of small-molecule drugs and therefore, may impact the safety and efficacy of medicines in broad therapeutic areas. CYP2D6 is highly polymorphic, and the frequency of variants can differ across racial and ethnic populations, significantly affecting enzymatic function and drug metabolism. However, rare variants of CYP2D6 present a unique challenge for academia, industry, and regulatory agencies alike due to the lack of feasibility of characterizing their clinical relevance in clinical trials, particularly in variants that exhibit population-specific frequencies in racial and ethnic groups that are poorly represented in clinical trials. Despite significant advancement in pharmacogenomics, the substrate specificity and related clinical relevance of these CYP2D6 rare variants remain largely unclear, and further efforts are warranted to characterize the burden of these variants on adverse drug reactions and drug efficacy. Thus, cell-based in vitro systems can be used to inform substrate-specific effects and the overall relevance of a rare variant. Liver microsomes, cell-based expression systems, ex vivo primary samples, and purified variant protein have all been used with various substrates to potentially predict the clinical impact of new substrates. In this review, we identify rare variants of CYP2D6 that demonstrate differences across races in prevalence and thus are often unassessed in clinical trials. Accordingly, we examine current pharmacogenomic in vitro models used to analyze the functional impact of these rare variants in a substrate-specific manner. SIGNIFICANCE STATEMENT: Variants of CYP2D6 play a clinically relevant role in drug metabolism, leading to potential safety and efficacy concerns. Although the influence of prevalent variants is often well characterized, rare variants are traditionally not included in clinical trials. This review captures the clinical relevance of rare variants in CYP2D6 by highlighting in vitro models that analyze their impact on the metabolism of CYP2D6 substrates.
Collapse
Affiliation(s)
- Sydney Stern
- Center for Drug Evaluation and Research, Office of Translational Science, Office of Clinical Pharmacology, US Food and Drug Administration, Silver Spring, Maryland
| | - Paula L Hyland
- Center for Drug Evaluation and Research, Office of Translational Science, Office of Clinical Pharmacology, US Food and Drug Administration, Silver Spring, Maryland
| | - Michael Pacanowski
- Center for Drug Evaluation and Research, Office of Translational Science, Office of Clinical Pharmacology, US Food and Drug Administration, Silver Spring, Maryland
| | - Robert N Schuck
- Center for Drug Evaluation and Research, Office of Translational Science, Office of Clinical Pharmacology, US Food and Drug Administration, Silver Spring, Maryland
| |
Collapse
|
43
|
Hernandez M, Cullell N, Cendros M, Serra-Llovich A, Arranz MJ. Clinical Utility and Implementation of Pharmacogenomics for the Personalisation of Antipsychotic Treatments. Pharmaceutics 2024; 16:244. [PMID: 38399298 PMCID: PMC10893329 DOI: 10.3390/pharmaceutics16020244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 01/24/2024] [Accepted: 01/29/2024] [Indexed: 02/25/2024] Open
Abstract
Decades of pharmacogenetic research have revealed genetic biomarkers of clinical response to antipsychotics. Genetic variants in antipsychotic targets, dopamine and serotonin receptors in particular, and in metabolic enzymes have been associated with the efficacy and toxicity of antipsychotic treatments. However, genetic prediction of antipsychotic response based on these biomarkers is far from accurate. Despite the clinical validity of these findings, the clinical utility remains unclear. Nevertheless, genetic information on CYP metabolic enzymes responsible for the biotransformation of most commercially available antipsychotics has proven to be effective for the personalisation of clinical dosing, resulting in a reduction of induced side effects and in an increase in efficacy. However, pharmacogenetic information is rarely used in psychiatric settings as a prescription aid. Lack of studies on cost-effectiveness, absence of clinical guidelines based on pharmacogenetic biomarkers for several commonly used antipsychotics, the cost of genetic testing and the delay in results delivery hamper the implementation of pharmacogenetic interventions in clinical settings. This narrative review will comment on the existing pharmacogenetic information, the clinical utility of pharmacogenetic findings, and their current and future implementations.
Collapse
Affiliation(s)
- Marta Hernandez
- PHAGEX Research Group, University Ramon Llull, 08022 Barcelona, Spain;
- School of Health Sciences Blanquerna, University Ramon Llull, 08022 Barcelona, Spain
| | - Natalia Cullell
- Fundació Docència i Recerca Mútua Terrassa, 08221 Terrassa, Spain; (N.C.); (A.S.-L.)
- Department of Neurology, Hospital Universitari Mútua Terrassa, 08221 Terrassa, Spain
| | - Marc Cendros
- EUGENOMIC Genómica y Farmacogenética, 08029 Barcelona, Spain;
| | | | - Maria J. Arranz
- PHAGEX Research Group, University Ramon Llull, 08022 Barcelona, Spain;
- Fundació Docència i Recerca Mútua Terrassa, 08221 Terrassa, Spain; (N.C.); (A.S.-L.)
| |
Collapse
|
44
|
Bharthi K, Zuberi R, Maruf AA, Shaheen SM, McCloud R, Heintz M, McAusland L, Arnold PD, Bousman CA. Impact of Cytochrome P450 Genetic Variation on Patient-Reported Symptom Improvement and Side Effects Among Children and Adolescents Treated with Fluoxetine. J Child Adolesc Psychopharmacol 2024; 34:21-27. [PMID: 38377520 DOI: 10.1089/cap.2023.0039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/22/2024]
Abstract
Background: Clinical practice guidelines recommend the use of fluoxetine, a selective serotonin reuptake inhibitor (SSRI), as a first-line pharmacotherapy for major depressive disorder (MDD) and obsessive compulsive disorder (OCD) in children and adolescents. However, response and tolerability to fluoxetine varies from child to child, which may in part, be a result of interindividual differences in fluoxetine metabolism. In this study, we examined whether genotype-predicted activity scores of cytochrome P450 enzymes were associated with patient-reported symptom improvement and side effects in children and adolescents treated with fluoxetine. Methods: Ninety children and adolescents aged 7-18 with a MDD or OCD diagnosis and a history of fluoxetine treatment were recruited from Western Canada. For each participant, fluoxetine dose and duration information were collected, as well as questions about adherence, side effects, and symptom improvement. DNA was extracted from a saliva sample and genotyped for CYP2D6, CYP2C19, CYP2C9, CYP3A4, and CYP3A5. Logistic regression models were fitted to assess the impact of activity scores on symptom improvement and side effects. Results: Increased CYP2D6 activity score was significantly associated with reduced odds of symptom improvement (odds ratio [OR] = 0.46, 95% confidence interval [CI] = 0.23-0.91, p = 0.028) as well as a trend association with reduced side effects (OR = 0.49, 95% CI = 0.22-1.07, p = 0.072), after adjusting for age, sex, diagnosis, dose, duration, adherence, and activity scores of the other assessed CYP enzymes. No associations with symptom improvement or side effects were detected for the other CYP enzymes examined. Conclusions: Our results suggest that an increase in the genotype-predicted CYP2D6 activity score was associated with a decrease in the odds of reporting symptom improvement among children and adolescents treated with fluoxetine. These findings will contribute to future updates of pharmacogenetic-based SSRI prescribing guidelines and if replicated, could inform fluoxetine treatment in children and adolescents with MDD or OCD. Clinical Trial Registration: NCT04797364.
Collapse
Affiliation(s)
- Kanika Bharthi
- Department of Biological Sciences, University of Calgary, Calgary, Canada
| | - Rayyan Zuberi
- Department of Medical Genetics, University of Calgary, Calgary, Canada
| | - Abdullah Al Maruf
- College of Pharmacy, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Canada
- The Mathison Centre for Mental Health Research and Education, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Canada
- Department of Psychiatry, University of Calgary, Calgary, Canada
| | - Sarker M Shaheen
- The Mathison Centre for Mental Health Research and Education, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Canada
- Department of Psychiatry, University of Calgary, Calgary, Canada
| | - Ryden McCloud
- The Mathison Centre for Mental Health Research and Education, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Canada
| | - Madison Heintz
- Department of Medical Genetics, University of Calgary, Calgary, Canada
- The Mathison Centre for Mental Health Research and Education, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Canada
| | - Laina McAusland
- Department of Biological Sciences, University of Calgary, Calgary, Canada
- Department of Medical Genetics, University of Calgary, Calgary, Canada
- The Mathison Centre for Mental Health Research and Education, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Canada
| | - Paul D Arnold
- Department of Medical Genetics, University of Calgary, Calgary, Canada
- The Mathison Centre for Mental Health Research and Education, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Canada
- Department of Psychiatry, University of Calgary, Calgary, Canada
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Canada
| | - Chad A Bousman
- Department of Medical Genetics, University of Calgary, Calgary, Canada
- The Mathison Centre for Mental Health Research and Education, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Canada
- Department of Psychiatry, University of Calgary, Calgary, Canada
- Department of Physiology and Pharmacology, University of Calgary, Calgary, Canada
- Department of Community Health Sciences, University of Calgary, Calgary, Canada
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Canada
| |
Collapse
|
45
|
Malik S, Verma P, Ruaño G, Al Siaghy A, Dilawar A, Bishop JR, Strawn JR, Namerow LB. Pharmacogenetics in Child and Adolescent Psychiatry: Background and Evidence-Based Clinical Applications. J Child Adolesc Psychopharmacol 2024; 34:4-20. [PMID: 38377525 DOI: 10.1089/cap.2023.0074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/22/2024]
Abstract
The efficacy and tolerability of psychotropic medications can vary significantly among children and adolescents, and some of this variability relates to pharmacogenetic factors. Pharmacogenetics (PGx) in child and adolescent psychiatry can potentially improve treatment outcomes and minimize adverse drug reactions. This article reviews key pharmacokinetic and pharmacodynamic genes and principles of pharmacogenetic testing and discusses the evidence base for clinical decision-making concerning PGx testing. This article reviews current guidelines from the United States Food and Drug Administration (FDA), the Clinical Pharmacogenetics Implementation Consortium (CPIC), and the Dutch Pharmacogenetics Working Group (DPWG) and explores potential future directions. This review discusses key clinical considerations for clinicians prescribing psychotropic medications in children and adolescents, focusing on antidepressants, antipsychotics, stimulants, norepinephrine reuptake inhibitors, and alpha-2 agonists. Finally, this review synthesizes the practical use of pharmacogenetic testing and clinical decision support systems.
Collapse
Affiliation(s)
- Salma Malik
- Department of Psychiatry, University of Connecticut School of Medicine, Farmington, Connecticut, USA
- Division of Child and Adolescent Psychiatry, Institute of Living/Hartford Hospital, Hartford, Connecticut, USA
| | - Pragya Verma
- Division of Child and Adolescent Psychiatry, Institute of Living/Hartford Hospital, Hartford, Connecticut, USA
| | - Gualberto Ruaño
- Department of Psychiatry, University of Connecticut School of Medicine, Farmington, Connecticut, USA
| | - Areej Al Siaghy
- Division of Child and Adolescent Psychiatry, Institute of Living/Hartford Hospital, Hartford, Connecticut, USA
| | | | - Jeffrey R Bishop
- Department of Experimental and Clinical Pharmacology, University of Minnesota College of Pharmacy, Minneapolis, Minnesota, USA
- Department of Psychiatry, University of Minnesota Medical School, Minneapolis, Minnesota, USA
| | - Jeffrey R Strawn
- Department of Psychiatry & Behavioral Neuroscience, University of Cincinnati, College of Medicine, Cincinnati, Ohio, USA
| | - Lisa B Namerow
- Department of Psychiatry, University of Connecticut School of Medicine, Farmington, Connecticut, USA
| |
Collapse
|
46
|
Tonti E, Lee YM, Gruenke N, Ferren J, Stutzman DL. Impact of Pharmacogenomics on Pediatric Psychotropic Medication Prescribing in an Ambulatory Care Setting. J Child Adolesc Psychopharmacol 2024; 34:52-60. [PMID: 38377527 DOI: 10.1089/cap.2023.0087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/22/2024]
Abstract
Objective: Evidence for pharmacogenomic (PGx) guided treatment in child and adolescent psychiatry is growing. This study evaluated the impact of PGx testing on psychotropic medication prescribing in an ambulatory child and adolescent psychiatry and a developmental pediatrics clinic. Methods: This was a single-center, retrospective, descriptive analysis of patients who underwent PGx testing between January 2015 and October 2022 at a child and adolescent psychiatry clinic or developmental pediatrics clinic. The primary outcome was the proportion of patients with at least one psychotropic medication modification made 6-month posttesting that could be attributed to CYP2C19, CYP2D6, HLA-B*15:02, or HLA-A*31:01. Secondary outcomes included reason for testing, types of therapeutic modifications made, and whether the therapeutic modifications concorded with PGx guidelines. Results: A total of 193 patients were analyzed. The average age was 10 ± 4 years old, 60% were male, 78% were Caucasian. Sixty-eight percent had a primary diagnosis of a neurodevelopmental disorder, namely autism spectrum disorder (51%), and attention-deficit/hyperactivity disorder (14%). The reasons for PGx testing included medication inefficacy (34%), medication intolerance (20%), and family request (19%). At the time of PGx testing, 37% of patients were taking ≥1 psychotropic medication with PGx annotation. Overall, 35 PGx-related therapeutic modifications were made in 32 (17%) patients. These included continuing current PGx medication (6.2%) and starting PGx medication (5.2%). These modifications mainly involved antidepressants. Out of these 35 PGx-related therapeutic modifications, 94% were concordant with PGx guidelines. Among 29 patients who were prescribed at least one CYP2D6 inhibitor, 25 (86%) underwent CYP2D6 phenoconversion. Conclusions: It is critical to apply pediatric age-specific considerations when utilizing PGx testing in child and adolescent psychiatry. PGx testing stewardship could provide a framework to guide the clinical utility of PGx in a pediatric population with mental health conditions, including neurodevelopmental disorders.
Collapse
Affiliation(s)
- Erica Tonti
- Department of Pharmacy, Children's Hospital Colorado, Aurora, Colorado, USA
| | - Yee Ming Lee
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado, Aurora, Colorado, USA
| | - Nathan Gruenke
- Department of Pharmacy, Children's Hospital Colorado, Aurora, Colorado, USA
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado, Aurora, Colorado, USA
| | - Janie Ferren
- Department of Pharmacy, Children's Hospital Colorado, Aurora, Colorado, USA
- Pediatric Mental Health Institute, Children's Hospital Colorado, Aurora, Colorado, USA
| | - Danielle L Stutzman
- Department of Pharmacy, Children's Hospital Colorado, Aurora, Colorado, USA
- Pediatric Mental Health Institute, Children's Hospital Colorado, Aurora, Colorado, USA
- Department of Clinical Pharmacy, Skaggs School of Pharmacy and Pharmaceutical Science, University of Colorado, Aurora, Colorado, USA
- Child and Adolescent Mental Health Division Department of Psychiatry, School of Medicine, University of Colorado, Aurora, Colorado, USA
| |
Collapse
|
47
|
Pelgrim TAD, Philipsen A, Young AH, Juruena M, Jimenez E, Vieta E, Jukić M, Van der Eycken E, Heilbronner U, Moldovan R, Kas MJH, Jagesar RR, Nöthen MM, Hoffmann P, Shomron N, Kilarski LL, van Amelsvoort T, Campforts B, van Westrhenen R. A New Intervention for Implementation of Pharmacogenetics in Psychiatry: A Description of the PSY-PGx Clinical Study. Pharmaceuticals (Basel) 2024; 17:151. [PMID: 38399366 PMCID: PMC10892863 DOI: 10.3390/ph17020151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 01/15/2024] [Accepted: 01/18/2024] [Indexed: 02/25/2024] Open
Abstract
(1) Background Pharmacological treatment for psychiatric disorders has shown to only be effective in about one-third of patients, as it is associated with frequent treatment failure, often because of side effects, and a long process of trial-and-error pharmacotherapy until an effective and tolerable treatment is found. This notion emphasizes the urgency for a personalized medicine approach in psychiatry. (2) Methods This prospective patient- and rater-blinded, randomized, controlled study will investigate the effect of dose-adjustment of antidepressants escitalopram and sertraline or antipsychotics risperidone and aripiprazole according to the latest state-of-the-art international dosing recommendations for CYP2C19 and CYP2D6 metabolizer status in patients with mood, anxiety, and psychotic disorders. A total sample of N = 2500 will be recruited at nine sites in seven countries (expected drop-out rate of 30%). Patients will be randomized to a pharmacogenetic group or a dosing-as-usual group and treated over a 24-week period with four study visits. The primary outcome is personal recovery using the Recovery Assessment Scale as assessed by the patient (RAS-DS), with secondary outcomes including clinical effects (response or symptomatic remission), side effects, general well-being, digital phenotyping, and psychosocial functioning. (3) Conclusions This is, to our knowledge, the first international, multi-center, non-industry-sponsored randomized controlled trial (RCT) that may provide insights into the effectiveness and utility of implementing pharmacogenetic-guided treatment of psychiatric disorders, and as such, results will be incorporated in already available dosing guidelines.
Collapse
Affiliation(s)
- Teuntje A. D. Pelgrim
- Department of Psychiatry, Parnassia Psychiatric Institute, 1062HN Amsterdam, The Netherlands
| | - Alexandra Philipsen
- Department of Psychiatry and Psychotherapy, University of Bonn, 53105 Bonn, Germany
| | - Allan H. Young
- Department of Psychological Medicine, Institute of Psychiatry, Psychology and Neuroscience, King’s College London & South London and Maudsley NHS Foundation Trust, Bethlem Royal Hospital, Monks Orchard Road Beckenham, Kent BR3 3BX, UK
| | - Mario Juruena
- Department of Psychological Medicine, Institute of Psychiatry, Psychology and Neuroscience, King’s College London & South London and Maudsley NHS Foundation Trust, Bethlem Royal Hospital, Monks Orchard Road Beckenham, Kent BR3 3BX, UK
| | - Ester Jimenez
- Bipolar and Depressive Disorders Unit, Department of Psychiatry and Psychology, Hospital Clinic & Institute of Neurosciences (UBNeuro), IDIBAPS, CIBERSAM, ISCIII, University of Barcelona, 08036 Catalonia, Spain
| | - Eduard Vieta
- Bipolar and Depressive Disorders Unit, Department of Psychiatry and Psychology, Hospital Clinic & Institute of Neurosciences (UBNeuro), IDIBAPS, CIBERSAM, ISCIII, University of Barcelona, 08036 Catalonia, Spain
| | - Marin Jukić
- Department of Physiology, Faculty of Pharmacy, University of Belgrade, 11000 Belgrade, Serbia
- Department of Physiology & Pharmacology, Karolinska Institute, 171 77 Stockholm, Sweden
| | - Erik Van der Eycken
- Global Alliance of Mental Illness Advocacy Networks-Europe (GAMIAN-Europe), 1050 Brussels, Belgium
| | - Urs Heilbronner
- Institute of Psychiatric Phenomics and Genomics (IPPG), LMU University Hospital, LMU Munich, 80336 Munich, Germany
| | - Ramona Moldovan
- Department of Psychology, Babeş-Bolyai University, 400015 Cluj-Napoca, Romania
- Division of Evolution, Infection and Genomic Sciences, School of Biological Sciences, University of Manchester, Manchester M13 9PT, UK
- Manchester Center for Genomic Medicine, St. Mary’s Hospital, Manchester University NHS Foundation Trust, Manchester M13 9WL, UK
| | - Martien J. H. Kas
- Groningen Institute for Evolutionary Life Sciences, University of Groningen, 9700CC Groningen, The Netherlands
| | - Raj R. Jagesar
- Groningen Institute for Evolutionary Life Sciences, University of Groningen, 9700CC Groningen, The Netherlands
| | - Markus M. Nöthen
- Institute of Human Genetics, University Hospital of Bonn and University of Bonn, 53127 Bonn, Germany
| | - Per Hoffmann
- Institute of Human Genetics, University Hospital of Bonn and University of Bonn, 53127 Bonn, Germany
| | - Noam Shomron
- Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Laura L. Kilarski
- Department of Psychiatry and Psychotherapy, University of Bonn, 53105 Bonn, Germany
| | - Thérèse van Amelsvoort
- Department of Psychiatry and Neuropsychology, Maastricht University, 6226NB Maastricht, The Netherlands
| | - Bea Campforts
- Department of Psychiatry and Neuropsychology, Maastricht University, 6226NB Maastricht, The Netherlands
| | | | - Roos van Westrhenen
- Department of Psychiatry, Parnassia Psychiatric Institute, 1062HN Amsterdam, The Netherlands
- Department of Psychological Medicine, Institute of Psychiatry, Psychology and Neuroscience, King’s College London & South London and Maudsley NHS Foundation Trust, Bethlem Royal Hospital, Monks Orchard Road Beckenham, Kent BR3 3BX, UK
- Department of Psychiatry and Neuropsychology, Maastricht University, 6226NB Maastricht, The Netherlands
- St. John’s National Academy of Health Sciences, Bangalore 560034, India
| |
Collapse
|
48
|
Tremmel R, Hofmann U, Haag M, Schaeffeler E, Schwab M. Circulating Biomarkers Instead of Genotyping to Establish Metabolizer Phenotypes. Annu Rev Pharmacol Toxicol 2024; 64:65-87. [PMID: 37585662 DOI: 10.1146/annurev-pharmtox-032023-121106] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/18/2023]
Abstract
Pharmacogenomics (PGx) enables personalized treatment for the prediction of drug response and to avoid adverse drug reactions. Currently, PGx mainly relies on the genetic information of absorption, distribution, metabolism, and excretion (ADME) targets such as drug-metabolizing enzymes or transporters to predict differences in the patient's phenotype. However, there is evidence that the phenotype-genotype concordance is limited. Thus, we discuss different phenotyping strategies using exogenous xenobiotics (e.g., drug cocktails) or endogenous compounds for phenotype prediction. In particular, minimally invasive approaches focusing on liquid biopsies offer great potential to preemptively determine metabolic and transport capacities. Early studies indicate that ADME phenotyping using exosomes released from the liver is reliable. In addition, pharmacometric modeling and artificial intelligence improve phenotype prediction. However, further prospective studies are needed to demonstrate the clinical utility of individualized treatment based on phenotyping strategies, not only relying on genetics. The present review summarizes current knowledge and limitations.
Collapse
Affiliation(s)
- Roman Tremmel
- Dr. Margarete Fischer-Bosch-Institute of Clinical Pharmacology, Stuttgart, Germany;
- University of Tuebingen, Tuebingen, Germany
| | - Ute Hofmann
- Dr. Margarete Fischer-Bosch-Institute of Clinical Pharmacology, Stuttgart, Germany;
- University of Tuebingen, Tuebingen, Germany
| | - Mathias Haag
- Dr. Margarete Fischer-Bosch-Institute of Clinical Pharmacology, Stuttgart, Germany;
- University of Tuebingen, Tuebingen, Germany
| | - Elke Schaeffeler
- Dr. Margarete Fischer-Bosch-Institute of Clinical Pharmacology, Stuttgart, Germany;
- University of Tuebingen, Tuebingen, Germany
- Cluster of Excellence iFIT (EXC2180) "Image-Guided and Functionally Instructed Tumor Therapies," University of Tuebingen, Tuebingen, Germany
| | - Matthias Schwab
- Dr. Margarete Fischer-Bosch-Institute of Clinical Pharmacology, Stuttgart, Germany;
- University of Tuebingen, Tuebingen, Germany
- Cluster of Excellence iFIT (EXC2180) "Image-Guided and Functionally Instructed Tumor Therapies," University of Tuebingen, Tuebingen, Germany
- Departments of Clinical Pharmacology, and Pharmacy and Biochemistry, University of Tuebingen, Tuebingen, Germany
- German Cancer Consortium (DKTK), German Cancer Research Center Heidelberg (DKFZ), Partner Site, Tübingen, Germany
| |
Collapse
|
49
|
Hurrell T, Naidoo J, Masimirembwa C, Scholefield J. The Case for Pre-Emptive Pharmacogenetic Screening in South Africa. J Pers Med 2024; 14:114. [PMID: 38276236 PMCID: PMC10817273 DOI: 10.3390/jpm14010114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 01/12/2024] [Accepted: 01/15/2024] [Indexed: 01/27/2024] Open
Abstract
Lack of equitable representation of global genetic diversity has hampered the implementation of genomic medicine in under-represented populations, including those on the African continent. Data from the multi-national Pre-emptive Pharmacogenomic Testing for Preventing Adverse Drug Reactions (PREPARE) study suggest that genotype guidance for prescriptions reduced the incidence of clinically relevant adverse drug reactions (ADRs) by 30%. In this study, hospital dispensary trends from a tertiary South African (SA) hospital (Steve Biko Academic Hospital; SBAH) were compared with the drugs monitored in the PREPARE study. Dispensary data on 29 drugs from the PREPARE study accounted for ~10% of total prescriptions and ~9% of the total expenditure at SBAH. VigiLyze data from the South African Health Products Regulatory Authority were interrogated for local ADRs related to these drugs; 27 were listed as being suspected, concomitant, or interacting in ADR reports. Furthermore, a comparison of pharmacogene allele frequencies between African and European populations was used to frame the potential impact of pre-emptive pharmacogenetic screening in SA. Enumerating the benefit of pre-emptive pharmacogenetic screening in SA will only be possible once we initiate its full application. However, regional genomic diversity, disease burden, and first-line treatment options could be harnessed to target stratified PGx today.
Collapse
Affiliation(s)
- Tracey Hurrell
- Bioengineering and Integrated Genomics Group, Future Production Chemicals Cluster, Council for Scientific and Industrial Research, Pretoria 0001, South Africa; (T.H.); (J.N.)
| | - Jerolen Naidoo
- Bioengineering and Integrated Genomics Group, Future Production Chemicals Cluster, Council for Scientific and Industrial Research, Pretoria 0001, South Africa; (T.H.); (J.N.)
| | - Collen Masimirembwa
- African Institute of Biomedical Science and Technology, Harare 00263, Zimbabwe;
- Sydney Brenner Institute for Molecular Biology, Division of Human Genetics, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg 2193, South Africa
| | - Janine Scholefield
- Bioengineering and Integrated Genomics Group, Future Production Chemicals Cluster, Council for Scientific and Industrial Research, Pretoria 0001, South Africa; (T.H.); (J.N.)
- Department of Human Biology, Faculty of Health Sciences, University of Cape Town, Cape Town 7925, South Africa
- Division of Human Genetics, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg 2193, South Africa
| |
Collapse
|
50
|
Jory J, Handelman K. Sudden-Onset Acute Obsessive-Compulsive Disorder Associated with Streptococcus and Brain MRI Hyperintensity in a Young Adult. Healthcare (Basel) 2024; 12:226. [PMID: 38255113 PMCID: PMC10815760 DOI: 10.3390/healthcare12020226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 01/06/2024] [Accepted: 01/12/2024] [Indexed: 01/24/2024] Open
Abstract
BACKGROUND Pediatric autoimmune neuropsychiatric disorders associated with streptococcal (strep) infections (PANDAS) are a recognized medical entity among children. But evidence for strep-mediated sudden-onset obsessive-compulsive disorder (OCD) in young adults is very limited. Delayed strep assessment and treatment may negatively impact clinical outcomes. METHODS We describe a young adult with acute sudden-onset OCD (age 24), treated unsuccessfully with medication and therapy for 3 years. At age 27, antistreptolysin-O (ASO) was tested, based on extensive pediatric history of strep infections. Antibiotic treatment was initiated. RESULTS Magnetic resonance imaging (MRI) identified a new temporal lobe hyperintensity at OCD onset (age 24), which persisted at ages 25 and 30. ASO titers were elevated from age 27 through 29. Following Amoxicillin treatment, ASO initially increased. Subsequent Amoxicillin + Clavulin treatment produced improved OCD symptoms and treatment response, with no adverse effects. CONCLUSION These results strongly suggest an association among strep infection, neuro-inflammation and sudden-onset OCD in this young adult whose response to medication and therapy was successful only after high-dose antibiotic intervention. Greater OCD remission potential may be possible with earlier identification and antibiotic treatment than 3 years post OCD onset. These findings add to the limited literature on strep as an etiology of the sudden-onset of OCD in young adults. They also lend urgency to increased frontline awareness for early strep and ASO assessment in sudden-onset acute OCD among young adults.
Collapse
Affiliation(s)
- Joan Jory
- Department of Family Relations and Applied Nutrition, University of Guelph, Guelph, ON L8N 3K7, Canada
| | - Kenneth Handelman
- Department of Psychiatry and Behavioural Neuroscience, McMaster University, Hamilton, ON L8N 3K7, Canada;
| |
Collapse
|