1
|
De Brabander EY, van Amelsvoort T, van Westrhenen R. Unidentified CYP2D6 genotype does not affect pharmacological treatment for patients with first episode psychosis. J Psychopharmacol 2024; 38:1111-1121. [PMID: 39344086 PMCID: PMC11528939 DOI: 10.1177/02698811241279022] [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] [Indexed: 10/01/2024]
Abstract
BACKGROUND Research on the pharmacogenetic influence of hepatic CYP450 enzyme 2D6 (CYP2D6) on metabolism of drugs for psychosis and associated outcome has been inconclusive. Some results suggest increased risk of adverse reactions in poor and intermediate metabolizers, while others find no relationship. However, retrospective designs may fail to account for the long-term pharmacological treatment of patients. Previous studies found that clinicians adapted risperidone dose successfully without knowledge of patient CYP2D6 phenotype. AIM Here, we aimed to replicate the results of those studies in a Dutch cohort of patients with psychosis (N = 418) on pharmacological treatment. METHOD We compared chlorpromazine-equivalent dose between CYP2D6 metabolizer phenotypes and investigated which factors were associated with dosage. This was repeated in two smaller subsets; patients prescribed pharmacogenetics-actionable drugs according to published guidelines, and risperidone-only as done previously. RESULTS We found no relationship between chlorpromazine-equivalent dose and phenotype in any sample (complete sample: p = 0.3, actionable-subset: p = 0.82, risperidone-only: p = 0.34). Only clozapine dose was weakly associated with CYP2D6 phenotype (p = 0.03). CONCLUSION Clinicians were thus not intuitively adapting dose to CYP2D6 activity in this sample, nor was CYP2D6 activity associated with prescribed dose. Although the previous studies could not be replicated, this study may provide support for existing and future pharmacogenetic research.
Collapse
Affiliation(s)
- Emma Y De Brabander
- Department of Psychiatry and Neuropsychology, Mental Health and Neuroscience Research Institute, Maastricht University Medical Centre, The Netherlands
| | - Thérèse van Amelsvoort
- Department of Psychiatry and Neuropsychology, Mental Health and Neuroscience Research Institute, Maastricht University Medical Centre, The Netherlands
| | - Roos van Westrhenen
- Department of Psychiatry, Parnassia Groep BV, The Netherlands
- Institute of Psychiatry, Psychology and Neurosciences, King’s College London, London, UK
- St. John’s National Academy of Health Sciences, Bangalore, India
| | | |
Collapse
|
2
|
Shilbayeh SAR, Adeen IS, Alhazmi AS, Ibrahim SF, Al Enazi FAR, Ghanem EH, Binduraihem AM. The Frequency of CYP2D6 and CYP3A4/5 Genotypes and The Impact of Their Allele Translation and Phenoconversion-Predicted Enzyme Activity on Risperidone Pharmacokinetics in Saudi Children with Autism. Biochem Genet 2024; 62:2907-2932. [PMID: 38041757 DOI: 10.1007/s10528-023-10580-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 10/28/2023] [Indexed: 12/03/2023]
Abstract
Data on the role of CYP2D6 and CYP3A4/5 polymorphisms in relation to risperidone (RIS) pharmacokinetics (PK) in children are relatively limited and inconsistent. This is partially attributable to the limited coverage of CYP2D6 and CYP3A4/5 metabolizer phenotypes, particularly those of poor and ultrarapid metabolizers (PMs and UMs), which has led to calls for studies of populations with a non-European background that may carry variants that are less frequent in Europeans. Children ≤ 18 years old with at least 8 weeks of a RIS-based regimen were recruited from three autism centers in Riyadh, Saudi Arabia. The primary outcomes measured were plasma concentrations of RIS and 9-hydroxyrisperidone (9-OH-RIS) and their dose-adjusted (C/D) ratios as a function of phenotypes and activity score (AS). For accurate DNA genotyping, targeted pharmacogenomic testing with the Axiom PharmacoFocus Array was performed via examination of a broad collection of probesets targeting CYP2D6 and CYP3A4/5 variants. The frequency of genotypes/phenotypes and the impact of their allele translation and phenoconversion-predicted enzyme activity were examined. The final cohort included 83 individuals. The most common CYP2D6 phenotype in our population was normal metabolizers (NMs, 66.3%). Inconsistent with some previous studies, the three phenotypes of intermediate metabolizers (IMs), NMs, and UMs were significantly different in terms of RIS concentration, the RIS/9-OH-RIS ratio, the RIS C/D ratio and the 9-OH-RIS C/D ratio. According to AS analyses, there were statistically significant differences in the RIS concentration (P = 0.013), RIS/9-OH-RIS ratio (P < 0.001) and RIS C/D ratio (P = 0.030) when patients were categorized into AS ≤ 1 vs. AS > 1. None of the CYP3A4/5 star allele translated phenotypes revealed a significant influence on any of the RIS PK parameters. Notably, neither CYP2D6 nor CYP3A4/5 phenotyping demonstrated a significant impact on the total active moiety, suggesting that other gene variants could modulate RIS PK. The study confirmed the previously reported partial impact of the CYP2D6 gene on RIS PK. However, future studies using contemporary genotyping techniques targeting a wide range of variants in other candidate genes must be conducted to further examine their interactive effects on RIS PK and the clinical response.
Collapse
Affiliation(s)
- Sireen Abdul Rahim Shilbayeh
- Department of Pharmacy Practice, College of Pharmacy, Princess Nourah bint Abdulrahman University (PNU), P.O. Box 84428, 11671, Riyadh, Saudi Arabia.
| | - Iman Sharaf Adeen
- Department of Pediatric Behavior and Development and Adolescent Medicine, King Fahad Medical City, Riyadh, Saudi Arabia
| | - Ayman Shawqi Alhazmi
- Department of Pediatric Behavior and Development and Adolescent Medicine, King Saud Medical City, Riyadh, Saudi Arabia
| | - Samah Fathy Ibrahim
- College of Medicine, Princess Nourah bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Fawwaz Abdul Razaq Al Enazi
- Department of Pediatric Behavior and Development and Adolescent Medicine, King Fahad Medical City, Riyadh, Saudi Arabia
| | - Ezzeldeen Hasan Ghanem
- Pharmaceutical Analysis Section, King Abdullah International Medical Research Center (KAIMRC), King Abdulaziz Medical City, Ministry of National Guard - Health Affairs, Riyadh, Saudi Arabia
| | - Adel Mohammed Binduraihem
- Health Sciences Research Center, King Abdullah Bin Abdulaziz University Hospital, Princess Nourah bint Abdulrahman University, Riyadh, Saudi Arabia
| |
Collapse
|
3
|
Korošec Hudnik L, Blagus T, Redenšek Trampuž S, Dolžan V, Bon J, Pjevac M. Case report: Avoiding intolerance to antipsychotics through a personalized treatment approach based on pharmacogenetics. Front Psychiatry 2024; 15:1363051. [PMID: 38566958 PMCID: PMC10985247 DOI: 10.3389/fpsyt.2024.1363051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Accepted: 02/05/2024] [Indexed: 04/04/2024] Open
Abstract
Introduction The standard approach to treatment in psychiatry is known as "treatment as usual" (TAU), in which the same types of treatment are administered to a group of patients. TAU often requires numerous dose adjustments and medication changes due to ineffectiveness and/or the occurrence of adverse drug reactions (ADRs). This process is not only time-consuming but also costly. Antipsychotic medications are commonly used to treat various psychiatric disorders such as schizophrenia and mood disorders. Some of the inter-individual differences in efficacy and ADRs observed in psychopharmacotherapy can be explained by genetic variability in the pharmacokinetics and pharmacodynamics of antipsychotics. A better understanding of (in)efficacy and possible ADRs can be achieved by pharmacogenetic analysis of genes involved in the metabolism of antipsychotics. Most psychotropic drugs are metabolized by genetically variable CYP2D6, CYP1A2, CYP3A4, and CYP2C19 enzymes. To demonstrate the utility of pharmacogenetic testing for tailoring antipsychotic treatment, in this paper, we present the case of a patient in whom a pharmacogenetic approach remarkably altered an otherwise intolerant or ineffective conventional TAU with antipsychotics. Methods In this case report, we present a 60-year-old patient with psychotic symptoms who suffered from severe extrapyramidal symptoms and a malignant neuroleptic syndrome during treatment with risperidone, fluphenazine, aripiprazole, brexpiprazole, and olanzapine. Therefore, we performed a pharmacogenetic analysis by genotyping common functional variants in genes involved in the pharmacokinetic pathways of prescribed antipsychotics, namely, CYP2D6, CYP3A4, CYP3A5, CYP1A2, ABCB1, and ABCG2. Treatment recommendations for drug-gene pairs were made according to available evidence-based pharmacogenetic recommendations from the Dutch Pharmacogenetics Working Group (DPWG) or Clinical Pharmacogenetics Implementation Consortium (CPIC). Results Pharmacogenetic testing revealed a specific metabolic profile and pharmacokinetic phenotype of the patient, which in retrospect provided possible explanations for the observed ADRs. Based on the pharmacogenetic results, the choice of an effective and safe medication proved to be much easier. The psychotic symptoms disappeared after treatment, while the negative symptoms persisted to a lesser extent. Conclusion With the case presented, we have shown that taking into account the pharmacogenetic characteristics of the patient can explain the response to antipsychotic treatment and associated side effects. In addition, pharmacogenetic testing enabled an informed choice of the most appropriate drug and optimal dose adjustment. This approach makes it possible to avoid or minimize potentially serious dose-related ADRs and treatment ineffectiveness. However, due to the complexity of psychopathology and the polypharmacy used in this field, it is of great importance to conduct further pharmacokinetic and pharmacogenetic studies to better assess gene-drug and gene-gene-drug interactions.
Collapse
Affiliation(s)
- Liam Korošec Hudnik
- Centre for Clinical Psychiatry, University Psychiatric Clinic Ljubljana, Ljubljana, Slovenia
| | - Tanja Blagus
- Pharmacogenetics Laboratory, Institute of Biochemistry and Molecular Genetics, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Sara Redenšek Trampuž
- Pharmacogenetics Laboratory, Institute of Biochemistry and Molecular Genetics, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Vita Dolžan
- Pharmacogenetics Laboratory, Institute of Biochemistry and Molecular Genetics, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Jurij Bon
- Centre for Clinical Psychiatry, University Psychiatric Clinic Ljubljana, Ljubljana, Slovenia
- Department of Psychiatry, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Milica Pjevac
- Centre for Clinical Psychiatry, University Psychiatric Clinic Ljubljana, Ljubljana, Slovenia
| |
Collapse
|
4
|
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
|
5
|
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
|
6
|
Zhang Z, Yang Y, Kong W, Huang S, Tan Y, Huang S, Zhang M, Lu H, Li Y, Li X, Liu S, Wen Y, Shang D. A Bibliometric and Visual Analysis of Single Nucleotide Polymorphism Studies in Depression. Curr Neuropharmacol 2024; 22:302-322. [PMID: 37581520 PMCID: PMC10788886 DOI: 10.2174/1570159x21666230815125430] [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: 10/23/2022] [Revised: 11/30/2022] [Accepted: 02/10/2023] [Indexed: 08/16/2023] Open
Abstract
BACKGROUND Genetic polymorphism has been proven to have an important association with depression, which can influence the risk of developing depression, the efficacy of medications, and adverse effects via metabolic and neurological pathways. Nonetheless, aspects of the association between single nucleotide polymorphisms and depression have not been systematically investigated by bibliometric analysis. OBJECTIVE The aim of this study was to analyze the current status and trends of single nucleotide polymorphism research on depression through bibliometric and visual analysis. METHODS The Web of Science Core Collection was used to retrieve 10,043 articles that were published between 1998 and 2021. CiteSpace (6.1 R4) was used to perform collaborative network analysis, co-citation analysis, co-occurrence analysis, and citation burst detection. RESULTS The most productive and co-cited journals were the Journal of Affective Disorders and Biological Psychiatry, respectively, and an analysis of the references showed that the most recent research focused on the largest thematic cluster, "5-HT", reflecting the important research base in this area. "CYP2D6" has been in the spotlight since its emergence in 2009 and has become a research hotspot since its outbreak in 2019. However, "BDNF ", "COMT ", "older adults", "loci", and "DNA methylation" are also the new frontier of research, and some of them are currently in the process of exploration. CONCLUSION These findings offer a useful perspective on existing research and potential future approaches in the study of the association between single nucleotide polymorphisms and depression, which may assist researchers in selecting appropriate collaborators or journals.
Collapse
Affiliation(s)
- Zi Zhang
- Department of Pharmacy, The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou 510370, China
| | - Ye Yang
- Department of Pharmacy, The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou 510370, China
| | - Wan Kong
- Department of Pharmacy, The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou 510370, China
| | - Shanqing Huang
- Department of Pharmacy, The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou 510370, China
| | - Yaqian Tan
- Department of Pharmacy, The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou 510370, China
| | - Shanshan Huang
- Department of Pharmacy, The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou 510370, China
| | - Ming Zhang
- Department of Pharmacy, The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou 510370, China
| | - Haoyang Lu
- Department of Pharmacy, The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou 510370, China
| | - Yuhua Li
- Department of Pharmacy, The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou 510370, China
| | - Xiaolin Li
- Department of Pharmacy, The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou 510370, China
| | - Shujing Liu
- Department of Pharmacy, The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou 510370, China
| | - Yuguan Wen
- Department of Pharmacy, The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou 510370, China
| | - Dewei Shang
- Department of Pharmacy, The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou 510370, China
| |
Collapse
|
7
|
Kang Z, Qin Y, Sun Y, Lu Z, Sun Y, Chen H, Feng X, Zhang Y, Guo H, Yan H, Yue W. Multigenetic Pharmacogenomics-Guided Treatment vs Treatment As Usual Among Hospitalized Men With Schizophrenia: A Randomized Clinical Trial. JAMA Netw Open 2023; 6:e2335518. [PMID: 37801319 PMCID: PMC10559185 DOI: 10.1001/jamanetworkopen.2023.35518] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Accepted: 08/19/2023] [Indexed: 10/07/2023] Open
Abstract
Importance Limited evidence supports multigenetic pharmacogenomics-guided treatment (MPGT) in schizophrenia. Objective To evaluate the clinical effectiveness of MPGT in schizophrenia in a randomized clinical trial (RCT). Design, Setting, and Participants This RCT was conducted from March 2020 to March 2022. Male Chinese Han inpatients aged 18 to 60 years diagnosed with schizophrenia with a Positive and Negative Symptom Scale (PANSS) score of 60 or more from 2 selected study hospitals were included. Patients and raters were masked to MPGT or treatment as usual (TAU) randomization. Interventions Participants were randomly assigned in a 1:1 ratio to receive either MPGT or TAU for 12 weeks. Main Outcomes and Measures The primary efficacy outcome was the percentage change in PANSS total scores (range, 30 to 210) from baseline to week 6 analyzed by a modified intention-to-treat mixed model for repeated measures. The secondary outcome included response and symptomatic remission rates. Results A total of 210 participants (mean [SD] age, 29.2 [8.8] years) were enrolled and analyzed, with 113 assigned to MPGT and 97 to TAU. Compared with those randomized to TAU, participants randomized to MPGT demonstrated a significantly higher percentage change in PANSS score (74.2% vs 64.9%; adjusted mean difference, 9.2 percentage points; 95% CI, 4.4-14.1 percentage points; P < .001) and a higher response rate (93 of 113 [82.3%] vs 63 of 97 [64.9%]; adjusted odds ratio, 2.48; 95% CI, 1.28-4.80; P = .01) at the end of week 6. Conclusions and Relevance In this RCT of MPGT, MPGT was more effective than TAU in treating patients with schizophrenia. These findings suggest that multigenetic pharmacogenomic testing could serve as an effective tool to guide the treatment of schizophrenia. Trial Registration Chinese Clinical Trial Registry Identifier: ChiCTR2000029671.
Collapse
Affiliation(s)
- Zhewei Kang
- Institute of Mental Health, Peking University Sixth Hospital, Beijing, China
- National Clinical Research Center for Mental Disorders, Peking University Sixth Hospital, Beijing, China
- NHC Key Laboratory of Mental Health, Research Unit of Diagnosis and Treatment of Mood Cognitive Disorder, Chinese Academy of Medical Sciences, Beijing, China
| | - Ying Qin
- The Second People’s Hospital of Guizhou Province, Guiyang, China
| | - Yutao Sun
- Tangshan Mental Health Center, Tangshan Fifth Hospital, Tangshan, China
| | - Zhe Lu
- Institute of Mental Health, Peking University Sixth Hospital, Beijing, China
- National Clinical Research Center for Mental Disorders, Peking University Sixth Hospital, Beijing, China
- NHC Key Laboratory of Mental Health, Research Unit of Diagnosis and Treatment of Mood Cognitive Disorder, Chinese Academy of Medical Sciences, Beijing, China
| | - Yaoyao Sun
- Institute of Mental Health, Peking University Sixth Hospital, Beijing, China
- National Clinical Research Center for Mental Disorders, Peking University Sixth Hospital, Beijing, China
- NHC Key Laboratory of Mental Health, Research Unit of Diagnosis and Treatment of Mood Cognitive Disorder, Chinese Academy of Medical Sciences, Beijing, China
| | - Huan Chen
- Shantou University Mental Health Center, Shantou, China
| | - Xiaoyang Feng
- Institute of Mental Health, Peking University Sixth Hospital, Beijing, China
- National Clinical Research Center for Mental Disorders, Peking University Sixth Hospital, Beijing, China
- NHC Key Laboratory of Mental Health, Research Unit of Diagnosis and Treatment of Mood Cognitive Disorder, Chinese Academy of Medical Sciences, Beijing, China
| | - Yuyanan Zhang
- Institute of Mental Health, Peking University Sixth Hospital, Beijing, China
- National Clinical Research Center for Mental Disorders, Peking University Sixth Hospital, Beijing, China
- NHC Key Laboratory of Mental Health, Research Unit of Diagnosis and Treatment of Mood Cognitive Disorder, Chinese Academy of Medical Sciences, Beijing, China
| | - Hua Guo
- Zhumadian Second People’s Hospital, Zhumadian, China
| | - Hao Yan
- Institute of Mental Health, Peking University Sixth Hospital, Beijing, China
- National Clinical Research Center for Mental Disorders, Peking University Sixth Hospital, Beijing, China
- NHC Key Laboratory of Mental Health, Research Unit of Diagnosis and Treatment of Mood Cognitive Disorder, Chinese Academy of Medical Sciences, Beijing, China
| | - Weihua Yue
- Institute of Mental Health, Peking University Sixth Hospital, Beijing, China
- National Clinical Research Center for Mental Disorders, Peking University Sixth Hospital, Beijing, China
- NHC Key Laboratory of Mental Health, Research Unit of Diagnosis and Treatment of Mood Cognitive Disorder, Chinese Academy of Medical Sciences, Beijing, China
- PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, China
- Chinese Institute for Brain Research, Beijing, China
| |
Collapse
|
8
|
van der Wouden CH, Guchelaar HJ, Swen JJ. Precision Medicine Using Pharmacogenomic Panel-Testing: Current Status and Future Perspectives. Clin Lab Med 2022; 42:587-602. [PMID: 36368784 DOI: 10.1016/j.cll.2022.09.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Cathelijne H van der Wouden
- Department of Clinical Pharmacy & Toxicology, Leiden University Medical Center, Albinusdreef 2, Leiden 2333ZA, The Netherlands; Leiden Network for Personalised Therapeutics, Leiden, The Netherlands
| | - Henk-Jan Guchelaar
- Department of Clinical Pharmacy & Toxicology, Leiden University Medical Center, Albinusdreef 2, Leiden 2333ZA, The Netherlands; Leiden Network for Personalised Therapeutics, Leiden, The Netherlands
| | - Jesse J Swen
- Department of Clinical Pharmacy & Toxicology, Leiden University Medical Center, Albinusdreef 2, Leiden 2333ZA, The Netherlands; Leiden Network for Personalised Therapeutics, Leiden, The Netherlands.
| |
Collapse
|
9
|
González-Rodríguez A, Monreal JA, Natividad M, Seeman MV. Collaboration between Psychiatrists and Other Allied Medical Specialists for the Treatment of Delusional Disorders. Healthcare (Basel) 2022; 10:healthcare10091729. [PMID: 36141341 PMCID: PMC9498439 DOI: 10.3390/healthcare10091729] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 09/06/2022] [Accepted: 09/07/2022] [Indexed: 11/16/2022] Open
Abstract
Background: There is increasing evidence that individuals with psychosis are at increased risk for cardiovascular disease, diabetes, metabolic syndrome, and several other medical comorbidities. In delusional disorder (DD), this is particularly so because of the relatively late onset age. Aims: The aim of this narrative review is to synthesize the literature on the necessity for medical collaboration between psychiatrists and other specialists. Methods: A non-systematic narrative review was carried out of papers addressing referrals and cooperation among specialists in the care of DD patients. Results: Psychiatrists, the primary care providers for DD patients, depend on neurology to assess cognitive defects and rule out organic sources of delusions. Neurologists rely on psychiatry to help with patient adherence to treatment and the management of psychotropic drug side effects. Psychiatrists require ophthalmology/otolaryngology to treat sensory deficits that often precede delusions; reciprocally, psychiatric consults can help in instances of functional sensory impairment. Close collaboration with dermatologists is essential for treating delusional parasitosis and dysmorphophobia to ensure timely referrals to psychiatry. Conclusions: This review offers many other examples from the literature of the extent of overlap among medical specialties in the evaluation and effective treatment of DD. Optimal patient care requires close collaboration among specialties.
Collapse
Affiliation(s)
- Alexandre González-Rodríguez
- Department of Mental Health, Mutua Terrassa University Hospital, 5 Dr. Robert Square, 08221 Terrassa, Spain
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), University of Barcelona, 08221 Terrassa, Spain
| | - José Antonio Monreal
- Department of Mental Health, Mutua Terrassa University Hospital, 5 Dr. Robert Square, 08221 Terrassa, Spain
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), University of Barcelona, 08221 Terrassa, Spain
- Institut de Neurociències, Universitat Autònoma de Barcelona (UAB), 08221 Terrassa, Spain
- Correspondence:
| | - Mentxu Natividad
- Department of Mental Health, Mutua Terrassa University Hospital, 5 Dr. Robert Square, 08221 Terrassa, Spain
| | - Mary V. Seeman
- Department of Psychiatry, University of Toronto, 605 260 Heath Street West, Toronto, ON M5P 3L6, Canada
| |
Collapse
|
10
|
Islam F, Marshe VS, Magarbeh L, Frey BN, Milev RV, Soares CN, Parikh SV, Placenza F, Strother SC, Hassel S, Taylor VH, Leri F, Blier P, Uher R, Farzan F, Lam RW, Turecki G, Foster JA, Rotzinger S, Kennedy SH, Müller DJ. Effects of CYP2C19 and CYP2D6 gene variants on escitalopram and aripiprazole treatment outcome and serum levels: results from the CAN-BIND 1 study. Transl Psychiatry 2022; 12:366. [PMID: 36068210 PMCID: PMC9448818 DOI: 10.1038/s41398-022-02124-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 08/16/2022] [Accepted: 08/18/2022] [Indexed: 11/25/2022] Open
Abstract
Cytochrome P450 drug-metabolizing enzymes may contribute to interindividual differences in antidepressant outcomes. We investigated the effects of CYP2C19 and CYP2D6 gene variants on response, tolerability, and serum concentrations. Patients (N = 178) were treated with escitalopram (ESC) from weeks 0-8 (Phase I), and at week 8, either continued ESC if they were responders or were augmented with aripiprazole (ARI) if they were non-responders (<50% reduction in Montgomery-Åsberg Depression Rating Scale from baseline) for weeks 8-16 (Phase II). Our results showed that amongst patients on ESC-Only, CYP2C19 intermediate and poor metabolizers (IM + PMs), with reduced or null enzyme function, trended towards significantly lower symptom improvement during Phase II compared to normal metabolizers (NMs), which was not observed in ESC + ARI. We further showed that CYP2D6 NMs and IM + PMs had a higher likelihood of reporting a treatment-related central nervous system side effect in ESC-Only and ESC + ARI, respectively. The differences in the findings between ESC-Only and ESC + ARI may be due to the altered pharmacokinetics of ESC by ARI coadministration in ESC + ARI. We provided evidence for this postulation when we showed that in ESC-Only, CYP2C19 and CYP2D6 IM + PMs demonstrated significantly higher ESC concentrations at Weeks 10 and 16 compared to NMs. In contrast, ESC + ARI showed an association with CYP2C19 but not with CYP2D6 metabolizer group. Instead, ESC + ARI showed an association between CYP2D6 metabolizer group and ARI metabolite-to-drug ratio suggesting potential competition between ESC and ARI for CYP2D6. Our findings suggest that dosing based on CYP2C19 and CYP2D6 genotyping could improve safety and outcome in patients on ESC monotherapy.
Collapse
Affiliation(s)
- Farhana Islam
- grid.155956.b0000 0000 8793 5925Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON Canada ,grid.17063.330000 0001 2157 2938Department of Pharmacology & Toxicology, University of Toronto, Toronto, ON Canada
| | - Victoria S. Marshe
- grid.155956.b0000 0000 8793 5925Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON Canada ,grid.17063.330000 0001 2157 2938Institute of Medical Science, University of Toronto, Toronto, ON Canada
| | - Leen Magarbeh
- grid.155956.b0000 0000 8793 5925Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON Canada ,grid.17063.330000 0001 2157 2938Department of Pharmacology & Toxicology, University of Toronto, Toronto, ON Canada
| | - Benicio N. Frey
- grid.25073.330000 0004 1936 8227Department of Psychiatry and Behavioural Neurosciences, McMaster University, Hamilton, ON Canada ,grid.416721.70000 0001 0742 7355St. Joseph’s Healthcare Hamilton, Hamilton, ON Canada
| | - Roumen V. Milev
- grid.410356.50000 0004 1936 8331Department of Psychiatry, Queen’s University, Providence Care, Kingston, ON Canada
| | - Claudio N. Soares
- grid.410356.50000 0004 1936 8331Department of Psychiatry, Queen’s University, Providence Care, Kingston, ON Canada
| | - Sagar V. Parikh
- grid.214458.e0000000086837370Department of Psychiatry, University of Michigan, Ann Arbor, MI USA
| | - Franca Placenza
- grid.231844.80000 0004 0474 0428Centre for Mental Health, University Health Network, Toronto, ON Canada
| | - Stephen C. Strother
- grid.17063.330000 0001 2157 2938Rotman Research Institute, Baycrest Medical Centre, and Medical Biophysics, University of Toronto, Toronto, ON Canada
| | - Stefanie Hassel
- grid.22072.350000 0004 1936 7697Department of Psychiatry, University of Calgary, Calgary, AB Canada
| | - Valerie H. Taylor
- grid.22072.350000 0004 1936 7697Department of Psychiatry, University of Calgary, Calgary, AB Canada
| | - Francesco Leri
- grid.34429.380000 0004 1936 8198Department of Psychology and Neuroscience, University of Guelph, Guelph, ON Canada
| | - Pierre Blier
- grid.414622.70000 0001 1503 7525The Royal Institute of Mental Health Research, Ottawa, ON Canada
| | - Rudolf Uher
- grid.55602.340000 0004 1936 8200Department of Psychiatry, Dalhousie University, Halifax, NS Canada
| | - Faranak Farzan
- grid.61971.380000 0004 1936 7494Mechatronic Systems Engineering, Simon Fraser University, Surrey, BC Canada
| | - Raymond W. Lam
- grid.17091.3e0000 0001 2288 9830University of British Columbia and Vancouver Coastal Health Authority, Vancouver, BC Canada
| | - Gustavo Turecki
- grid.14709.3b0000 0004 1936 8649McGill Group for Suicide Studies, Douglas Mental Health University Institute, McGill University, Verdun, QC Canada
| | - Jane A. Foster
- grid.25073.330000 0004 1936 8227Department of Psychiatry and Behavioural Neurosciences, McMaster University, Hamilton, ON Canada ,grid.416721.70000 0001 0742 7355St. Joseph’s Healthcare Hamilton, Hamilton, ON Canada ,grid.231844.80000 0004 0474 0428Centre for Mental Health, University Health Network, Toronto, ON Canada
| | - Susan Rotzinger
- grid.17063.330000 0001 2157 2938Department of Psychiatry, St Michael’s Hospital, University of Toronto, Toronto, ON Canada ,grid.17063.330000 0001 2157 2938Department of Psychiatry, University of Toronto, Toronto, ON Canada
| | - Sidney H. Kennedy
- grid.17063.330000 0001 2157 2938Institute of Medical Science, University of Toronto, Toronto, ON Canada ,grid.231844.80000 0004 0474 0428Centre for Mental Health, University Health Network, Toronto, ON Canada ,grid.17063.330000 0001 2157 2938Department of Psychiatry, St Michael’s Hospital, University of Toronto, Toronto, ON Canada ,grid.17063.330000 0001 2157 2938Department of Psychiatry, University of Toronto, Toronto, ON Canada ,grid.415502.7Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St Michael’s Hospital, Toronto, ON Canada
| | - Daniel J. Müller
- grid.155956.b0000 0000 8793 5925Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON Canada ,grid.17063.330000 0001 2157 2938Department of Pharmacology & Toxicology, University of Toronto, Toronto, ON Canada ,grid.17063.330000 0001 2157 2938Institute of Medical Science, University of Toronto, Toronto, ON Canada ,grid.17063.330000 0001 2157 2938Department of Psychiatry, University of Toronto, Toronto, ON Canada ,grid.411760.50000 0001 1378 7891Department of Psychiatry, Psychosomatics and Psychotherapy, Center of Mental Health, University Clinic of Würzburg, Würzburg, Germany
| |
Collapse
|
11
|
Metabolizing status of CYP2C19 in response and side effects to medications for depression: Results from a naturalistic study. Eur Neuropsychopharmacol 2022; 56:100-111. [PMID: 35152032 DOI: 10.1016/j.euroneuro.2022.01.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 01/17/2022] [Accepted: 01/19/2022] [Indexed: 12/19/2022]
Abstract
Major depressive disorder (MDD) is one of the leading causes of disability worldwide. Polymorphisms in cytochrome P450 genes (CYP450) were demonstrated to play a significant role in antidepressant response and side effects, but their effect in real-world clinical practice is poorly known. We determined the metabolic status of CYP2C19 based on the combination of *1, *2, *3 and *17 alleles extracted from genome-wide data in 1239 patients with MDD, pharmacologically treated in a naturalistic setting. Symptom improvement and side effects were assessed using the Montgomery and Åsberg Depression Rating Scale and the Udvalg for Kliniske Undersøgelse scale, respectively. We tested if symptom improvement, response and side effects were associated with CYP2C19 metabolic status adjusting for potential confounders. We considered patients treated with drugs for depression having CYP2C19 genotyping recommended by guidelines (T1 Drugs); secondarily, with all psychotropic drugs having CYP2C19 as relevant metabolic path (T2 Drugs). In the group treated with T1 drugs (n = 540), poor metabolizers (PMs) showed higher response and higher symptom improvement compared to normal metabolizers (p = 0.023 and p = 0.009, respectively), but also higher risk of autonomic and neurological side effects (p = 0.022 and p = 0.022 respectively). In patients treated with T2 drugs (n = 801), similar results were found. No associations between metabolizer status and other types of side effects were found (psychic and other side effects). Our study suggests potential advantages of CYP2C19 pharmacogenetic testing to guide treatment prescription, that may not be limited to the drugs currently recommended by guidelines.
Collapse
|
12
|
Cacabelos R, Naidoo V, Corzo L, Cacabelos N, Carril JC. Genophenotypic Factors and Pharmacogenomics in Adverse Drug Reactions. Int J Mol Sci 2021; 22:ijms222413302. [PMID: 34948113 PMCID: PMC8704264 DOI: 10.3390/ijms222413302] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 12/05/2021] [Accepted: 12/06/2021] [Indexed: 02/06/2023] Open
Abstract
Adverse drug reactions (ADRs) rank as one of the top 10 leading causes of death and illness in developed countries. ADRs show differential features depending upon genotype, age, sex, race, pathology, drug category, route of administration, and drug–drug interactions. Pharmacogenomics (PGx) provides the physician effective clues for optimizing drug efficacy and safety in major problems of health such as cardiovascular disease and associated disorders, cancer and brain disorders. Important aspects to be considered are also the impact of immunopharmacogenomics in cutaneous ADRs as well as the influence of genomic factors associated with COVID-19 and vaccination strategies. Major limitations for the routine use of PGx procedures for ADRs prevention are the lack of education and training in physicians and pharmacists, poor characterization of drug-related PGx, unspecific biomarkers of drug efficacy and toxicity, cost-effectiveness, administrative problems in health organizations, and insufficient regulation for the generalized use of PGx in the clinical setting. The implementation of PGx requires: (i) education of physicians and all other parties involved in the use and benefits of PGx; (ii) prospective studies to demonstrate the benefits of PGx genotyping; (iii) standardization of PGx procedures and development of clinical guidelines; (iv) NGS and microarrays to cover genes with high PGx potential; and (v) new regulations for PGx-related drug development and PGx drug labelling.
Collapse
Affiliation(s)
- Ramón Cacabelos
- Department of Genomic Medicine, International Center of Neuroscience and Genomic Medicine, EuroEspes Biomedical Research Center, Bergondo, 15165 Corunna, Spain
- Correspondence: ; Tel.: +34-981-780-505
| | - Vinogran Naidoo
- Department of Neuroscience, International Center of Neuroscience and Genomic Medicine, EuroEspes Biomedical Research Center, Bergondo, 15165 Corunna, Spain;
| | - Lola Corzo
- Department of Medical Biochemistry, International Center of Neuroscience and Genomic Medicine, EuroEspes Biomedical Research Center, Bergondo, 15165 Corunna, Spain;
| | - Natalia Cacabelos
- Department of Medical Documentation, International Center of Neuroscience and Genomic Medicine, EuroEspes Biomedical Research Center, Bergondo, 15165 Corunna, Spain;
| | - Juan C. Carril
- Departments of Genomics and Pharmacogenomics, International Center of Neuroscience and Genomic Medicine, EuroEspes Biomedical Research Center, Bergondo, 15165 Corunna, Spain;
| |
Collapse
|
13
|
Lisoway AJ, Chen CC, Zai CC, Tiwari AK, Kennedy JL. Toward personalized medicine in schizophrenia: Genetics and epigenetics of antipsychotic treatment. Schizophr Res 2021; 232:112-124. [PMID: 34049235 DOI: 10.1016/j.schres.2021.05.010] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 04/30/2021] [Accepted: 05/02/2021] [Indexed: 12/21/2022]
Abstract
Schizophrenia is a complex psychiatric disorder where genetic, epigenetic, and environmental factors play a role in disease onset, course of illness, and treatment outcome. Pharmaco(epi)genetic research presents an important opportunity to improve patient care through prediction of medication side effects and response. In this narrative review, we discuss the current state of research and important progress of both genetic and epigenetic factors involved in antipsychotic response, over the past five years. The review is largely focused on the following frequently prescribed antipsychotics: olanzapine, risperidone, aripiprazole, and clozapine. Several consistent pharmacogenetic findings have emerged, in particular pharmacokinetic genes (primarily cytochrome P450 enzymes) and pharmacodynamic genes involving dopamine, serotonin, and glutamate neurotransmission. In addition to studies analysing DNA sequence variants, there are also several pharmacoepigenetic studies of antipsychotic response that have focused on the measurement of DNA methylation. Although pharmacoepigenetics is still in its infancy, consideration of both genetic and epigenetic factors contributing to antipsychotic response and side effects no doubt will be increasingly important in personalized medicine. We provide recommendations for next steps in research and clinical evaluation.
Collapse
Affiliation(s)
- Amanda J Lisoway
- Molecular Brain Science Department, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON, Canada; Institute of Medical Science, University of Toronto, Canada
| | - Cheng C Chen
- Molecular Brain Science Department, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON, Canada; Institute of Medical Science, University of Toronto, Canada
| | - Clement C Zai
- Molecular Brain Science Department, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON, Canada; Institute of Medical Science, University of Toronto, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, Canada; Department of Psychiatry, University of Toronto, Canada
| | - Arun K Tiwari
- Molecular Brain Science Department, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON, Canada; Department of Psychiatry, University of Toronto, Canada
| | - James L Kennedy
- Molecular Brain Science Department, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON, Canada; Institute of Medical Science, University of Toronto, Canada; Department of Psychiatry, University of Toronto, Canada.
| |
Collapse
|
14
|
Keeling NJ, Dunn TJ, Bentley JP, Ramachandran S, Hoffman JM, Rosenthal M. Approaches to assessing the provider experience with clinical pharmacogenomic information: a scoping review. Genet Med 2021; 23:1589-1603. [PMID: 33927377 DOI: 10.1038/s41436-021-01186-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Accepted: 04/11/2021] [Indexed: 11/09/2022] Open
Abstract
PURPOSE Barriers to the implementation of pharmacogenomics in clinical practice have been thoroughly discussed over the past decade. METHODS The objective of this scoping review was to characterize the peer-reviewed literature surrounding the experiences and actions of prescribers, pharmacists, or genetic counselors when using pharmacogenomic information in real-world or hypothetical research settings. RESULTS A total of 33 studies were included in the scoping review. The majority of studies were conducted in the United States (70%), used quantitative or mixed methods (79%) with physician or pharmacist respondents (100%). The qualitative content analysis revealed five major methodological approaches: hypothetical clinical case scenarios, real-world studies evaluating prescriber response to recommendations or alerts, cross-sectional quantitative surveys, cross-sectional qualitative surveys/interviews, and a quasi-experimental real-world study. CONCLUSION The findings of this scoping review can guide further research on the factors needed to successfully integrate pharmacogenomics into clinical care.
Collapse
Affiliation(s)
- Nicholas J Keeling
- Department of Pharmacy Administration, University of Mississippi School of Pharmacy, University, MS, USA
| | - Tyler J Dunn
- Department of Pharmacy Administration, University of Mississippi School of Pharmacy, University, MS, USA.
| | - John P Bentley
- Department of Pharmacy Administration, University of Mississippi School of Pharmacy, University, MS, USA
| | - Sujith Ramachandran
- Department of Pharmacy Administration, University of Mississippi School of Pharmacy, University, MS, USA
| | - James M Hoffman
- Department of Pharmaceutical Sciences and Office of Quality and Patient Care, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Meagen Rosenthal
- Department of Pharmacy Administration, University of Mississippi School of Pharmacy, University, MS, USA
| |
Collapse
|
15
|
Islam F, Gorbovskaya I, Müller DJ. Pharmacogenetic/Pharmacogenomic Tests for Treatment Prediction in Depression. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1305:231-255. [PMID: 33834403 DOI: 10.1007/978-981-33-6044-0_13] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Genetic factors play a significant but complex role in antidepressant (AD) response and tolerability. During recent years, there is growing enthusiasm in the promise of pharmacogenetic/pharmacogenomic (PGx) tools for optimizing and personalizing treatment outcomes for patients with major depressive disorder (MDD). The influence of pharmacokinetic and pharmacodynamic genes on response and tolerability has been investigated, including those encoding the cytochrome P450 superfamily, P-glycoprotein, monoaminergic transporters and receptors, intracellular signal transduction pathways, and the stress hormone system. Genome-wide association studies are also identifying new genetic variants associated with AD response phenotypes, which, combined with methods such as polygenic risk scores (PRS), is opening up new avenues for novel personalized treatment approaches for MDD. This chapter describes the basic concepts in PGx of AD response, reviews the major pharmacokinetic and pharmacodynamic genes involved in AD outcome, discusses PRS as a promising approach for predicting AD efficacy and tolerability, and addresses key challenges to the development and application of PGx tests.
Collapse
Affiliation(s)
- Farhana Islam
- Pharmacogenetics Research Clinic, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Canada
- Department of Pharmacology & Toxicology, University of Toronto, Toronto, ON, Canada
| | - Ilona Gorbovskaya
- Pharmacogenetics Research Clinic, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Canada
| | - Daniel J Müller
- Pharmacogenetics Research Clinic, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Canada.
- Department of Pharmacology & Toxicology, University of Toronto, Toronto, ON, Canada.
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
16
|
Baldelli S, Cheli S, Montrasio C, Cattaneo D, Clementi E. Therapeutic drug monitoring and pharmacogenetics of antipsychotics and antidepressants in real life settings: A 5-year single centre experience. World J Biol Psychiatry 2021; 22:34-45. [PMID: 32212950 DOI: 10.1080/15622975.2020.1747112] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
OBJECTIVES Exposure and clinical response to CNS drugs are largely variable. AGNP guidelines suggest therapy individualisation with therapeutic drug monitoring of plasma concentrations and pharmacogenetic testing. We present the retrospective analysis of the last 5 years' data collected in real life settings as indirect evidence of the applications of the AGNP guidelines in the routine clinical management of psychiatric patients requiring pharmacologic treatments. METHODS Plasma concentrations were quantified using a liquid chromatography/tandem mass spectrometry method. Genomic DNA was isolated using an automatic DNA extraction system. All genotypes were determined by Real-Time PCR. RESULTS We collected a total of 4582 requests for TDM and 212 requests for pharmacogenetic analysis. A wide distribution in the trough concentrations was observed for most drugs indicating a high interpatient variability. Nearly 45% of the samples had trough levels below the minimum effective drug concentrations set by the AGNP guidelines; only 8% of the samples had high concentrations. For pharmacogenetics analysis, among antipsychotics, clozapine, haloperidol and aripiprazole were the most requested (78%); while for antidepressants SSRIs were the most frequently prescribed. CONCLUSIONS These data suggest that physicians are becoming more confident with the laboratory pharmacologic tools to optimise treatments and/or that the pharmacological treatment of patients with psychiatric disorders is becoming more challenging. TDM and PGx might significantly contribute to the rational selection of the best drug and best dose in individual cases.
Collapse
Affiliation(s)
- Sara Baldelli
- Unit of Clinical Pharmacology, ASST Fatebenefratelli Sacco University Hospital, Milan, Italy
| | - Stefania Cheli
- Unit of Clinical Pharmacology, ASST Fatebenefratelli Sacco University Hospital, Milan, Italy
| | - Cristina Montrasio
- Unit of Clinical Pharmacology, ASST Fatebenefratelli Sacco University Hospital, Milan, Italy
| | - Dario Cattaneo
- Unit of Clinical Pharmacology, ASST Fatebenefratelli Sacco University Hospital, Milan, Italy
| | - Emilio Clementi
- Clinical Pharmacology Unit, Consiglio Nazionale delle Ricerche Institute of Neuroscience, Department of Biomedical and Clinical Sciences L, Sacco University Hospital, Università degli Studi di Milano, Milan, Italy.,Eugenio Medea Scientific Institute, Bosisio Parini, Italy
| |
Collapse
|
17
|
Arranz MJ, Salazar J, Hernández MH. Pharmacogenetics of antipsychotics: Clinical utility and implementation. Behav Brain Res 2020; 401:113058. [PMID: 33316324 DOI: 10.1016/j.bbr.2020.113058] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 11/23/2020] [Accepted: 12/04/2020] [Indexed: 02/06/2023]
Abstract
Decades of research have produced extensive evidence of the contribution of genetic factors to the efficacy and toxicity of antipsychotics. Numerous genetic variants in genes controlling drug availability or involved in antipsychotic processes have been linked to treatment variability. The complex mechanism of action and multitarget profile of most antipsychotic drugs hinder the identification of pharmacogenetic markers of clinical value. Nevertheless, the validity of associations between variants in CYP1A2, CYP2D6, CYP2C19, ABCB1, DRD2, DRD3, HTR2A, HTR2C, BDNF, COMT, MC4R genes and antipsychotic response has been confirmed in independent candidate gene studies. Genome wide pharmacogenomic studies have proven the role of the glutamatergic pathway in mediating antipsychotic activity and have reported novel associations with antipsychotic response. However, only a limited number of the findings, mainly functional variants of CYP metabolic enzymes, have been shown to be of clinical utility and translated into useful pharmacogenetic markers. Based on the currently available information, actionable pharmacogenetics should be reduced to antipsychotics' dose adjustment according to the genetically predicted metabolic status (CYPs' profile) of the patient. Growing evidence suggests that such interventions will reduce antipsychotics' side-effects and increase treatment safety. Despite this evidence, the use of pharmacogenetics in psychiatric wards is minimal. Hopefully, further evidence on the clinical and economic benefits, the development of clinical protocols based on pharmacogenetic information, and improved and cheaper genetic testing will increase the implementation of pharmacogenetic guided prescription in clinical settings.
Collapse
Affiliation(s)
- Maria J Arranz
- Fundació Docència i Recerca Mútua Terrassa, Spain; Centro de investigación en Red de Salud Mental, CIBERSAM, Madrid, Spain; PHAGEX Research Group, Universitat Ramon LLull, Spain.
| | - Juliana Salazar
- Translational Medical Oncology Laboratory, Institut d'Investigació Biomèdica Sant Pau (IIB-Sant Pau), Barcelona, Spain; U705, ISCIII Center for Biomedical Research on Rare Diseases (CIBERER), Barcelona, Spain; PHAGEX Research Group, Universitat Ramon LLull, Spain
| | - Marta H Hernández
- PHAGEX Research Group, Universitat Ramon LLull, Spain; School of Health Sciences Blanquerna. University Ramon Llull, Barcelona, Spain
| |
Collapse
|
18
|
Bousman CA, Bengesser SA, Aitchison KJ, Amare AT, Aschauer H, Baune BT, Asl BB, Bishop JR, Burmeister M, Chaumette B, Chen LS, Cordner ZA, Deckert J, Degenhardt F, DeLisi LE, Folkersen L, Kennedy JL, Klein TE, McClay JL, McMahon FJ, Musil R, Saccone NL, Sangkuhl K, Stowe RM, Tan EC, Tiwari AK, Zai CC, Zai G, Zhang J, Gaedigk A, Müller DJ. Review and Consensus on Pharmacogenomic Testing in
Psychiatry. PHARMACOPSYCHIATRY 2020; 54:5-17. [DOI: 10.1055/a-1288-1061] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
AbstractThe implementation of pharmacogenomic (PGx) testing in psychiatry remains modest,
in part due to divergent perceptions of the quality and completeness of the
evidence base and diverse perspectives on the clinical utility of PGx testing
among psychiatrists and other healthcare providers. Recognizing the current lack
of consensus within the field, the International Society of Psychiatric Genetics
assembled a group of experts to conduct a narrative synthesis of the PGx
literature, prescribing guidelines, and product labels related to psychotropic
medications as well as the key considerations and limitations related to the use
of PGx testing in psychiatry. The group concluded that to inform medication
selection and dosing of several commonly-used antidepressant and antipsychotic
medications, current published evidence, prescribing guidelines, and product
labels support the use of PGx testing for 2 cytochrome P450 genes (CYP2D6,
CYP2C19). In addition, the evidence supports testing for human leukocyte
antigen genes when using the mood stabilizers carbamazepine (HLA-A and
HLA-B), oxcarbazepine (HLA-B), and phenytoin (CYP2C9, HLA-B). For
valproate, screening for variants in certain genes (POLG, OTC, CSP1) is
recommended when a mitochondrial disorder or a urea cycle disorder is suspected.
Although barriers to implementing PGx testing remain to be fully resolved, the
current trajectory of discovery and innovation in the field suggests these
barriers will be overcome and testing will become an important tool in
psychiatry.
Collapse
Affiliation(s)
- Chad A. Bousman
- Departments of Medical Genetics, Psychiatry, Physiology &
Pharmacology, University of Calgary, Calgary, AB, Canada
- Hotchkiss Brain Institute, Cumming School of Medicine, University of
Calgary, Calgary, AB, Canada
- Alberta Children’s Hospital Research Institute, Calgary, AB,
Canada
- Department of Psychiatry, Melbourne Medical School, The University of
Melbourne, Melbourne, VIC, Australia
| | - Susanne A. Bengesser
- Department of Psychiatry and Psychotherapeutic Medicine, Medical
University of Graz, Austria
| | - Katherine J. Aitchison
- Departments of Psychiatry, Medical Genetics and the Neuroscience and
Mental Health Institute, University of Alberta, Edmonton, AB,
Canada
| | - Azmeraw T. Amare
- Discipline of Psychiatry, School of Medicine, University of Adelaide,
Adelaide, SA, Australia
- South Australian Health and Medical Research Institute (SAHMRI),
Adelaide, SA, Australia
| | - Harald Aschauer
- Biopsychosocial Corporation (BioPsyC), non-profit association, Vienna,
Austria
| | - Bernhard T. Baune
- Department of Psychiatry and Psychotherapy, University of
Münster, Germany
- Department of Psychiatry, Melbourne Medical School, The University of
Melbourne, Melbourne, VIC, Australia
- The Florey Institute of Neuroscience and Mental Health, The University
of Melbourne, Parkville, VIC, Australia
| | - Bahareh Behroozi Asl
- Departments of Psychiatry, Medical Genetics and the Neuroscience and
Mental Health Institute, University of Alberta, Edmonton, AB,
Canada
| | - Jeffrey R. Bishop
- Department of Experimental and Clinical Pharmacology, University of
Minnesota College of Pharmacy and Department of Psychiatry, University of
Minnesota Medical School, Minneapolis, MN, USA
| | - Margit Burmeister
- Michigan Neuroscience Institute and Departments of Computational
Medicine & Bioinformatics, Human Genetics and Psychiatry, The University
of Michigan, Ann Arbor MI, USA
| | - Boris Chaumette
- Institute of Psychiatry and Neuroscience of Paris, GHU Paris
Psychiatrie & Neurosciences, University of Paris, Paris,
France
- Department of Psychiatry, McGill University, Montreal,
Canada
| | - Li-Shiun Chen
- Departments of Psychiatry and Genetics, Washington University School of
Medicine in St. Louis, USA
| | - Zachary A. Cordner
- Department of Psychiatry & Behavioral Sciences, Johns Hopkins
University School of Medicine, Baltimore, MD, USA
| | - Jürgen Deckert
- Department of Psychiatry, Psychosomatics and Psychotherapy, Center of
Mental Health, Würzburg, Germany
| | - Franziska Degenhardt
- Institute of Human Genetics, University of Bonn, School of Medicine
& University Hospital Bonn, Bonn, Germany
- Department of Child and Adolescent Psychiatry, Psychosomatics and
Psychotherapy, University Hospital Essen, University of Duisburg-Essen,
Duisburg, Germany
| | - Lynn E. DeLisi
- Department of Psychiatry, Harvard Medical School, Cambridge Health
Alliance, Cambridge, Massachusetts, USA
| | - Lasse Folkersen
- Institute of Biological Psychiatry, Capital Region Hospitals,
Copenhagen, Denmark
| | - James L. Kennedy
- Department of Psychiatry, University of Toronto, Toronto, Ontario,
Canada
- Centre for Addiction and Mental Health, University of Toronto, Toronto,
Ontario, Canada
| | - Teri E. Klein
- Department of Biomedical Data Science, Stanford University, Stanford,
California, USA
| | - Joseph L. McClay
- Department of Pharmacotherapy and Outcome Science, Virginia
Commonwealth University School of Pharmacy, Richmond, VA, USA
| | - Francis J. McMahon
- Human Genetics Branch, National Institute of Mental Health, Bethesda,
MD, USA
| | - Richard Musil
- Department of Psychiatry and Psychotherapy,
Ludwig-Maximilians-University, Munich, Germany
| | - Nancy L. Saccone
- Departments of Psychiatry and Genetics, Washington University School of
Medicine in St. Louis, USA
| | - Katrin Sangkuhl
- Department of Biomedical Data Science, Stanford University, Stanford,
California, USA
| | - Robert M. Stowe
- Departments of Psychiatry and Neurology (Medicine), University of
British Columbia, USA
| | - Ene-Choo Tan
- KK Research Centre, KK Women’s and Children’s Hospital,
Singapore, Singapore
| | - Arun K. Tiwari
- Department of Psychiatry, University of Toronto, Toronto, Ontario,
Canada
- Centre for Addiction and Mental Health, University of Toronto, Toronto,
Ontario, Canada
| | - Clement C. Zai
- Department of Psychiatry, University of Toronto, Toronto, Ontario,
Canada
- Centre for Addiction and Mental Health, University of Toronto, Toronto,
Ontario, Canada
| | - Gwyneth Zai
- Department of Psychiatry, University of Toronto, Toronto, Ontario,
Canada
- Centre for Addiction and Mental Health, University of Toronto, Toronto,
Ontario, Canada
| | - Jianping Zhang
- Department of Psychiatry, Weill Cornell Medical College, New
York-Presbyterian Westchester Division, White Plains, NY, USA
| | - Andrea Gaedigk
- Division of Clinical Pharmacology, Toxicology & Therapeutic
Innovation, Children’s Mercy Kansas City, Kansas City and School of
Medicine, University of Missouri-Kansas City, Kansas City, MO, USA
| | - Daniel J Müller
- Department of Psychiatry, University of Toronto, Toronto, Ontario,
Canada
- Centre for Addiction and Mental Health, University of Toronto, Toronto,
Ontario, Canada
| |
Collapse
|
19
|
No association between CYP2C19 genetic polymorphism with treatment remission to antidepressant venlafaxine in Han Chinese population. Psychiatr Genet 2020; 30:30-33. [PMID: 31842058 DOI: 10.1097/ypg.0000000000000246] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
OBJECTIVE Major depressive disorder (MDD) is a global mental health problem. As a serotonin-noradrenaline reuptake inhibitor (SNRI), the antidepressant venlafaxine is used to alleviate MDD clinically. Recent research has shown that Cytochrome P450 (CYP) enzymes affect venlafaxine efficacy by mediating its metabolism. The present study investigates genetic polymorphisms of cytochrome P450 family 2 subfamily C member 19 (CYP2C19) are associated with remission after venlafaxine treatment for MDD. METHODS A total of 175 Han Chinese patients with depression were recruited to accept a 6-week treatment with venlafaxine. Three single-nucleotide polymorphisms of CYP2C19 were selected from dbSNP and previous literature to compare the allele and genotype frequencies between patients in remission and nonremission. Seventeen items Hamilton Depression Scale (17-HAMD) was used to access the outcomes of patients' depressive symptoms through the study. Our results denied the role of CYP2C19 polymorphisms for remission after venlafaxine treatment in MDD patients. RESULT & CONCLUSION CYP2C19 genetic polymorphism may not have association with SNRI venlafaxine treatment remission in the Han Chinese population.
Collapse
|
20
|
van der Wouden CH, Böhringer S, Cecchin E, Cheung KC, Dávila-Fajardo CL, Deneer VH, Dolžan V, Ingelman-Sundberg M, Jönsson S, Karlsson MO, Kriek M, Mitropoulou C, Patrinos GP, Pirmohamed M, Rial-Sebbag E, Samwald M, Schwab M, Steinberger D, Stingl J, Sunder-Plassmann G, Toffoli G, Turner RM, van Rhenen MH, van Zwet E, Swen JJ, Guchelaar HJ. Generating evidence for precision medicine: considerations made by the Ubiquitous Pharmacogenomics Consortium when designing and operationalizing the PREPARE study. Pharmacogenet Genomics 2020; 30:131-144. [PMID: 32317559 PMCID: PMC7331826 DOI: 10.1097/fpc.0000000000000405] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Accepted: 03/02/2020] [Indexed: 02/06/2023]
Abstract
OBJECTIVES Pharmacogenetic panel-based testing represents a new model for precision medicine. A sufficiently powered prospective study assessing the (cost-)effectiveness of a panel-based pharmacogenomics approach to guide pharmacotherapy is lacking. Therefore, the Ubiquitous Pharmacogenomics Consortium initiated the PREemptive Pharmacogenomic testing for prevention of Adverse drug Reactions (PREPARE) study. Here, we provide an overview of considerations made to mitigate multiple methodological challenges that emerged during the design. METHODS An evaluation of considerations made when designing the PREPARE study across six domains: study aims and design, primary endpoint definition and collection of adverse drug events, inclusion and exclusion criteria, target population, pharmacogenomics intervention strategy, and statistical analyses. RESULTS Challenges and respective solutions included: (1) defining and operationalizing a composite primary endpoint enabling measurement of the anticipated effect, by including only severe, causal, and drug genotype-associated adverse drug reactions; (2) avoiding overrepresentation of frequently prescribed drugs within the patient sample while maintaining external validity, by capping drugs of enrolment; (3) designing the pharmacogenomics intervention strategy to be applicable across ethnicities and healthcare settings; and (4) designing a statistical analysis plan to avoid dilution of effect by initially excluding patients without a gene-drug interaction in a gatekeeping analysis. CONCLUSION Our design considerations will enable quantification of the collective clinical utility of a panel of pharmacogenomics-markers within one trial as a proof-of-concept for pharmacogenomics-guided pharmacotherapy across multiple actionable gene-drug interactions. These considerations may prove useful to other investigators aiming to generate evidence for precision medicine.
Collapse
Affiliation(s)
- Cathelijne H. van der Wouden
- Department of Clinical Pharmacy and Toxicology, Leiden University Medical Center
- Leiden University Medical Center, Leiden Network for Personalised Therapeutics
| | - Stefan Böhringer
- Department of Biomedical Data Sciences, Leiden University Medical Center, Leiden, The Netherlands
| | - Erika Cecchin
- Experimental and Clinical Pharmacology, Experimental and Clinical Pharmacology; Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, National Cancer Institute, Aviano, Italy
| | - Ka-Chun Cheung
- Medicine Information Centre, Royal Dutch Pharmacists Association (KNMP), The Hague, The Netherlands
| | - Cristina Lucía Dávila-Fajardo
- Department of Clinical Pharmacy, San Cecilio University Hospital, Instituto de investigación biosanitaria de Granada, ibs.Granada, Granada, Spain
| | - Vera H.M. Deneer
- Department of Clinical Pharmacy, Division of Laboratories, Pharmacy, and Biomedical Genetics, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Vita Dolžan
- Pharmacogenetics Laboratory, Faculty of Medicine, Institute of Biochemistry, University of Ljubljana, Slovenia
| | - Magnus Ingelman-Sundberg
- Department of Physiology and Pharmacology, Section of Pharmacogenetics, Karolinska Institutet, Stockholm
| | - Siv Jönsson
- Department of Physiology and Pharmacology, Section of Pharmacogenetics, Karolinska Institutet, Stockholm
| | - Mats O. Karlsson
- Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden
| | - Marjolein Kriek
- Department of Clinical Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | | | - George P. Patrinos
- Department of Pharmacy, University of Patras, School of Health Sciences, University Campus, Rion, Patras, Greece
| | - Munir Pirmohamed
- Department of Molecular and Clinical Pharmacology, University of Liverpool, and Royal Liverpool University Hospital, Liverpool, UK
| | | | - Matthias Samwald
- Center for Medical Statistics, Informatics, and Intelligent Systems, Medical University of Vienna, Vienna, Austria
| | - Matthias Schwab
- Department of Clinical Pharmacology, Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany and University of Tübingen
- Department of Clinical Pharmacology, University Hospital Tübingen
- Department of Pharmacy and Biochemistry, University of Tübingen, Tübingen
| | - Daniela Steinberger
- bio.logis Center for Human Genetics, Frankfurt am Main
- Institute of Human Genetics, Justus Liebig University Giessen
| | - Julia Stingl
- Institute of Clinical Pharmacology, University Hospital of RWTH Aachen, Aachen, Germany
| | - Gere Sunder-Plassmann
- Department of Internal Medicine III, Division of Nephrology and Dialysis, Medical University of Venna, Vienna, Austria
| | - Giuseppe Toffoli
- Experimental and Clinical Pharmacology, Experimental and Clinical Pharmacology; Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, National Cancer Institute, Aviano, Italy
| | - Richard M. Turner
- Department of Molecular and Clinical Pharmacology, University of Liverpool, and Royal Liverpool University Hospital, Liverpool, UK
| | - Mandy H. van Rhenen
- Medicine Information Centre, Royal Dutch Pharmacists Association (KNMP), The Hague, The Netherlands
| | - Erik van Zwet
- Department of Biomedical Data Sciences, Leiden University Medical Center, Leiden, The Netherlands
| | - Jesse J. Swen
- Department of Clinical Pharmacy and Toxicology, Leiden University Medical Center
- Leiden University Medical Center, Leiden Network for Personalised Therapeutics
| | - Henk-Jan Guchelaar
- Department of Clinical Pharmacy and Toxicology, Leiden University Medical Center
- Leiden University Medical Center, Leiden Network for Personalised Therapeutics
| |
Collapse
|
21
|
Fu Y, Tian X, Han L, Li Y, Peng Y, Zheng J. Mechanism-based inactivation of cytochrome P450 2D6 by Notopterol. Chem Biol Interact 2020; 322:109053. [PMID: 32198085 DOI: 10.1016/j.cbi.2020.109053] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Revised: 01/28/2020] [Accepted: 03/09/2020] [Indexed: 11/17/2022]
Abstract
Notopterol (NOT) is a major bioactive ingredient extracted from the rhizomes of either Notopterygium incisum Ting ex H. T. Chang or N. forbesii Boiss (Qianghuo in Chinese), a botanical drug that was adopted as a traditional Chinese medicine. NOT is suggested to show analgesic and anti-inflammatory effects in clinical practice. The inhibitory effects of NOT on human cytochrome P450 enzymes were investigated in the present study. Our results indicate that NOT inhibited the activity of CYP2D6 in a time-, concentration- and NADPH-dependent manner. The values of KI and kinact were 10.8 μM and 0.62 min-1, respectively. The calculated kobs at 10 μM was 0.29 min-1, above the 0.02 min-1 risk level. After incubation with NOT at 10 μM for 9 min, approximately 92% of CYP2D6 activity was inhibited. Such loss of enzyme activity was not restored through dialysis, which indicates that the observed enzyme inhibition was irreversible. Partition ratio of the inactivation was approximately 29. Quinidine, a competitive CYP2D6 inhibitor, demonstrated protection on enzymes against the NOT-induced inactivation, but such protection was not found in incubation systems fortified with glutathione or catalase/superoxide dismutase. Additionally, CYP3A4 was observed to function as an enzyme mainly involved in the biotransformation of NOT. Taken together, these findings indicate that NOT served as a mechanism-based inactivator of CYP2D6, meanwhile, those observed effects may induce the latent drug-drug interactions. The metabolic activation of NOT may be the key to trigger the inactivation of the enzyme.
Collapse
Affiliation(s)
- Yao Fu
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning, 110016, PR China
| | - Xiaoxiao Tian
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning, 110016, PR China
| | - Lingling Han
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning, 110016, PR China
| | - Yilin Li
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning, 110016, PR China
| | - Ying Peng
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning, 110016, PR China.
| | - Jiang Zheng
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning, 110016, PR China; State Key Laboratory of Functions and Applications of Medicinal Plants, Key Laboratory of Pharmaceutics of Guizhou Province, Guizhou Medical University, Guiyang, Guizhou, 550025, PR China; Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, 550025, PR China.
| |
Collapse
|
22
|
The effect of CYP2D6 variation on antipsychotic-induced hyperprolactinaemia: a systematic review and meta-analysis. THE PHARMACOGENOMICS JOURNAL 2020; 20:629-637. [PMID: 32015455 DOI: 10.1038/s41397-019-0142-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Revised: 12/13/2019] [Accepted: 12/17/2019] [Indexed: 11/09/2022]
Abstract
Hyperprolactinemia is a known adverse drug reaction to antipsychotic treatment. Antipsychotic blood levels are influenced by cytochrome P450 enzymes, primarily CYP2D6. Variation in CYP450 genes may affect the risk of antipsychotic-induced hyperprolactinemia. We undertook a systematic review and meta-analysis to assess whether CYP2D6 functional genetic variants are associated with antipsychotic-induced hyperprolactinemia. The systematic review identified 16 relevant papers, seven of which were suitable for the meta-analysis (n = 303 participants including 134 extreme metabolisers). Participants were classified into four phenotype groups as poor, intermediate, extensive, and ultra-rapid metabolisers. A random effects meta-analysis was used and Cohen's d calculated as the effect size for each primary study. We found no significant differences in prolactin levels between CYP2D6 metabolic groups. Current evidence does not support using CYP2D6 genotyping to reduce risk of antipsychotic-induced hyperprolactinemia. However, statistical power is limited. Future studies with larger samples and including a range of prolactin-elevating drugs are needed.
Collapse
|
23
|
van der Wouden CH, van Rhenen MH, Jama WOM, Ingelman-Sundberg M, Lauschke VM, Konta L, Schwab M, Swen JJ, Guchelaar HJ. One non-believer: Response to "Obviously Nine Believers: Actionable Germline Genetic Variants for Pre-emptive Pharmacogenetic Testing". Basic Clin Pharmacol Toxicol 2020; 126:7-8. [PMID: 31670867 DOI: 10.1111/bcpt.13336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Accepted: 09/25/2019] [Indexed: 01/19/2023]
Affiliation(s)
- Cathelijne H van der Wouden
- Department of Clinical Pharmacy & Toxicology, Leiden University Medical Center, Leiden, The Netherlands.,Leiden Network for Personalised Therapeutics, Leiden, The Netherlands
| | | | - Wafa O M Jama
- Royal Dutch Pharmacists Association (KNMP), The Hague, The Netherlands
| | - Magnus Ingelman-Sundberg
- Department of Physiology and Pharmacology, Section of Pharmacogenetics, Biomedicum 5B Karolinska Institute, Stockholm, Sweden
| | - Volker M Lauschke
- Department of Physiology and Pharmacology, Section of Pharmacogenetics, Biomedicum 5B Karolinska Institute, Stockholm, Sweden
| | - Lidija Konta
- bio.logis Center for Human Genetics, Frankfurt am Main, Germany
| | - Matthias Schwab
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany.,Department of Clinical Pharmacology and Departments of Biochemistry and Pharmacy, University Tuebingen, Tuebingen, Germany
| | - Jesse J Swen
- Department of Clinical Pharmacy & Toxicology, Leiden University Medical Center, Leiden, The Netherlands.,Leiden Network for Personalised Therapeutics, Leiden, The Netherlands
| | - Henk-Jan Guchelaar
- Department of Clinical Pharmacy & Toxicology, Leiden University Medical Center, Leiden, The Netherlands.,Leiden Network for Personalised Therapeutics, Leiden, The Netherlands
| |
Collapse
|
24
|
Omics in schizophrenia: current progress and future directions of antipsychotic treatments. JOURNAL OF BIO-X RESEARCH 2019. [DOI: 10.1097/jbr.0000000000000049] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
|
25
|
Arranz MJ, Gonzalez-Rodriguez A, Perez-Blanco J, Penadés R, Gutierrez B, Ibañez L, Arias B, Brunet M, Cervilla J, Salazar J, Catalan R. A pharmacogenetic intervention for the improvement of the safety profile of antipsychotic treatments. Transl Psychiatry 2019; 9:177. [PMID: 31346157 PMCID: PMC6658489 DOI: 10.1038/s41398-019-0511-9] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 05/09/2019] [Accepted: 05/31/2019] [Indexed: 12/24/2022] Open
Abstract
Antipsychotic drugs fail to achieve adequate response in 30-50% of treated patients and about 50% of them develop severe and lasting side effects. Treatment failure results in poorer prognosis with devastating repercussions for the patients, carers and broader society. Our study evaluated the clinical benefits of a pharmacogenetic intervention for the personalisation of antipsychotic treatment. Pharmacogenetic information in key CYP polymorphisms was used to adjust clinical doses in a group of patients who started or switched treatment with antipsychotic drugs (PharmG+, N = 123), and their results were compared with those of a group of patients treated following existing clinical guides (PharmG-, N = 167). There was no evidence of significant differences in side effects between the two arms. Although patients who had their antipsychotic dose adjusted according to CYPs polymorphisms (PharmG+) had a bigger reduction in side effects than those treated as usual (PharmG-), the difference was not statistically significant (p > 0.05 for all comparisons). However, PharmG+ patients treated with CYP2D6 substrates that were carriers of CYP2D6 UMs or PMs variants showed a significantly higher improvement in global, psychic and other UKU side effects than PharmG- patients (p = 0.02, p = 0.05 and p = 0.01, respectively). PharmG+ clozapine treated patients with CYP1A2 or CYP2C19 UM and PMs variants also showed higher reductions in UKU scores than PharmG- clozapine patients in general. However, those differences were not statistically significant. Pharmacogenetic interventions may improve the safety of antipsychotic treatments by reducing associated side effects. This intervention may be particularly useful when considering treatment with antipsychotics with one major metabolic pathway, and therefore more susceptible to be affected by functional variants of CYP enzymes.
Collapse
Affiliation(s)
- Maria J. Arranz
- 0000 0004 1794 4956grid.414875.bFundació Docència i Recerca Mútua Terrassa, Terrassa, Spain ,Centro de Investigación en Red de Salud Mental, CIBERSAM, Madrid, Spain
| | - Alex Gonzalez-Rodriguez
- 0000 0000 9238 6887grid.428313.fDepartment of Mental Health, Parc Taulí University Hospital Sabadell, Barcelona, Spain
| | - Josefina Perez-Blanco
- Centro de Investigación en Red de Salud Mental, CIBERSAM, Madrid, Spain ,0000 0004 1768 8905grid.413396.aDepartment of Psychiatry, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Rafael Penadés
- Centro de Investigación en Red de Salud Mental, CIBERSAM, Madrid, Spain ,Barcelona Clinic Schizophrenia Unit (BCSU), Neurosciences Institute, Hospital Clinic of Barcelona, University of Barcelona, IDIBAPS, Barcelona, Spain
| | - Blanca Gutierrez
- 0000000121678994grid.4489.1Department of Psychiatry, University of Granada, Granada, Spain
| | - Laura Ibañez
- 0000 0004 1794 4956grid.414875.bFundació Docència i Recerca Mútua Terrassa, Terrassa, Spain
| | - Barbara Arias
- Centro de Investigación en Red de Salud Mental, CIBERSAM, Madrid, Spain ,0000 0004 1937 0247grid.5841.8Department of Biologia Evolutiva, Ecologia i Ciències Ambientals, Facultat de Biologia, Institut de Biomedicina de la Universitat de Barcelona (IBUB), Barcelona, Spain
| | - Mercè Brunet
- 0000 0000 9635 9413grid.410458.cPharmacology and Toxicology Unit, Department of Biochemistry and Molecular Genetics, Hospital Clinic, Barcelona, Spain
| | - Jorge Cervilla
- 0000000121678994grid.4489.1Department of Psychiatry, University of Granada, Granada, Spain
| | - Juliana Salazar
- 0000 0004 1768 8905grid.413396.aGenetics Department, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Rosa Catalan
- Centro de Investigación en Red de Salud Mental, CIBERSAM, Madrid, Spain. .,Barcelona Clinic Schizophrenia Unit (BCSU), Neurosciences Institute, Hospital Clinic of Barcelona, University of Barcelona, IDIBAPS, Barcelona, Spain.
| |
Collapse
|
26
|
van der Wouden CH, van Rhenen MH, Jama WOM, Ingelman-Sundberg M, Lauschke VM, Konta L, Schwab M, Swen JJ, Guchelaar HJ. Development of the PGx-Passport: A Panel of Actionable Germline Genetic Variants for Pre-Emptive Pharmacogenetic Testing. Clin Pharmacol Ther 2019; 106:866-873. [PMID: 31038729 PMCID: PMC6771671 DOI: 10.1002/cpt.1489] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Accepted: 04/21/2019] [Indexed: 12/19/2022]
Abstract
Pre-emptive pharmacogenetics (PGx) testing of a panel of germline genetic variants represents a new model for personalized medicine. Clinical impact of PGx testing is maximized when all variant alleles for which actionable clinical guidelines are available are included in the test panel. However, no such standardized panel has been presented to date, impeding adoption, exchange, and continuity of PGx testing. We, therefore, developed such a panel, hereafter called the PGx-Passport, based on the actionable Dutch Pharmacogenetics Working Group (DPWG) guidelines. Germline-variant alleles were systematically selected using predefined criteria regarding allele population frequencies, effect on protein functionality, and association with drug response. A PGx-Passport of 58 germline variant alleles, located within 14 genes (CYP2B6, CYP2C9, CYP2C19, CYP2D6, CYP3A5, DPYD, F5, HLA-A, HLA-B, NUDT15, SLCO1B1, TPMT, UGT1A1, and VKORC1) was composed. This PGx-Passport can be used in combination with the DPWG guidelines to optimize drug prescribing for 49 commonly prescribed drugs.
Collapse
Affiliation(s)
- Cathelijne H van der Wouden
- Department of Clinical Pharmacy and Toxicology, Leiden University Medical Center, Leiden, The Netherlands.,Leiden Network for Personalised Therapeutics, Leiden, The Netherlands
| | | | - Wafa O M Jama
- Royal Dutch Pharmacists Association (KNMP), The Hague, The Netherlands
| | - Magnus Ingelman-Sundberg
- Section of Pharmacogenetics, Department of Physiology and Pharmacology, Biomedicum 5B Karolinska Institutet, Stockholm, Sweden
| | - Volker M Lauschke
- Section of Pharmacogenetics, Department of Physiology and Pharmacology, Biomedicum 5B Karolinska Institutet, Stockholm, Sweden
| | - Lidija Konta
- bio.logis Center for Human Genetics, Frankfurt am Main, Germany
| | - Matthias Schwab
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany.,Department of Clinical Pharmacology and Departments of Biochemistry and Pharmacy,, University Tuebingen, Tuebingen, Germany
| | - Jesse J Swen
- Department of Clinical Pharmacy and Toxicology, Leiden University Medical Center, Leiden, The Netherlands.,Leiden Network for Personalised Therapeutics, Leiden, The Netherlands
| | - Henk-Jan Guchelaar
- Department of Clinical Pharmacy and Toxicology, Leiden University Medical Center, Leiden, The Netherlands.,Leiden Network for Personalised Therapeutics, Leiden, The Netherlands
| |
Collapse
|
27
|
Jarvis JP, Peter AP, Shaman JA. Consequences of CYP2D6 Copy-Number Variation for Pharmacogenomics in Psychiatry. Front Psychiatry 2019; 10:432. [PMID: 31281270 PMCID: PMC6595891 DOI: 10.3389/fpsyt.2019.00432] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Accepted: 06/03/2019] [Indexed: 12/20/2022] Open
Abstract
Pharmacogenomics represents a potentially powerful enhancement to the current standard of care for psychiatric patients. However, a variety of biological and technical challenges must be addressed in order to provide adequate clinical decision support for personalized prescribing and dosing based on genomic data. This is particularly true in the case of CYP2D6, a key drug-metabolizing gene, which not only harbors multiple genetic variants known to affect enzyme function but also shows a broad range of copy-number and hybrid alleles in various patient populations. Here, we describe several challenges in the accurate measurement and interpretation of data from the CYP2D6 locus including the clinical consequences of increased copy number. We discuss best practices for overcoming these challenges and then explore various current and future applications of pharmacogenomic analysis of CYP2D6 in psychiatry.
Collapse
|
28
|
Mukerjee G, Huston A, Kabakchiev B, Piquette-Miller M, van Schaik R, Dorfman R. User considerations in assessing pharmacogenomic tests and their clinical support tools. NPJ Genom Med 2018; 3:26. [PMID: 30210808 PMCID: PMC6133969 DOI: 10.1038/s41525-018-0065-4] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Revised: 08/10/2018] [Accepted: 08/13/2018] [Indexed: 12/18/2022] Open
Abstract
Pharmacogenomic (PGx) testing is gaining recognition from physicians, pharmacists and patients as a tool for evidence-based medication management. However, seemingly similar PGx testing panels (and PGx-based decision support tools) can diverge in their technological specifications, as well as the genetic factors that determine test specificity and sensitivity, and hence offer different values for users. Reluctance to embrace PGx testing is often the result of unfamiliarity with PGx technology, a lack of knowledge about the availability of curated guidelines/evidence for drug dosing recommendations, and an absence of wide-spread institutional implementation efforts and educational support. Demystifying an often confusing and variable PGx marketplace can lead to greater acceptance of PGx as a standard-of-care practice that improves drug outcomes and provides a lifetime value for patients. Here, we highlight the key underlying factors of a PGx test that should be considered, and discuss the current progress of PGx implementation.
Collapse
Affiliation(s)
| | - Andrea Huston
- GeneYouIn Inc., 156 Front St. W., Toronto, ON Canada
| | - Boyko Kabakchiev
- GeneYouIn Inc., 156 Front St. W., Toronto, ON Canada.,2Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON Canada
| | | | - Ron van Schaik
- 4International Expert Center Pharmacogenetics, Department of Clinical Chemistry, Erasmus University Medical Center, Rotterdam, The Netherlands
| | | |
Collapse
|