1
|
Lock SK, Kappel DB, Owen MJ, Walters JTR, O'Donovan MC, Pardiñas AF, Legge SE. Antipsychotic and pharmacogenomic effects on cross-sectional symptom severity and cognitive ability in schizophrenia. EBioMedicine 2025; 116:105745. [PMID: 40347835 PMCID: PMC12139436 DOI: 10.1016/j.ebiom.2025.105745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 04/09/2025] [Accepted: 04/23/2025] [Indexed: 05/14/2025] Open
Abstract
BACKGROUND People with schizophrenia differ in the type and severity of symptoms experienced, as well as their response to medication. A better understanding of the factors that influence this heterogeneity is necessary for the development of individualised patient care. Here, we sought to investigate the relationships between phenotypic severity and both medication and pharmacogenomic variables in a cross-sectional sample of people with schizophrenia or schizoaffective disorder depressed type. METHODS Confirmatory factor analysis derived five dimensions relating to current symptom severity (positive symptoms, negative symptoms of diminished expressivity, negative symptoms of reduced motivation and pleasure, depression and suicide) and cognitive ability in participants prescribed with antipsychotic medication. Linear models were fit to test for associations between medication and pharmacogenomic variables with dimension scores in the full sample (N = 585), and in a sub-sample of participants prescribed clozapine (N = 215). FINDINGS Lower cognitive ability was associated with higher chlorpromazine-equivalent daily antipsychotic dose (β = -0.12; 95% CI, -0.19 to -0.05; p = 0.001) and with the prescription of clozapine (β = -0.498; 95% CI, -0.65 to -0.35; p = 3 × 10-10) and anticholinergic medication (β = -0.345; 95% CI, -0.55 to -0.14; p = 8 × 10-4). We also found associations between pharmacogenomics-inferred cytochrome P450 (CYP) enzyme activity and symptom dimensions. Increased genotype-predicted CYP2C19 and CYP3A5 activity were associated with reduced severity of the positive (β = -0.108; 95% CI, -0.19 to -0.03; p = 0.009) and both negative symptom dimensions (β = -0.113; 95% CI, -0.19 to -0.03; p = 0.007; β = -0.106; 95% CI, -0.19 to -0.02; p = 0.012), respectively. Faster predicted CYP1A2 activity was associated with higher cognitive dimension scores in people taking clozapine (β = 0.17; 95% CI, 0.05-0.29; p = 0.005). INTERPRETATION Our results confirm the importance of taking account of medication history (and particularly antipsychotic type and dose) in assessing potential correlates of cognitive impairment or poor functioning in patients with schizophrenia. We also highlight the potential for pharmacogenomic variation to be a useful tool to help guide drug prescription, although these findings require further validation. FUNDING Medical Research Council (MR/Y004094/1) and The National Center for Mental Health, funded by the Welsh Government through Health and Care Research Wales. SKL was funded by a PhD studentship from Mental Health Research UK (MHRUK). DBK, JTRW, MCOD and AFP were supported by the European Union's Horizon 2020 research and innovation programme under grant agreement 964874.
Collapse
Affiliation(s)
- Siobhan K Lock
- Centre for Neuropsychiatric Genetics and Genomics, Cardiff University, Hadyn Ellis Building, Maindy Road, Cardiff, Wales, CF24 4HQ, United Kingdom
| | - Djenifer B Kappel
- Centre for Neuropsychiatric Genetics and Genomics, Cardiff University, Hadyn Ellis Building, Maindy Road, Cardiff, Wales, CF24 4HQ, United Kingdom
| | - Michael J Owen
- Centre for Neuropsychiatric Genetics and Genomics, Cardiff University, Hadyn Ellis Building, Maindy Road, Cardiff, Wales, CF24 4HQ, United Kingdom
| | - James T R Walters
- Centre for Neuropsychiatric Genetics and Genomics, Cardiff University, Hadyn Ellis Building, Maindy Road, Cardiff, Wales, CF24 4HQ, United Kingdom
| | - Michael C O'Donovan
- Centre for Neuropsychiatric Genetics and Genomics, Cardiff University, Hadyn Ellis Building, Maindy Road, Cardiff, Wales, CF24 4HQ, United Kingdom
| | - Antonio F Pardiñas
- Centre for Neuropsychiatric Genetics and Genomics, Cardiff University, Hadyn Ellis Building, Maindy Road, Cardiff, Wales, CF24 4HQ, United Kingdom.
| | - Sophie E Legge
- Centre for Neuropsychiatric Genetics and Genomics, Cardiff University, Hadyn Ellis Building, Maindy Road, Cardiff, Wales, CF24 4HQ, United Kingdom.
| |
Collapse
|
2
|
Owen MJ, Bray NJ, Walters JTR, O'Donovan MC. Genomics of schizophrenia, bipolar disorder and major depressive disorder. Nat Rev Genet 2025:10.1038/s41576-025-00843-0. [PMID: 40355602 DOI: 10.1038/s41576-025-00843-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/10/2025] [Indexed: 05/14/2025]
Abstract
Schizophrenia, bipolar disorder and major depressive disorder - which are the most common adult disorders requiring psychiatric care - contribute substantially to premature mortality and morbidity globally. Treatments for these disorders are suboptimal, there are no diagnostic pathologies or biomarkers and their pathophysiologies are poorly understood. Novel therapeutic and diagnostic approaches are thus badly needed. Given the high heritability of psychiatric disorders, psychiatry has potentially much to gain from the application of genomics to identify molecular risk mechanisms and to improve diagnosis. Recent large-scale, genome-wide association studies and sequencing studies, together with advances in functional genomics, have begun to illuminate the genetic architectures of schizophrenia, bipolar disorder and major depressive disorder and to identify potential biological mechanisms. Genomic findings also point to the aetiological relationships between different diagnoses and to the relationships between adult psychiatric disorders and childhood neurodevelopmental conditions.
Collapse
Affiliation(s)
- Michael J Owen
- Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff, UK.
| | - Nicholas J Bray
- Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff, UK
| | - James T R Walters
- Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff, UK
| | - Michael C O'Donovan
- Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff, UK
| |
Collapse
|
3
|
Zhou S, Hu X, Zhou P, Si J, Jiang Y. Nomograms based on clinical factors to predict abnormal metabolism of psychotropic drugs. Biomed Rep 2025; 22:83. [PMID: 40151799 PMCID: PMC11948297 DOI: 10.3892/br.2025.1961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Accepted: 02/17/2025] [Indexed: 03/29/2025] Open
Abstract
Interindividual variability in drug metabolism serves a critical role in the occurrence of adverse drug reactions. Factors such as age, sex, body mass index (BMI) and liver and renal function can influence the metabolism of antipsychotic medications. To the best of our knowledge, however, clinical prediction models based on these factors for estimating drug-metabolizing capacity have not yet been developed. Between January 2022 and September 2023, 185 adult patients (aged ≥18 years) who did not have cancer and were not critically ill, with or without comorbidities such diabetes, hypertension and liver and kidney diseases, who underwent pharmacogenetic testing at The First Hospital of Jilin University (Changchun, China) were enrolled. Clinical data were collected, and the participants were divided into training and validation cohorts. Logistic regression was performed to identify significant risk factors, which were incorporated into multivariable models to construct nomograms predicting psychotropic drug metabolism. A total of eight clinical indicators (BMI, hypertension, alkaline phosphatase, aspartate aminotransferase, cholinesterase, albumin to globulin ratio, urea, and uric acid) were significantly associated with psychotropic drug metabolism (all P<0.05). Based on these indicators, along with age and sex, prediction models for psychotropic drug metabolism were developed. The areas under the receiver operating characteristic curves for haloperidol, olanzapine, paroxetine, mirtazapine/venlafaxine and oxazepam/lorazepam in the validation dataset were 0.767, 0.767, 0.705, 0.740 and 0.789, respectively, indicating the models had moderate diagnostic efficiency. Nomograms were constructed to demonstrate the contribution of each indicator to drug metabolism capacity. To the best of our knowledge, the present study is the first to develop predictive models for psychotropic drug metabolism. These models offer clinicians practical tools to identify patients with impaired drug-metabolizing capacity, thereby enabling more precise and personalized medication management.
Collapse
Affiliation(s)
- Shuai Zhou
- Genetic Diagnosis Center, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Xinyuan Hu
- Genetic Diagnosis Center, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Peiwen Zhou
- Genetic Diagnosis Center, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Junzhuo Si
- Genetic Diagnosis Center, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Yanfang Jiang
- Genetic Diagnosis Center, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| |
Collapse
|
4
|
Mansoori A, Bazrafshan A, Ahmadi J, Mosavat SH. Adjunctive ketamine vs. buprenorphine in co-occurring major depressive disorder and opioid use disorder: a randomized, double-blind clinical trial assessing anxiety symptom severity and craving intensity. Trials 2025; 26:133. [PMID: 40247293 PMCID: PMC12004569 DOI: 10.1186/s13063-025-08836-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 04/07/2025] [Indexed: 04/19/2025] Open
Abstract
BACKGROUND The concomitant presence of major depressive disorder (MDD) and opioid use disorder (OUD) poses a formidable clinical challenge, warranting effective interventions that address both psychiatric and addictive components. AIMS This study sought to compare the efficacy of adjunctive ketamine and buprenorphine in mitigating anxiety symptom severity and craving intensity in individuals with co-occurring MDD and OUD. METHODS A randomized, double-blind clinical trial was conducted, involving individuals meeting diagnostic criteria for both MDD and OUD. Participants were randomly assigned to receive adjunctive ketamine or buprenorphine, in conjunction with standard psychiatric and addiction treatments. Anxiety symptom severity and craving intensity were assessed using Hamilton Anxiety Rating Scale (HAM-A), and the Opioid Craving Scale after 2 h, 24 h, and 7 days. RESULTS The findings revealed distinct treatment trajectories, with ketamine demonstrating rapid and substantial reduction in anxiety symptom severity within hours of administration, accompanied by a pronounced decline in opioid craving intensity. In contrast, buprenorphine was associated with a more gradual but sustained improvement in anxiety symptoms over several days, paralleled by a modest initial reduction in opioid craving, followed by persistent attenuation. CONCLUSIONS In conclusion, this randomized clinical trial provides evidence supporting the efficacy of adjunctive Ketamine and Buprenorphine in reducing anxiety symptoms and craving intensity in patients with co-occurring MDD and OUD. TRIAL REGISTRATION IRCT20211214053411N1.
Collapse
Affiliation(s)
- Arash Mansoori
- Research Center for Psychiatry and Behavioral Science, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Amir Bazrafshan
- Research Center for Psychiatry and Behavioral Science, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Jamshid Ahmadi
- Substance Abuse Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- IMCES (Institute for Multicultural Counseling & Educational Services), Los Angeles, CA, USA
| | - Seyed Hamdollah Mosavat
- Research Center for Traditional Medicine and History of Medicine, Department of Persian Medicine, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
5
|
Mongelli M, De Angelis B, delle Cave V, Greco G, De Arcangelis A, Bernagozzi A, Salvemini C, Calabrese M, Christille JM, Cavalli A, Gustincich S, Monaci MG. The Public Knowledge of Precision Medicine and Genomic Research: A Survey in the Aosta Valley. J Pers Med 2025; 15:80. [PMID: 40137396 PMCID: PMC11943031 DOI: 10.3390/jpm15030080] [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: 12/19/2024] [Revised: 02/14/2025] [Accepted: 02/20/2025] [Indexed: 03/27/2025] Open
Abstract
Background: Precision medicine (PM) considers the genetic variability of individuals to identify tailored diagnosis and treatments. It relies on the possibility of gathering the widest possible health data and genetic information from individuals to obtain a broad pool of comparative data. To achieve this goal, the Region of Valle d'Aosta, since 2019, has co-financed the research center CMP3VdA, aiming to sequence 5000 genomes of patients with neurodevelopmental, neurodegenerative, oncological, and organ transplantation diseases, and to investigate the genetic variability of the resident population. Methods: This paper presents the results of an online survey of 472 (328F) respondents regarding willingness to participate in the genomic project and awareness, attitudes, and concerns about PM. Results: The main results show that the vast majority (92.6%) would be willing to participate-a higher percentage than in previous studies. Age, education, and prior experience in the healthcare sector are significant factors influencing the awareness of PM. Additionally, subgroups organized by age, gender, and religiosity show significant differences with respect to participants' reasons for participating in research and which types of biological samples they would be willing to donate. Conclusions: Our findings can serve as a guide for stakeholders-particularly policymakers-to target institutional communication and achieve maximum participation in genomic research projects.
Collapse
Affiliation(s)
- Matteo Mongelli
- CMP3VdA, Istituto Italiano di Tecnologia (IIT), Via Lavoratori Vittime del Col du Mont, 28, 11100 Aosta, Italy; (M.M.); (B.D.A.); (A.D.A.); (A.B.); (C.S.); (M.C.); (J.M.C.); (A.C.); (S.G.)
| | - Biagio De Angelis
- CMP3VdA, Istituto Italiano di Tecnologia (IIT), Via Lavoratori Vittime del Col du Mont, 28, 11100 Aosta, Italy; (M.M.); (B.D.A.); (A.D.A.); (A.B.); (C.S.); (M.C.); (J.M.C.); (A.C.); (S.G.)
| | - Valeria delle Cave
- Communication and External Relations Directorate, Istituto Italiano di Tecnologia (IIT), Via Morego 30, 16163 Genova, Italy (G.G.)
| | - Giuliano Greco
- Communication and External Relations Directorate, Istituto Italiano di Tecnologia (IIT), Via Morego 30, 16163 Genova, Italy (G.G.)
| | - Arianna De Arcangelis
- CMP3VdA, Istituto Italiano di Tecnologia (IIT), Via Lavoratori Vittime del Col du Mont, 28, 11100 Aosta, Italy; (M.M.); (B.D.A.); (A.D.A.); (A.B.); (C.S.); (M.C.); (J.M.C.); (A.C.); (S.G.)
| | - Andrea Bernagozzi
- CMP3VdA, Istituto Italiano di Tecnologia (IIT), Via Lavoratori Vittime del Col du Mont, 28, 11100 Aosta, Italy; (M.M.); (B.D.A.); (A.D.A.); (A.B.); (C.S.); (M.C.); (J.M.C.); (A.C.); (S.G.)
- Fondazione Clément Fillietroz, Astronomical Observatory of the Autonomous Region of the Aosta Valley (OAVdA), Loc. Lignan 39, 11020 Nus, Italy
| | - Chiara Salvemini
- CMP3VdA, Istituto Italiano di Tecnologia (IIT), Via Lavoratori Vittime del Col du Mont, 28, 11100 Aosta, Italy; (M.M.); (B.D.A.); (A.D.A.); (A.B.); (C.S.); (M.C.); (J.M.C.); (A.C.); (S.G.)
- Fondazione Clément Fillietroz, Astronomical Observatory of the Autonomous Region of the Aosta Valley (OAVdA), Loc. Lignan 39, 11020 Nus, Italy
| | - Matteo Calabrese
- CMP3VdA, Istituto Italiano di Tecnologia (IIT), Via Lavoratori Vittime del Col du Mont, 28, 11100 Aosta, Italy; (M.M.); (B.D.A.); (A.D.A.); (A.B.); (C.S.); (M.C.); (J.M.C.); (A.C.); (S.G.)
- Fondazione Clément Fillietroz, Astronomical Observatory of the Autonomous Region of the Aosta Valley (OAVdA), Loc. Lignan 39, 11020 Nus, Italy
| | - Jean Marc Christille
- CMP3VdA, Istituto Italiano di Tecnologia (IIT), Via Lavoratori Vittime del Col du Mont, 28, 11100 Aosta, Italy; (M.M.); (B.D.A.); (A.D.A.); (A.B.); (C.S.); (M.C.); (J.M.C.); (A.C.); (S.G.)
- Fondazione Clément Fillietroz, Astronomical Observatory of the Autonomous Region of the Aosta Valley (OAVdA), Loc. Lignan 39, 11020 Nus, Italy
| | - Andrea Cavalli
- CMP3VdA, Istituto Italiano di Tecnologia (IIT), Via Lavoratori Vittime del Col du Mont, 28, 11100 Aosta, Italy; (M.M.); (B.D.A.); (A.D.A.); (A.B.); (C.S.); (M.C.); (J.M.C.); (A.C.); (S.G.)
- Computational and Chemical Biology, Istituto Italiano di Tecnologia (IIT), Via Morego 30, 16163 Genova, Italy
| | - Stefano Gustincich
- CMP3VdA, Istituto Italiano di Tecnologia (IIT), Via Lavoratori Vittime del Col du Mont, 28, 11100 Aosta, Italy; (M.M.); (B.D.A.); (A.D.A.); (A.B.); (C.S.); (M.C.); (J.M.C.); (A.C.); (S.G.)
- Center for Human Technologies, Non-Coding RNAs and RNA-Based Therapeutics, Istituto Italiano di Tecnologia (IIT), Via Enrico Melen 83, 16152 Genova, Italy
| | - Maria Grazia Monaci
- Department of Human and Social Science, University of Valle d’Aosta, Strada Cappuccini 2A, 11100 Aosta, Italy
| |
Collapse
|
6
|
Koch E, Pardiñas AF, O'Connell KS, Selvaggi P, Camacho Collados J, Babic A, Marshall SE, Van der Eycken E, Angulo C, Lu Y, Sullivan PF, Dale AM, Molden E, Posthuma D, White N, Schubert A, Djurovic S, Heimer H, Stefánsson H, Stefánsson K, Werge T, Sønderby I, O'Donovan MC, Walters JTR, Milani L, Andreassen OA. How Real-World Data Can Facilitate the Development of Precision Medicine Treatment in Psychiatry. Biol Psychiatry 2024; 96:543-551. [PMID: 38185234 PMCID: PMC11758919 DOI: 10.1016/j.biopsych.2024.01.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 12/20/2023] [Accepted: 01/02/2024] [Indexed: 01/09/2024]
Abstract
Precision medicine has the ambition to improve treatment response and clinical outcomes through patient stratification and holds great potential for the treatment of mental disorders. However, several important factors are needed to transform current practice into a precision psychiatry framework. Most important are 1) the generation of accessible large real-world training and test data including genomic data integrated from multiple sources, 2) the development and validation of advanced analytical tools for stratification and prediction, and 3) the development of clinically useful management platforms for patient monitoring that can be integrated into health care systems in real-life settings. This narrative review summarizes strategies for obtaining the key elements-well-powered samples from large biobanks integrated with electronic health records and health registry data using novel artificial intelligence algorithms-to predict outcomes in severe mental disorders and translate these models into clinical management and treatment approaches. Key elements are massive mental health data and novel artificial intelligence algorithms. For the clinical translation of these strategies, we discuss a precision medicine platform for improved management of mental disorders. We use cases to illustrate how precision medicine interventions could be brought into psychiatry to improve the clinical outcomes of mental disorders.
Collapse
Affiliation(s)
- Elise Koch
- Norwegian Centre for Mental Disorders Research, Division of Mental Health and Addiction, Oslo University Hospital, and Institute of Clinical Medicine, University of Oslo, Oslo, Norway.
| | - Antonio F Pardiñas
- Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Kevin S O'Connell
- Norwegian Centre for Mental Disorders Research, Division of Mental Health and Addiction, Oslo University Hospital, and Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Pierluigi Selvaggi
- Department of Translational Biomedicine and Neuroscience, University of Bari Aldo Moro, Bari, Italy
| | - José Camacho Collados
- CardiffNLP, School of Computer Science and Informatics, Cardiff University, Cardiff, United Kingdom
| | | | | | - Erik Van der Eycken
- Global Alliance of Mental Illness Advocacy Networks-Europe, Brussels, Belgium
| | - Cecilia Angulo
- Global Alliance of Mental Illness Advocacy Networks-Europe, Brussels, Belgium
| | - Yi Lu
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Solna, Sweden
| | - Patrick F Sullivan
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Solna, Sweden; Departments of Genetics and Psychiatry, University of North Carolina, Chapel Hill, North Carolina
| | - Anders M Dale
- Multimodal Imaging Laboratory, University of California San Diego, La Jolla, California; Departments of Radiology, Psychiatry, and Neurosciences, University of California, San Diego, La Jolla, California
| | - Espen Molden
- Center for Psychopharmacology, Diakonhjemmet Hospital, Oslo, Norway
| | - Danielle Posthuma
- Department of Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Nathan White
- CorTechs Laboratories, Inc., San Diego, California
| | | | - Srdjan Djurovic
- Department of Medical Genetics, Oslo University Hospital, Oslo, Norway; The Norwegian Centre for Mental Disorders Research Centre, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Hakon Heimer
- Norwegian Centre for Mental Disorders Research, Division of Mental Health and Addiction, Oslo University Hospital, and Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Nordic Society of Human Genetics and Precision Medicine, Copenhagen, Denmark
| | | | | | - Thomas Werge
- Institute of Biological Psychiatry, Mental Health Center Sct. Hans, Mental Health Services Copenhagen, Roskilde, Denmark; Lundbeck Foundation Initiative for Integrative Psychiatric Research, Copenhagen, Denmark; Lundbeck Foundation GeoGenetics Centre, GLOBE Institute, University of Copenhagen, Copenhagen, Denmark
| | - Ida Sønderby
- Norwegian Centre for Mental Disorders Research, Division of Mental Health and Addiction, Oslo University Hospital, and Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Department of Medical Genetics, Oslo University Hospital, Oslo, Norway; KG Jebsen Centre for Neurodevelopmental Disorders, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Michael C O'Donovan
- Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - James T R Walters
- Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Lili Milani
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia; Genetics and Personalized Medicine Clinic, Tartu University Hospital, Tartu, Estonia
| | - Ole A Andreassen
- Norwegian Centre for Mental Disorders Research, Division of Mental Health and Addiction, Oslo University Hospital, and Institute of Clinical Medicine, University of Oslo, Oslo, Norway; KG Jebsen Centre for Neurodevelopmental Disorders, University of Oslo and Oslo University Hospital, Oslo, Norway.
| |
Collapse
|
7
|
Sharew NT, Clark SR, Schubert KO, Amare AT. Pharmacogenomic scores in psychiatry: systematic review of current evidence. Transl Psychiatry 2024; 14:322. [PMID: 39107294 PMCID: PMC11303815 DOI: 10.1038/s41398-024-02998-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Revised: 06/21/2024] [Accepted: 06/27/2024] [Indexed: 08/10/2024] Open
Abstract
In the past two decades, significant progress has been made in the development of polygenic scores (PGSs). One specific application of PGSs is the development and potential use of pharmacogenomic- scores (PGx-scores) to identify patients who can benefit from a specific medication or are likely to experience side effects. This systematic review comprehensively evaluates published PGx-score studies in psychiatry and provides insights into their potential clinical use and avenues for future development. A systematic literature search was conducted across PubMed, EMBASE, and Web of Science databases until 22 August 2023. This review included fifty-three primary studies, of which the majority (69.8%) were conducted using samples of European ancestry. We found that over 90% of PGx-scores in psychiatry have been developed based on psychiatric and medical diagnoses or trait variants, rather than pharmacogenomic variants. Among these PGx-scores, the polygenic score for schizophrenia (PGSSCZ) has been most extensively studied in relation to its impact on treatment outcomes (32 publications). Twenty (62.5%) of these studies suggest that individuals with higher PGSSCZ have negative outcomes from psychotropic treatment - poorer treatment response, higher rates of treatment resistance, more antipsychotic-induced side effects, or more psychiatric hospitalizations, while the remaining studies did not find significant associations. Although PGx-scores alone accounted for at best 5.6% of the variance in treatment outcomes (in schizophrenia treatment resistance), together with clinical variables they explained up to 13.7% (in bipolar lithium response), suggesting that clinical translation might be achieved by including PGx-scores in multivariable models. In conclusion, our literature review found that there are still very few studies developing PGx-scores using pharmacogenomic variants. Research with larger and diverse populations is required to develop clinically relevant PGx-scores, using biology-informed and multi-phenotypic polygenic scoring approaches, as well as by integrating clinical variables with these scores to facilitate their translation to psychiatric practice.
Collapse
Affiliation(s)
- Nigussie T Sharew
- Discipline of Psychiatry, Adelaide Medical School, The University of Adelaide, Adelaide, SA, Australia
- Asrat Woldeyes Health Science Campus, Debre Berhan University, Debre Berhan, Ethiopia
| | - Scott R Clark
- Discipline of Psychiatry, Adelaide Medical School, The University of Adelaide, Adelaide, SA, Australia
| | - K Oliver Schubert
- Discipline of Psychiatry, Adelaide Medical School, The University of Adelaide, Adelaide, SA, Australia
- Division of Mental Health, Northern Adelaide Local Health Network, SA Health, Adelaide, Australia
- Headspace Adelaide Early Psychosis - Sonder, Adelaide, SA, Australia
| | - Azmeraw T Amare
- Discipline of Psychiatry, Adelaide Medical School, The University of Adelaide, Adelaide, SA, Australia.
| |
Collapse
|
8
|
Tesfamicael KG, Zhao L, Fernández-Rodríguez R, Adelson DL, Musker M, Polasek TM, Lewis MD. Efficacy and safety of pharmacogenomic-guided antidepressant prescribing in patients with depression: an umbrella review and updated meta-analysis. Front Psychiatry 2024; 15:1276410. [PMID: 39086729 PMCID: PMC11289719 DOI: 10.3389/fpsyt.2024.1276410] [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: 08/11/2023] [Accepted: 06/26/2024] [Indexed: 08/02/2024] Open
Abstract
Aim To determine the efficacy and safety of pharmacogenomics (PGx)-guided antidepressant prescribing in patients with depression through an umbrella review and updated meta-analysis. Methods A comprehensive systematic search was conducted on PsycINFO, PubMed, Embase and the Cochrane databases. The pooled effect sizes of randomized controlled trials (RCTs) were expressed as mean differences for continuous data and risk ratios for noncontinuous data. Results Patients who received PGx-guided medications were 41% to 78% more likely to achieve remission and 20% to 49% more likely to respond to antidepressants than patients receiving treatment-as-usual (TAU). Conclusion PGx-guided antidepressant prescribing improves the treatment of depression. However, the significance and magnitude of the benefit varies widely between studies and different PGx testing panels. Systematic review registration https://www.crd.york.ac.uk/prospero/, identifier CRD42022321324.
Collapse
Affiliation(s)
- Kiflu G. Tesfamicael
- School of Biological Sciences, University of Adelaide, Adelaide, SA, Australia
- Lifelong Health, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, SA, Australia
| | - Lijun Zhao
- Lifelong Health, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, SA, Australia
- Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
| | | | - David L. Adelson
- School of Biological Sciences, University of Adelaide, Adelaide, SA, Australia
| | - Michael Musker
- Adelaide Nursing School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, South Australia, Australia
| | - Thomas M. Polasek
- Centre for Medicine Use and Safety, Monash University, Melbourne, VIC, Australia
| | - Martin David Lewis
- School of Biological Sciences, University of Adelaide, Adelaide, SA, Australia
- Lifelong Health, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, SA, Australia
| |
Collapse
|
9
|
Abdel‐latif R, Badji R, Mohammed S, Al‐Muftah W, Mbarek H, Darwish D, Assaf D, Al‐Badriyeh D, Elewa H, Afifi N, Masoodi NA, Omar AS, Al Suwaidi J, Bujassoum S, Al Hail M, Ismail SI, Althani A. QPGx-CARES: Qatar pharmacogenetics clinical applications and research enhancement strategies. Clin Transl Sci 2024; 17:e13800. [PMID: 38818903 PMCID: PMC11140449 DOI: 10.1111/cts.13800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 04/03/2024] [Accepted: 04/05/2024] [Indexed: 06/01/2024] Open
Abstract
Pharmacogenetic (PGx)-informed medication prescription is a cutting-edge genomic application in contemporary medicine, offering the potential to overcome the conventional "trial-and-error" approach in drug prescription. The ability to use an individual's genetic profile to predict drug responses allows for personalized drug and dosage selection, thereby enhancing the safety and efficacy of treatments. However, despite significant scientific and clinical advancements in PGx, its integration into routine healthcare practices remains limited. To address this gap, the Qatar Genome Program (QGP) has embarked on an ambitious initiative known as QPGx-CARES (Qatar Pharmacogenetics Clinical Applications and Research Enhancement Strategies), which aims to set a roadmap for optimizing PGx research and clinical implementation on a national scale. The goal of QPGx-CARES initiative is to integrate PGx testing into clinical settings with the aim of improving patient health outcomes. In 2022, QGP initiated several implementation projects in various clinical settings. These projects aimed to evaluate the clinical utility of PGx testing, gather valuable insights into the effective dissemination of PGx data to healthcare professionals and patients, and identify the gaps and the challenges for wider adoption. QPGx-CARES strategy aimed to integrate evidence-based PGx findings into clinical practice, focusing on implementing PGx testing for cardiovascular medications, supported by robust scientific evidence. The current initiative sets a precedent for the nationwide implementation of precision medicine across diverse clinical domains.
Collapse
Affiliation(s)
- Rania Abdel‐latif
- Qatar Genome Program, Qatar Precision Health InstituteQatar FoundationDohaQatar
| | - Radja Badji
- Qatar Genome Program, Qatar Precision Health InstituteQatar FoundationDohaQatar
| | | | - Wadha Al‐Muftah
- Qatar Genome Program, Qatar Precision Health InstituteQatar FoundationDohaQatar
| | - Hamdi Mbarek
- Qatar Genome Program, Qatar Precision Health InstituteQatar FoundationDohaQatar
| | - Dima Darwish
- Qatar Genome Program, Qatar Precision Health InstituteQatar FoundationDohaQatar
| | - Duha Assaf
- Qatar Genome Program, Qatar Precision Health InstituteQatar FoundationDohaQatar
| | | | - Hazem Elewa
- College of Pharmacy, QU HealthQatar UniversityDohaQatar
| | - Nahla Afifi
- Qatar Biobank for Medical ResearchQatar Foundation for Education, Science, and CommunityDohaQatar
| | | | - Amr Salah Omar
- Cardiology and Cardiovascular SurgeryDepartment Hamad Medical CorporationDohaQatar
| | - Jassim Al Suwaidi
- Cardiology and Cardiovascular SurgeryDepartment Hamad Medical CorporationDohaQatar
| | - Salha Bujassoum
- Medical Oncology, National Center for Cancer Care and ResearchDepartment Hamad Medical CorporationDohaQatar
| | - Moza Al Hail
- Pharmacy DepartmentHamad Medical CorporationDohaQatar
| | - Said I. Ismail
- Qatar Genome Program, Qatar Precision Health InstituteQatar FoundationDohaQatar
| | - Asma Althani
- Biomedical Research CenterQatar UniversityDohaQatar
| |
Collapse
|
10
|
Kappel DB, Rees E, Fenner E, King A, Jansen J, Helthuis M, Owen MJ, O'Donovan MC, Walters JTR, Pardiñas AF. Rare variants in pharmacogenes influence clozapine metabolism in individuals with schizophrenia. Eur Neuropsychopharmacol 2024; 80:47-54. [PMID: 38310750 DOI: 10.1016/j.euroneuro.2023.12.007] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 12/14/2023] [Accepted: 12/18/2023] [Indexed: 02/06/2024]
Abstract
Clozapine is the only licensed medication for treatment-resistant schizophrenia (TRS). Few predictors for variation in response to clozapine have been identified, but clozapine metabolism is known to influence therapeutic response and adverse side effects. Here, we expand on genome-wide studies of clozapine metabolism, previously focused on common genetic variation, by analysing whole-exome sequencing data from 2062 individuals with schizophrenia taking clozapine in the UK. We investigated whether rare genomic variation in genes and gene sets involved in the clozapine metabolism pathway influences plasma concentrations of clozapine metabolites, assessed through the longitudinal analysis of 6585 pharmacokinetic assays. We observed a statistically significant association between the burden of rare damaging coding variants (MAF ≤ 1 %) in gene sets broadly related to drug pharmacokinetics and lower clozapine (β = -0.054, SE = 0.019, P-value = 0.005) concentrations in plasma. We estimate that the effects in clozapine plasma concentrations of a single damaging allele in this gene set are akin to reducing the clozapine dose by about 35 mg/day. The gene-based analysis identified rare variants in CYP1A2, which encodes the enzyme responsible for converting clozapine to norclozapine, as having the strongest effects of any gene on clozapine metabolism (β = 0.324, SE = 0.124, P = 0.009). Our findings support the hypothesis that rare genetic variants in known drug-metabolising enzymes and transporters can markedly influence clozapine plasma concentrations; these results suggest that pharmacogenomic efforts trying to predict clozapine metabolism and personalise drug therapy could benefit from the inclusion of rare damaging variants in pharmacogenes beyond those already identified and catalogued as PGx star alleles.
Collapse
Affiliation(s)
- Djenifer B Kappel
- Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Elliott Rees
- Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Eilidh Fenner
- Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Adrian King
- Magna Laboratories Ltd., Ross-on-Wye, United Kingdom
| | - John Jansen
- Leyden Delta B.V., Nijmegen, The Netherlands
| | | | - Michael J Owen
- Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Michael C O'Donovan
- Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - James T R Walters
- Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Antonio F Pardiñas
- Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff, United Kingdom.
| |
Collapse
|
11
|
Patel W, Shankar RG, Smith MA, Snodgrass HR, Pirmohamed M, Jorgensen AL, Alfirevic A, Dickens D. Role of Transporters and Enzymes in Metabolism and Distribution of 4-Chlorokynurenine (AV-101). Mol Pharm 2024; 21:550-563. [PMID: 38261609 PMCID: PMC10848289 DOI: 10.1021/acs.molpharmaceut.3c00700] [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: 08/07/2023] [Revised: 12/08/2023] [Accepted: 12/08/2023] [Indexed: 01/25/2024]
Abstract
4-Chlorokynurenine (4-Cl-KYN, AV-101) is a prodrug of a NMDA receptor antagonist and is in clinical development for potential CNS indications. We sought to further understand the distribution and metabolism of 4-Cl-KYN, as this information might provide a strategy to enhance the clinical development of this drug. We used excretion studies in rats, in vitro transporter assays, and pharmacogenetic analysis of clinical trial data to determine how 4-Cl-KYN and metabolites are distributed. Our data indicated that a novel acetylated metabolite (N-acetyl-4-Cl-KYN) did not affect the uptake of 4-Cl-KYN across the blood-brain barrier via LAT1. 4-Cl-KYN and its metabolites were found to be renally excreted in rodents. In addition, we found that N-acetyl-4-Cl-KYN inhibited renal and hepatic transporters involved in excretion. Thus, this metabolite has the potential to limit the excretion of a range of compounds. Our pharmacogenetic analysis found that a SNP in N-acetyltransferase 8 (NAT8, rs13538) was linked to levels of N-acetyl-4-Cl-KYN relative to 4-Cl-KYN found in the plasma and that a SNP in SLC7A5 (rs28582913) was associated with the plasma levels of the active metabolite, 7-Cl-KYNA. Thus, we have a pharmacogenetics-based association for plasma drug level that could aid in the drug development of 4-Cl-KYN and have investigated the interaction of a novel metabolite with drug transporters.
Collapse
Affiliation(s)
- Waseema Patel
- Department
of Pharmacology and Therapeutics, University
of Liverpool, Liverpool L69 3GL, United
Kingdom
| | - Ravi G. Shankar
- Institute
of Population Health, University of Liverpool, Liverpool L69 3GL, United Kingdom
| | - Mark A. Smith
- Vistagen
Therapeutics, Inc., 343 Allerton Ave, South San Francisco, California 94080, United States
- Medical
College of Georgia, 1120
15th St, Augusta, Georgia 30912, United States
| | - H. Ralph Snodgrass
- Formerly
at Vistagen Therapeutics, Inc., 343 Allerton Ave, South San Francisco, California 94080, United States
| | - Munir Pirmohamed
- Department
of Pharmacology and Therapeutics, University
of Liverpool, Liverpool L69 3GL, United
Kingdom
| | - Andrea L. Jorgensen
- Institute
of Population Health, University of Liverpool, Liverpool L69 3GL, United Kingdom
| | - Ana Alfirevic
- Department
of Pharmacology and Therapeutics, University
of Liverpool, Liverpool L69 3GL, United
Kingdom
| | - David Dickens
- Department
of Pharmacology and Therapeutics, University
of Liverpool, Liverpool L69 3GL, United
Kingdom
| |
Collapse
|
12
|
Cheng Y, Liu H, Yuan R, Yuan K, Yu S. Effectiveness of pharmacogenomics on the response and remission of treatment-resistant depression: a meta-analysis of randomised controlled trials. Gen Psychiatr 2023; 36:e101050. [PMID: 38155841 PMCID: PMC10753713 DOI: 10.1136/gpsych-2023-101050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Accepted: 10/27/2023] [Indexed: 12/30/2023] Open
Abstract
Background Pharmacogenomics (PGx) is a promising tool to realise tailored drug therapy for depression. Aims To investigate the treatment efficacy of PGx for treatment-resistant depression (TRD) compared with treatment as usual. Methods A systematic search was conducted in PubMed, Embase, the Cochrane Library, Web of Science and PsycINFO to identify relevant studies published from inception to 15 April 2023. Two-arm randomised controlled trials (RCTs) exploring the efficacy of PGx-guided versus unguided treatment for TRD were included. The risk of bias in the included studies was evaluated using the Cochrane risk of bias assessment tool. The overall quality of evidence was assessed using the Grading of Recommendations, Assessment, Development and Evaluation (GRADE) approach. Results Seven RCTs (n=3003) comparing PGx-guided (n=1492) and unguided (n=1511) groups were identified and analysed. PGx-guided treatment was superior to treatment as usual in response (relative risk (RR)=1.31; 95% confidence interval (95% CI): 1.15 to 1.49; p<0.001) and remission (RR=1.40; 95% CI: 1.09 to 1.80; p=0.009) improvements. Effect sizes for acceptability (RR=0.90; 95% CI: 0.80 to 1.02; p=0.100) and side effect burden (RR=0.58; 95% CI: 0.29 to 1.15; p=0.120) between the two groups were not statistically different. The overall quality of evidence was rated from 'very low' (25%) to 'low' (75%) based on the GRADE criteria. Conclusions PGx-guided treatment has shown a small overall effect in improving the response and remission rates for patients with TRD. However, these results should be interpreted cautiously because of the few included studies and the low quality of evidence. Further high-quality clinical trials are warranted to confirm the findings. PROSPERO registration number CRD42022340182.
Collapse
Affiliation(s)
- Yu Cheng
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hongmei Liu
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ruixue Yuan
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Kai Yuan
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shunying Yu
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
13
|
Vuic B, Milos T, Tudor L, Nikolac Perkovic M, Konjevod M, Nedic Erjavec G, Farkas V, Uzun S, Mimica N, Svob Strac D. Pharmacogenomics of Dementia: Personalizing the Treatment of Cognitive and Neuropsychiatric Symptoms. Genes (Basel) 2023; 14:2048. [PMID: 38002991 PMCID: PMC10671071 DOI: 10.3390/genes14112048] [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: 10/10/2023] [Revised: 10/30/2023] [Accepted: 11/02/2023] [Indexed: 11/26/2023] Open
Abstract
Dementia is a syndrome of global and progressive deterioration of cognitive skills, especially memory, learning, abstract thinking, and orientation, usually affecting the elderly. The most common forms are Alzheimer's disease, vascular dementia, and other (frontotemporal, Lewy body disease) dementias. The etiology of these multifactorial disorders involves complex interactions of various environmental and (epi)genetic factors and requires multiple forms of pharmacological intervention, including anti-dementia drugs for cognitive impairment, antidepressants, antipsychotics, anxiolytics and sedatives for behavioral and psychological symptoms of dementia, and other drugs for comorbid disorders. The pharmacotherapy of dementia patients has been characterized by a significant interindividual variability in drug response and the development of adverse drug effects. The therapeutic response to currently available drugs is partially effective in only some individuals, with side effects, drug interactions, intolerance, and non-compliance occurring in the majority of dementia patients. Therefore, understanding the genetic basis of a patient's response to pharmacotherapy might help clinicians select the most effective treatment for dementia while minimizing the likelihood of adverse reactions and drug interactions. Recent advances in pharmacogenomics may contribute to the individualization and optimization of dementia pharmacotherapy by increasing its efficacy and safety via a prediction of clinical outcomes. Thus, it can significantly improve the quality of life in dementia patients.
Collapse
Affiliation(s)
- Barbara Vuic
- Laboratory for Molecular Neuropsychiatry, Division of Molecular Medicine, Rudjer Boskovic Institute, 10000 Zagreb, Croatia; (B.V.); (T.M.); (L.T.); (M.N.P.); (M.K.); (G.N.E.); (V.F.)
| | - Tina Milos
- Laboratory for Molecular Neuropsychiatry, Division of Molecular Medicine, Rudjer Boskovic Institute, 10000 Zagreb, Croatia; (B.V.); (T.M.); (L.T.); (M.N.P.); (M.K.); (G.N.E.); (V.F.)
| | - Lucija Tudor
- Laboratory for Molecular Neuropsychiatry, Division of Molecular Medicine, Rudjer Boskovic Institute, 10000 Zagreb, Croatia; (B.V.); (T.M.); (L.T.); (M.N.P.); (M.K.); (G.N.E.); (V.F.)
| | - Matea Nikolac Perkovic
- Laboratory for Molecular Neuropsychiatry, Division of Molecular Medicine, Rudjer Boskovic Institute, 10000 Zagreb, Croatia; (B.V.); (T.M.); (L.T.); (M.N.P.); (M.K.); (G.N.E.); (V.F.)
| | - Marcela Konjevod
- Laboratory for Molecular Neuropsychiatry, Division of Molecular Medicine, Rudjer Boskovic Institute, 10000 Zagreb, Croatia; (B.V.); (T.M.); (L.T.); (M.N.P.); (M.K.); (G.N.E.); (V.F.)
| | - Gordana Nedic Erjavec
- Laboratory for Molecular Neuropsychiatry, Division of Molecular Medicine, Rudjer Boskovic Institute, 10000 Zagreb, Croatia; (B.V.); (T.M.); (L.T.); (M.N.P.); (M.K.); (G.N.E.); (V.F.)
| | - Vladimir Farkas
- Laboratory for Molecular Neuropsychiatry, Division of Molecular Medicine, Rudjer Boskovic Institute, 10000 Zagreb, Croatia; (B.V.); (T.M.); (L.T.); (M.N.P.); (M.K.); (G.N.E.); (V.F.)
| | - Suzana Uzun
- Department for Biological Psychiatry and Psychogeriatry, University Hospital Vrapce, 10000 Zagreb, Croatia; (S.U.); (N.M.)
- School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
| | - Ninoslav Mimica
- Department for Biological Psychiatry and Psychogeriatry, University Hospital Vrapce, 10000 Zagreb, Croatia; (S.U.); (N.M.)
- School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
| | - Dubravka Svob Strac
- Laboratory for Molecular Neuropsychiatry, Division of Molecular Medicine, Rudjer Boskovic Institute, 10000 Zagreb, Croatia; (B.V.); (T.M.); (L.T.); (M.N.P.); (M.K.); (G.N.E.); (V.F.)
| |
Collapse
|
14
|
Niemis W, Peterson SR, Javier C, Nguyen A, Subiah S, Palmer RHC. On the utilization of the induced pluripotent stem cell (iPSC) model to study substance use disorders: A scoping review protocol. PLoS One 2023; 18:e0292238. [PMID: 37824561 PMCID: PMC10569547 DOI: 10.1371/journal.pone.0292238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2023] [Accepted: 09/13/2023] [Indexed: 10/14/2023] Open
Abstract
INTRODUCTION Induced pluripotent stem cells (iPSCs) are cells derived from somatic cells via reprogramming techniques. The iPSC approach has been increasingly used in neuropsychiatric research in the last decade. Though substance use disorders (SUDs) are a commonly occurring psychiatric disorder, the application of iPSC model in addiction research has been limited. No comprehensive review has been reported. We conducted a scoping review to collate existing evidence on the iPSC technologies applied to SUD research. We aim to identify current knowledge gaps and limitations in order to advance the use of iPSCs in the SUD field. METHODS AND ANALYSIS We employed a scoping review using the methodological framework first created by Arksey and O'Malley and further updated by Levac et al. and the Joanna Briggs Institute (JBI). We adopted the Preferred Reporting Items for Systematic reviews and Meta-Analyses extension for Protocols (PRISMA-P) to report items for the protocol. We searched evidence from four electronic databases: PubMed®, Embase®, Web of Science™, and Scopus®. Primary research, systematic reviews, and meta-analyses were included and limited to studies published in English, at the time from 2007 to March 2022. This is an "ongoing" scoping review. Searched studies will be independently screened, selected, and extracted by two reviewers. Disagreement will be solved by the third reviewer and discussion. Extracted data will be analyzed in descriptive and quantitative approaches, then summarized and presented in appropriate formats. Results will be reported following the Preferred Reporting Items for Systematic reviews and Meta-Analyses extension for Scoping Reviews (PRISMA-ScR) guideline and disseminated through a peer-reviewed publication and conference presentations. CONCLUSION To our best knowledge, this is the first comprehensive scoping review of iPSC methods specifically applied to a broad range of addictive drugs/substances that lead to SUDs or misuse behavior. REGISTRATION This protocol is registered on Zenodo repository (https://zenodo.org/) with doi:10.5281/zenodo.7915252.
Collapse
Affiliation(s)
- Wasiri Niemis
- Behavioral Genetics of Addiction Laboratory, Department of Psychology, Emory University, Atlanta, GA, United States of America
| | - Shenita R. Peterson
- Woodruff Health Sciences Center Library, Emory University, Atlanta, GA, United States of America
| | - Chrisabella Javier
- Behavioral Genetics of Addiction Laboratory, Department of Psychology, Emory University, Atlanta, GA, United States of America
| | - Amy Nguyen
- Behavioral Genetics of Addiction Laboratory, Department of Psychology, Emory University, Atlanta, GA, United States of America
| | - Sanchi Subiah
- Behavioral Genetics of Addiction Laboratory, Department of Psychology, Emory University, Atlanta, GA, United States of America
| | - Rohan H. C. Palmer
- Behavioral Genetics of Addiction Laboratory, Department of Psychology, Emory University, Atlanta, GA, United States of America
| |
Collapse
|
15
|
O'Connell KS, Koch E, Lenk HÇ, Akkouh IA, Hindley G, Jaholkowski P, Smith RL, Holen B, Shadrin AA, Frei O, Smeland OB, Steen NE, Dale AM, Molden E, Djurovic S, Andreassen OA. Polygenic overlap with body-mass index improves prediction of treatment-resistant schizophrenia. Psychiatry Res 2023; 325:115217. [PMID: 37146461 PMCID: PMC10788293 DOI: 10.1016/j.psychres.2023.115217] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 04/03/2023] [Accepted: 04/21/2023] [Indexed: 05/07/2023]
Abstract
Treatment resistant schizophrenia (TRS) is characterized by repeated treatment failure with antipsychotics. A recent genome-wide association study (GWAS) of TRS showed a polygenic architecture, but no significant loci were identified. Clozapine is shown to be the superior drug in terms of clinical effect in TRS; at the same time it has a serious side effect profile, including weight gain. Here, we sought to increase power for genetic discovery and improve polygenic prediction of TRS, by leveraging genetic overlap with Body Mass Index (BMI). We analysed GWAS summary statistics for TRS and BMI applying the conditional false discovery rate (cFDR) framework. We observed cross-trait polygenic enrichment for TRS conditioned on associations with BMI. Leveraging this cross-trait enrichment, we identified 2 novel loci for TRS at cFDR <0.01, suggesting a role of MAP2K1 and ZDBF2. Further, polygenic prediction based on the cFDR analysis explained more variance in TRS when compared to the standard TRS GWAS. These findings highlight putative molecular pathways which may distinguish TRS patients from treatment responsive patients. Moreover, these findings confirm that shared genetic mechanisms influence both TRS and BMI and provide new insights into the biological underpinnings of metabolic dysfunction and antipsychotic treatment.
Collapse
Affiliation(s)
- Kevin S O'Connell
- NORMENT, Centre for Mental Disorders Research, Division of Mental Health and Addiction, Oslo University Hospital, and Institute of Clinical Medicine, University of Oslo, Oslo, Norway.
| | - Elise Koch
- NORMENT, Centre for Mental Disorders Research, Division of Mental Health and Addiction, Oslo University Hospital, and Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Hasan Çağın Lenk
- Center for Psychopharmacology, Diakonhjemmet Hospital, Oslo, Norway; Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, University of Oslo, Oslo, Norway
| | - Ibrahim A Akkouh
- NORMENT, Centre for Mental Disorders Research, Division of Mental Health and Addiction, Oslo University Hospital, and Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Department of Medical Genetics, Oslo University Hospital, Oslo, Norway
| | - Guy Hindley
- NORMENT, Centre for Mental Disorders Research, Division of Mental Health and Addiction, Oslo University Hospital, and Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Psychosis Studies, Institute of Psychiatry, Psychology and Neurosciences, King's College London, United Kingdom
| | - Piotr Jaholkowski
- NORMENT, Centre for Mental Disorders Research, Division of Mental Health and Addiction, Oslo University Hospital, and Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Robert Løvsletten Smith
- NORMENT, Centre for Mental Disorders Research, Division of Mental Health and Addiction, Oslo University Hospital, and Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Center for Psychopharmacology, Diakonhjemmet Hospital, Oslo, Norway
| | - Børge Holen
- NORMENT, Centre for Mental Disorders Research, Division of Mental Health and Addiction, Oslo University Hospital, and Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Alexey A Shadrin
- NORMENT, Centre for Mental Disorders Research, Division of Mental Health and Addiction, Oslo University Hospital, and Institute of Clinical Medicine, University of Oslo, Oslo, Norway; KG Jebsen Centre for Neurodevelopmental disorders, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Oleksandr Frei
- NORMENT, Centre for Mental Disorders Research, Division of Mental Health and Addiction, Oslo University Hospital, and Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Center for Bioinformatics, Department of Informatics, University of Oslo, 0316 Oslo, Norway
| | - Olav B Smeland
- NORMENT, Centre for Mental Disorders Research, Division of Mental Health and Addiction, Oslo University Hospital, and Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Nils Eiel Steen
- NORMENT, Centre for Mental Disorders Research, Division of Mental Health and Addiction, Oslo University Hospital, and Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Anders M Dale
- Department of Radiology, University of California, San Diego, La Jolla, CA 92093, USA; Multimodal Imaging Laboratory, University of California San Diego, La Jolla, CA 92093, USA; Department of Psychiatry, University of California, San Diego, La Jolla, CA, USA; Department of Neurosciences, University of California San Diego, La Jolla, CA 92093, USA
| | - Espen Molden
- Center for Psychopharmacology, Diakonhjemmet Hospital, Oslo, Norway; Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, University of Oslo, Oslo, Norway
| | - Srdjan Djurovic
- Department of Medical Genetics, Oslo University Hospital, Oslo, Norway; NORMENT Centre, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Ole A Andreassen
- NORMENT, Centre for Mental Disorders Research, Division of Mental Health and Addiction, Oslo University Hospital, and Institute of Clinical Medicine, University of Oslo, Oslo, Norway; KG Jebsen Centre for Neurodevelopmental disorders, University of Oslo and Oslo University Hospital, Oslo, Norway.
| |
Collapse
|
16
|
Boissart C, Lasbareilles M, Tournois J, Chatrousse L, Poullion T, Benchoua A. Identification of signaling pathways modifying human dopaminergic neuron development using a pluripotent stem cell-based high-throughput screening automated system: purinergic pathways as a proof-of-principle. Front Pharmacol 2023; 14:1152180. [PMID: 37435497 PMCID: PMC10331426 DOI: 10.3389/fphar.2023.1152180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Accepted: 06/12/2023] [Indexed: 07/13/2023] Open
Abstract
Introduction: Alteration in the development, maturation, and projection of dopaminergic neurons has been proposed to be associated with several neurological and psychiatric disorders. Therefore, understanding the signals modulating the genesis of human dopaminergic neurons is crucial to elucidate disease etiology and develop effective countermeasures. Methods: In this study, we developed a screening model using human pluripotent stem cells to identify the modulators of dopaminergic neuron genesis. We set up a differentiation protocol to obtained floorplate midbrain progenitors competent to produce dopaminergic neurons and seeded them in a 384-well screening plate in a fully automated manner. Results and Discussion: These progenitors were treated with a collection of small molecules to identify the compounds increasing dopaminergic neuron production. As a proof-of-principle, we screened a library of compounds targeting purine- and adenosine-dependent pathways and identified an adenosine receptor 3 agonist as a candidate molecule to increase dopaminergic neuron production under physiological conditions and in cells invalidated for the HPRT1 gene. This screening model can provide important insights into the etiology of various diseases affecting the dopaminergic circuit development and plasticity and be used to identify therapeutic molecules for these diseases.
Collapse
Affiliation(s)
- Claire Boissart
- CECS, I-STEM, AFM, Neuroplasticity and Therapeutics, Corbeil-Essonnes, France
| | - Marie Lasbareilles
- CECS, I-STEM, AFM, Neuroplasticity and Therapeutics, Corbeil-Essonnes, France
- INSERM UMR 861, I-STEM, AFM, Corbeil-Essonnes, France
- UEVE UMR 861, I-STEM, AFM, Corbeil-Essonnes, France
| | - Johana Tournois
- CECS, I-STEM, AFM, Research and Technological Innovation, High Throughput Screening Plateform, Corbeil-Essonnes, France
| | - Laure Chatrousse
- CECS, I-STEM, AFM, Neuroplasticity and Therapeutics, Corbeil-Essonnes, France
| | - Thifaine Poullion
- CECS, I-STEM, AFM, Neuroplasticity and Therapeutics, Corbeil-Essonnes, France
| | - Alexandra Benchoua
- CECS, I-STEM, AFM, Neuroplasticity and Therapeutics, Corbeil-Essonnes, France
- CECS, I-STEM, AFM, Research and Technological Innovation, High Throughput Screening Plateform, Corbeil-Essonnes, France
| |
Collapse
|
17
|
Pardiñas AF, Kappel DB, Roberts M, Tipple F, Shitomi-Jones LM, King A, Jansen J, Helthuis M, Owen MJ, O'Donovan MC, Walters JTR. Pharmacokinetics and pharmacogenomics of clozapine in an ancestrally diverse sample: a longitudinal analysis and genome-wide association study using UK clinical monitoring data. Lancet Psychiatry 2023; 10:209-219. [PMID: 36804072 PMCID: PMC10824469 DOI: 10.1016/s2215-0366(23)00002-0] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 12/21/2022] [Accepted: 12/22/2022] [Indexed: 02/17/2023]
Abstract
BACKGROUND The antipsychotic, clozapine, is the only licensed drug against the treatment-resistant symptoms that affect 20-30% of people with schizophrenia. Clozapine is markedly underprescribed, partly because of concerns about its narrow therapeutic range and adverse drug reaction profile. Both concerns are linked to drug metabolism, which varies across populations globally and is partly genetically determined. Our study aimed to use a cross-ancestry genome-wide association study (GWAS) design to investigate variations in clozapine metabolism within and between genetically inferred ancestral backgrounds, to discover genomic associations to clozapine plasma concentrations, and to assess the effects of pharmacogenomic predictors across different ancestries. METHODS In this GWAS, we analysed data from the UK Zaponex Treatment Access System clozapine monitoring service as part of the CLOZUK study. We included all available individuals with clozapine pharmacokinetic assays requested by their clinicians. We excluded people younger than 18 years, or whose records contained clerical errors, or with blood drawn 6-24 h after dose, a clozapine or norclozapine concentration less than 50 ng/mL, a clozapine concentration of more than 2000 ng/mL, a clozapine-to-norclozapine ratio outside of the 0·5-3·0 interval, or a clozapine dose of more than 900 mg/day. Using genomic information, we identified five biogeographical ancestries: European, sub-Saharan African, north African, southwest Asian, and east Asian. We did pharmacokinetic modelling, a GWAS, and a polygenic risk score association analysis using longitudinal regression analysis with three primary outcome variables: two metabolite plasma concentrations (clozapine and norclozapine) and the clozapine-to-norclozapine ratio. FINDINGS 19 096 pharmacokinetic assays were available for 4760 individuals in the CLOZUK study. After data quality control, 4495 individuals (3268 [72·7%] male and 1227 [27·3%] female; mean age 42·19 years [range 18-85]) linked to 16 068 assays were included in this study. We found a faster average clozapine metabolism in people of sub-Saharan African ancestry than in those of European ancestry. By contrast, individuals with east Asian or southwest Asian ancestry were more likely to be slow clozapine metabolisers than those with European ancestry. Eight pharmacogenomic loci were identified in the GWAS, seven with significant effects in non-European groups. Polygenic scores generated from these loci were associated with clozapine outcome variables in the whole sample and within individual ancestries; the maximum variance explained was 7·26% for the metabolic ratio. INTERPRETATION Longitudinal cross-ancestry GWAS can discover pharmacogenomic markers of clozapine metabolism that, individually or as polygenic scores, have consistent effects across ancestries. Our findings suggest that ancestral differences in clozapine metabolism could be considered for optimising clozapine prescription protocols for diverse populations. FUNDING UK Academy of Medical Sciences, UK Medical Research Council, and European Commission.
Collapse
Affiliation(s)
- Antonio F Pardiñas
- MRC Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff, UK.
| | - Djenifer B Kappel
- MRC Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff, UK
| | - Milly Roberts
- MRC Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff, UK
| | - Francesca Tipple
- MRC Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff, UK
| | - Lisa M Shitomi-Jones
- MRC Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff, UK
| | | | | | | | - Michael J Owen
- MRC Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff, UK
| | - Michael C O'Donovan
- MRC Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff, UK
| | - James T R Walters
- MRC Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff, UK
| |
Collapse
|
18
|
Kappel DB, Legge SE, Hubbard L, Willcocks IR, O'Connell KS, Smith RL, Molden E, Andreassen OA, King A, Jansen J, Helthuis M, Owen MJ, O'Donovan MC, Walters JTR, Pardiñas AF. Genomic Stratification of Clozapine Prescription Patterns Using Schizophrenia Polygenic Scores. Biol Psychiatry 2023; 93:149-156. [PMID: 36244804 PMCID: PMC10804961 DOI: 10.1016/j.biopsych.2022.07.014] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 07/14/2022] [Accepted: 07/19/2022] [Indexed: 11/28/2022]
Abstract
BACKGROUND Treatment-resistant schizophrenia affects approximately 30% of individuals with the disorder. Clozapine is the medication of choice in treatment-resistant schizophrenia, but optimizing administration and dose titration is complex. The identification of factors influencing clozapine prescription and response, including genetics, is of interest in a precision psychiatry framework. METHODS We used linear regression models accounting for demographic, pharmacological, and clinical covariates to determine whether a polygenic risk score (PRS) for schizophrenia would be associated with the highest dose recorded during clozapine treatment. Analyses were performed across 2 independent multiancestry samples of individuals from a UK patient monitoring system, CLOZUK2 (n = 3133) and CLOZUK3 (n = 909), and a European sample from a Norwegian therapeutic drug monitoring service (n = 417). In a secondary analysis merging both UK cohorts, logistic regression models were used to estimate the relationship between schizophrenia PRSs and clozapine doses classified as low, standard, or high. RESULTS After controlling for relevant covariates, the schizophrenia PRS was correlated with the highest clozapine dose on record for each individual across all samples: CLOZUK2 (β = 12.22, SE = 3.78, p = .001), CLOZUK3 (β = 12.73, SE = 5.99, p = .034), and the Norwegian cohort (β = 46.45, SE = 18.83, p = .014). In a secondary analysis, the schizophrenia PRS was associated with taking clozapine doses >600 mg/day (odds ratio = 1.279, p = .006). CONCLUSIONS The schizophrenia PRS was associated with the highest clozapine dose prescribed for an individual in records from 3 independent samples, suggesting that the genetic liability for schizophrenia might index factors associated with therapeutic decisions in cohorts of patients with treatment-resistant schizophrenia.
Collapse
Affiliation(s)
- Djenifer B Kappel
- MRC Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Sophie E Legge
- MRC Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Leon Hubbard
- MRC Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Isabella R Willcocks
- MRC Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Kevin S O'Connell
- Norwegian Centre for Mental Disorders Research, Division of Mental Health and Addiction, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Robert L Smith
- Norwegian Centre for Mental Disorders Research, Division of Mental Health and Addiction, University of Oslo and Oslo University Hospital, Oslo, Norway; Center for Psychopharmacology, Diakonhjemmet Hospital, Oslo, Norway
| | - Espen Molden
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, University of Oslo, Oslo, Norway; Center for Psychopharmacology, Diakonhjemmet Hospital, Oslo, Norway
| | - Ole A Andreassen
- Norwegian Centre for Mental Disorders Research, Division of Mental Health and Addiction, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Adrian King
- Magna Laboratories Ltd., Ross-on-Wye, United Kingdom
| | | | | | - Michael J Owen
- MRC Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Michael C O'Donovan
- MRC Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - James T R Walters
- MRC Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Antonio F Pardiñas
- MRC Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff, United Kingdom.
| |
Collapse
|
19
|
Zhou Y, Koutsilieri S, Eliasson E, Lauschke VM. A paradigm shift in pharmacogenomics: From candidate polymorphisms to comprehensive sequencing. Basic Clin Pharmacol Toxicol 2022; 131:452-464. [PMID: 35971800 PMCID: PMC9805052 DOI: 10.1111/bcpt.13779] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Revised: 07/28/2022] [Accepted: 08/09/2022] [Indexed: 01/09/2023]
Abstract
Genetic factors have long been recognized as important determinants of interindividual variability in drug efficacy and toxicity. However, despite the increasing number of established gene-drug associations, candidate polymorphisms can only explain a fraction of the genetically encoded functional variability in drug disposition. Advancements in genetic profiling methods now allow to analyse the landscape of human pharmacogenetic variations comprehensively, which opens new opportunities to identify novel factors that could explain the "missing heritability." Here, we provide an updated overview of the landscape of pharmacogenomic variability based on recent analyses of population-scale sequencing projects. We then summarize the current state-of-the-art how the functional consequences of variants with unknown effects can be quantitatively estimated while discussing challenges and peculiarities that are specific to pharmacogenes. In the last sections, we discuss the importance of considering ethnogeographic diversity to provide equitable benefits of pharmacogenomics and summarize current roadblocks for the implementation of sequencing-based guidance of clinical decision-making. Based on the current state of the field, we conclude that testing is likely to gradually shift from the interrogation of selected candidate polymorphisms to comprehensive sequencing, which allows to consider the full spectrum of pharmacogenomic variations for a true personalization of genomic prescribing.
Collapse
Affiliation(s)
- Yitian Zhou
- Department of Physiology and PharmacologyKarolinska InstitutetStockholmSweden,Department of Laboratory MedicineKarolinska InstitutetStockholmSweden
| | | | - Erik Eliasson
- Department of Laboratory MedicineKarolinska InstitutetStockholmSweden
| | - Volker M. Lauschke
- Department of Physiology and PharmacologyKarolinska InstitutetStockholmSweden,Dr Margarete Fischer‐Bosch Institute of Clinical PharmacologyStuttgartGermany,University of TübingenTübingenGermany
| |
Collapse
|
20
|
Advances in Pharmacy Practice: A Look towards the Future. PHARMACY 2022; 10:pharmacy10050125. [PMID: 36287446 PMCID: PMC9608826 DOI: 10.3390/pharmacy10050125] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 09/21/2022] [Accepted: 09/21/2022] [Indexed: 11/07/2022] Open
Abstract
This review looks at the factors that may influence practice in the future. Transformation could occur at 3 levels. Firstly, the traditional profession of the pharmacist as a dispenser of medicines is expanding. Secondly, the pharmacist’s activities are progressing into new healthcare fields. Thirdly, other changes are stimulated by global developments. This review may be helpful for pharmacy and healthcare leaders looking at the future configuration and aims of their pharmacy services.
Collapse
|
21
|
Baune BT. Clinical use of pharmacogenomics in Psychiatry: The future has not yet arrived. Eur Neuropsychopharmacol 2022; 58:4-6. [PMID: 35093788 DOI: 10.1016/j.euroneuro.2022.01.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 01/11/2022] [Accepted: 01/14/2022] [Indexed: 11/04/2022]
Affiliation(s)
- B T Baune
- Department of Psychiatry, University of Münster, Münster, Germany; Department of Psychiatry, Melbourne Medical School, The University of Melbourne, Melbourne, Australia; The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC, Australia.
| |
Collapse
|
22
|
Gill PS, Elchynski AL, Porter-Gill PA, Goodson BG, Scott MA, Lipinski D, Seay A, Kehn C, Balmakund T, Schaefer GB. Multidisciplinary Consulting Team for Complicated Cases of Neurodevelopmental and Neurobehavioral Disorders: Assessing the Opportunities and Challenges of Integrating Pharmacogenomics into a Team Setting. J Pers Med 2022; 12:jpm12040599. [PMID: 35455715 PMCID: PMC9024886 DOI: 10.3390/jpm12040599] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 03/16/2022] [Accepted: 04/06/2022] [Indexed: 12/15/2022] Open
Abstract
Neurodevelopmental disorders have steadily increased in incidence in the United States. Over the past decade, there have been significant changes in clinical diagnoses and treatments some of which are due to the increasing adoption of pharmacogenomics (PGx) by clinicians. In this pilot study, a multidisciplinary team at the Arkansas Children’s Hospital North West consulted on 27 patients referred for difficult-to-manage neurodevelopmental and/or neurobehavioral disorders. The 27 patients were evaluated by the team using records review, team discussion, and pharmacogenetic testing. OneOme RightMed® (Minneapolis, MN, USA) and the Arkansas Children’s Hospital comprehensive PGx test were used for drug prescribing guidance. Of the 27 patients’ predicted phenotypes, the normal metabolizer was 11 (40.8%) for CYP2C19 and 16 (59.3%) for CYP2D6. For the neurodevelopmental disorders, the most common comorbid conditions included attention-deficit hyperactivity disorder (66.7%), anxiety disorder (59.3%), and autism (40.7%). Following the team assessment and PGx testing, 66.7% of the patients had actionable medication recommendations. This included continuing current therapy, suggesting an appropriate alternative medication, starting a new therapy, or adding adjunct therapy (based on their current medication use). Moreover, 25.9% of patients phenoconverted to a CYP2D6 poor metabolizer. This retrospective chart review pilot study highlights the value of a multidisciplinary treatment approach to deliver precision healthcare by improving physician clinical decisions and potentially impacting patient outcomes. It also shows the feasibility to implement PGx testing in neurodevelopmental/neurobehavioral disorders.
Collapse
Affiliation(s)
- Pritmohinder S. Gill
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR 72202, USA; (T.B.); (G.B.S.)
- Arkansas Children’s Research Institute, Little Rock, AR 72202, USA;
- Correspondence: ; Tel.: +1-(501)-364-1418; Fax: +1-(501)-364-3654
| | | | | | - Bradley G. Goodson
- Schmieding Developmental Center, Springdale, AR 72762, USA; (B.G.G.); (M.A.S.); (D.L.); (A.S.); (C.K.)
| | - Mary Ann Scott
- Schmieding Developmental Center, Springdale, AR 72762, USA; (B.G.G.); (M.A.S.); (D.L.); (A.S.); (C.K.)
| | - Damon Lipinski
- Schmieding Developmental Center, Springdale, AR 72762, USA; (B.G.G.); (M.A.S.); (D.L.); (A.S.); (C.K.)
| | - Amy Seay
- Schmieding Developmental Center, Springdale, AR 72762, USA; (B.G.G.); (M.A.S.); (D.L.); (A.S.); (C.K.)
- Arkansas Children’s Hospital Northwest, Springdale, AR 72762, USA
| | - Christina Kehn
- Schmieding Developmental Center, Springdale, AR 72762, USA; (B.G.G.); (M.A.S.); (D.L.); (A.S.); (C.K.)
| | - Tonya Balmakund
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR 72202, USA; (T.B.); (G.B.S.)
- Arkansas Children’s Hospital Northwest, Springdale, AR 72762, USA
| | - G. Bradley Schaefer
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR 72202, USA; (T.B.); (G.B.S.)
- Arkansas Children’s Research Institute, Little Rock, AR 72202, USA;
- Schmieding Developmental Center, Springdale, AR 72762, USA; (B.G.G.); (M.A.S.); (D.L.); (A.S.); (C.K.)
- University of Arkansas for Medical Sciences Northwest, Fayetteville, AR 72701, USA
| |
Collapse
|
23
|
Pardiñas AF, Smart SE, Willcocks IR, Holmans PA, Dennison CA, Lynham AJ, Legge SE, Baune BT, Bigdeli TB, Cairns MJ, Corvin A, Fanous AH, Frank J, Kelly B, McQuillin A, Melle I, Mortensen PB, Mowry BJ, Pato CN, Periyasamy S, Rietschel M, Rujescu D, Simonsen C, St Clair D, Tooney P, Wu JQ, Andreassen OA, Kowalec K, Sullivan PF, Murray RM, Owen MJ, MacCabe JH, O’Donovan MC, Walters JTR, and the Genetics Workstream of the Schizophrenia Treatment Resistance and Therapeutic Advances (STRATA) Consortium and the Schizophrenia Working Group of the Psychiatric Genomics Consortium (PGC), Ajnakina O, Alameda L, Barnes TRE, Berardi D, Bonora E, Camporesi S, Cleusix M, Conus P, Crespo-Facorro B, D'Andrea G, Demjaha A, Do KQ, Doody GA, Eap CB, Ferchiou A, Di Forti M, Guidi L, Homman L, Jenni R, Joyce EM, Kassoumeri L, Khadimallah I, Lastrina O, Muratori R, Noyan H, O'Neill FA, Pignon B, Restellini R, Richard JR, Schürhoff F, Španiel F, Szöke A, Tarricone I, Tortelli A, Üçok A, Vázquez-Bourgon J. Interaction Testing and Polygenic Risk Scoring to Estimate the Association of Common Genetic Variants With Treatment Resistance in Schizophrenia. JAMA Psychiatry 2022; 79:260-269. [PMID: 35019943 PMCID: PMC8756361 DOI: 10.1001/jamapsychiatry.2021.3799] [Citation(s) in RCA: 55] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
IMPORTANCE About 20% to 30% of people with schizophrenia have psychotic symptoms that do not respond adequately to first-line antipsychotic treatment. This clinical presentation, chronic and highly disabling, is known as treatment-resistant schizophrenia (TRS). The causes of treatment resistance and their relationships with causes underlying schizophrenia are largely unknown. Adequately powered genetic studies of TRS are scarce because of the difficulty in collecting data from well-characterized TRS cohorts. OBJECTIVE To examine the genetic architecture of TRS through the reassessment of genetic data from schizophrenia studies and its validation in carefully ascertained clinical samples. DESIGN, SETTING, AND PARTICIPANTS Two case-control genome-wide association studies (GWASs) of schizophrenia were performed in which the case samples were defined as individuals with TRS (n = 10 501) and individuals with non-TRS (n = 20 325). The differences in effect sizes for allelic associations were then determined between both studies, the reasoning being such differences reflect treatment resistance instead of schizophrenia. Genotype data were retrieved from the CLOZUK and Psychiatric Genomics Consortium (PGC) schizophrenia studies. The output was validated using polygenic risk score (PRS) profiling of 2 independent schizophrenia cohorts with TRS and non-TRS: a prevalence sample with 817 individuals (Cardiff Cognition in Schizophrenia [CardiffCOGS]) and an incidence sample with 563 individuals (Genetics Workstream of the Schizophrenia Treatment Resistance and Therapeutic Advances [STRATA-G]). MAIN OUTCOMES AND MEASURES GWAS of treatment resistance in schizophrenia. The results of the GWAS were compared with complex polygenic traits through a genetic correlation approach and were used for PRS analysis on the independent validation cohorts using the same TRS definition. RESULTS The study included a total of 85 490 participants (48 635 [56.9%] male) in its GWAS stage and 1380 participants (859 [62.2%] male) in its PRS validation stage. Treatment resistance in schizophrenia emerged as a polygenic trait with detectable heritability (1% to 4%), and several traits related to intelligence and cognition were found to be genetically correlated with it (genetic correlation, 0.41-0.69). PRS analysis in the CardiffCOGS prevalence sample showed a positive association between TRS and a history of taking clozapine (r2 = 2.03%; P = .001), which was replicated in the STRATA-G incidence sample (r2 = 1.09%; P = .04). CONCLUSIONS AND RELEVANCE In this GWAS, common genetic variants were differentially associated with TRS, and these associations may have been obscured through the amalgamation of large GWAS samples in previous studies of broadly defined schizophrenia. Findings of this study suggest the validity of meta-analytic approaches for studies on patient outcomes, including treatment resistance.
Collapse
Affiliation(s)
- Antonio F. Pardiñas
- MRC Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Sophie E. Smart
- MRC Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff, United Kingdom,Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London, United Kingdom
| | - Isabella R. Willcocks
- MRC Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Peter A. Holmans
- MRC Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Charlotte A. Dennison
- MRC Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Amy J. Lynham
- MRC Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Sophie E. Legge
- MRC Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Bernhard T. Baune
- Department of Psychiatry, University of Münster, Münster, Germany,Department of Psychiatry, Melbourne Medical School, The University of Melbourne, Melbourne, Australia,The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Melbourne, Australia
| | - Tim B. Bigdeli
- Department of Psychiatry and the Behavioral Sciences, State University of New York Downstate Medical Center, Brooklyn,Institute for Genomic Health, State University of New York Downstate Medical Center, Brooklyn,Department of Psychiatry, Veterans Affairs New York Harbor Healthcare System, Brooklyn
| | - Murray J. Cairns
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Newcastle, Australia,Centre for Brain and Mental Health Research, University of Newcastle, Newcastle, Australia,Hunter Medical Research Institute, Newcastle, Australia
| | - Aiden Corvin
- Neuropsychiatric Genetics Research Group, Department of Psychiatry, Trinity College Dublin, Dublin, Ireland
| | - Ayman H. Fanous
- Department of Psychiatry and the Behavioral Sciences, State University of New York Downstate Medical Center, Brooklyn,Institute for Genomic Health, State University of New York Downstate Medical Center, Brooklyn
| | - Josef Frank
- Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Heidelberg, Mannheim, Germany
| | - Brian Kelly
- School of Medicine & Public Health, The University of Newcastle, Newcastle, Australia
| | - Andrew McQuillin
- Molecular Psychiatry Laboratory, Division of Psychiatry, University College London, London, United Kingdom
| | - Ingrid Melle
- Norwegian Centre for Mental Disorders Research, Institute of Clinical Medicine, University of Oslo, Oslo, Norway,Division of Mental Health and Addiction, Institute of Clinical Medicine, Oslo University Hospital, Oslo, Norway
| | - Preben B. Mortensen
- National Centre for Register-based Research, Aarhus University, Aarhus, Denmark,The Lundbeck Foundation Initiative for Integrative Psychiatric Research, Aarhus, Denmark
| | - Bryan J. Mowry
- Queensland Brain Institute, The University of Queensland, Brisbane, Australia,Queensland Centre for Mental Health Research, The University of Queensland, Brisbane, Australia
| | - Carlos N. Pato
- Department of Psychiatry and Behavioral Sciences, State University of New York Downstate Medical Center, Brooklyn,Department of Psychiatry and Zilkha Neurogenetics Institute, Keck School of Medicine, University of Southern California, Los Angeles,Institute for Genomic Health, State University of New York Downstate Medical Center, Brooklyn
| | - Sathish Periyasamy
- Queensland Brain Institute, The University of Queensland, Brisbane, Australia,Queensland Centre for Mental Health Research, The University of Queensland, Brisbane, Australia
| | - Marcella Rietschel
- Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Heidelberg, Mannheim, Germany
| | - Dan Rujescu
- University Clinic and Outpatient Clinic for Psychiatry, Psychotherapy and Psychosomatics, Martin Luther University of Halle-Wittenberg, Halle, Germany,Division of General Psychiatry, Department of Psychiatry and Psychotherapy, Medical University of Vienna, Vienna, Austria
| | - Carmen Simonsen
- Norwegian Centre for Mental Disorders Research, Institute of Clinical Medicine, University of Oslo, Oslo, Norway,Early Intervention in Psychosis Advisory Unit for South-East Norway, Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
| | - David St Clair
- Institute of Medical Sciences, University of Aberdeen, Aberdeen, United Kingdom
| | - Paul Tooney
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Newcastle, Australia,Hunter Medical Research Institute, Newcastle, Australia
| | - Jing Qin Wu
- Baker Heart and Diabetes Institute, Melbourne, Australia
| | - Ole A. Andreassen
- Norwegian Centre for Mental Disorders Research, Institute of Clinical Medicine, University of Oslo, Oslo, Norway,Division of Mental Health and Addiction, Institute of Clinical Medicine, Oslo University Hospital, Oslo, Norway
| | - Kaarina Kowalec
- College of Pharmacy, University of Manitoba, Winnipeg, Manitoba, Canada,Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Patrick F. Sullivan
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden,Department of Psychiatry, Icahn School of Medicine, Mount Sinai Hospital, New York, New York,Department of Genetics, University of North Carolina, Chapel Hill
| | - Robin M. Murray
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London, United Kingdom
| | - Michael J. Owen
- MRC Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - James H. MacCabe
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London, United Kingdom
| | - Michael C. O’Donovan
- MRC Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - James T. R. Walters
- MRC Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | | | - Olesya Ajnakina
- Department of Biostatistics & Health Informatics, Institute of Psychiatry, Psychology & Neuroscience, King's College London, University of London, London, United Kingdom.,Department of Behavioural Science and Health, Institute of Epidemiology and Health Care, University College London, London, United Kingdom
| | - Luis Alameda
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, United Kingdom.,Centro de Investigacion Biomedica en Red de Salud Mental, Spanish Network for Research in Mental Health, Sevilla, Spain.,Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocio, Departamento de Psiquiatria, Universidad de Sevilla, Sevilla, Spain.,Treatment and Early Intervention in Psychosis Program, Service of General Psychiatry, Department of Psychiatry, Lausanne University Hospital, Lausanne, Switzerland
| | - Thomas R E Barnes
- Division of Psychiatry, Imperial College London, London, United Kingdom
| | - Domenico Berardi
- Department of Biomedical and Neuro-motor Sciences, Psychiatry Unit, Alma Mater Studiorum Università di Bologna, Bologna, Italy
| | - Elena Bonora
- Department of Medical and Surgical Sciences, Bologna Transcultural Psychosomatic Team, Alma Mater Studiorum, University of Bologna, Bologna, Italy
| | - Sara Camporesi
- Treatment and Early Intervention in Psychosis Program, Service of General Psychiatry, Department of Psychiatry, Lausanne University Hospital, Lausanne, Switzerland.,Unit for Research in Schizophrenia, Center for Psychiatric Neuroscience, Department of Psychiatry, Lausanne University Hospital, Lausanne, Switzerland
| | - Martine Cleusix
- Unit for Research in Schizophrenia, Center for Psychiatric Neuroscience, Department of Psychiatry, Lausanne University Hospital, Lausanne, Switzerland
| | - Philippe Conus
- Treatment and Early Intervention in Psychosis Program, Service of General Psychiatry, Department of Psychiatry, Lausanne University Hospital, Lausanne, Switzerland
| | - Benedicto Crespo-Facorro
- Centro de Investigacion Biomedica en Red de Salud Mental, Spanish Network for Research in Mental Health, Sevilla, Spain.,Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocio, Departamento de Psiquiatria, Universidad de Sevilla, Sevilla, Spain
| | - Giuseppe D'Andrea
- Department of Biomedical and Neuro-motor Sciences, Psychiatry Unit, Alma Mater Studiorum Università di Bologna, Bologna, Italy
| | - Arsime Demjaha
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, United Kingdom
| | - Kim Q Do
- Unit for Research in Schizophrenia, Center for Psychiatric Neuroscience, Department of Psychiatry, Lausanne University Hospital, Lausanne, Switzerland
| | - Gillian A Doody
- Department of Medical Education, University of Nottingham Faculty of Medicine and Health Sciences, Nottingham, United Kingdom
| | - Chin B Eap
- Unit of Pharmacogenetics and Clinical Psychopharmacology, Centre for Psychiatric Neuroscience, Department of Psychiatry, Lausanne University Hospital, University of Lausanne, Prilly, Switzerland.,School of Pharmaceutical Sciences, University of Geneva, Geneva, Switzerland.,Center for Research and Innovation in Clinical Pharmaceutical Sciences, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland.,Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, Geneva, Switzerland
| | - Aziz Ferchiou
- University Paris-Est Créteil, Institut national de la santé et de la recherche médicale, Mondor Institute for Biomedical Research, Translational Neuropsychiatry, Fondation FondaMental, Créteil, France
| | - Marta Di Forti
- Social Genetics and Developmental Psychiatry, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, United Kingdom.,South London and Maudsley National Health Service Mental Health Foundation Trust, London, United Kingdom
| | - Lorenzo Guidi
- Department of Medical and Surgical Sciences, Bologna Transcultural Psychosomatic Team, Alma Mater Studiorum, University of Bologna, Bologna, Italy
| | - Lina Homman
- Department of Social and Welfare Studies, Department of Behavioural Sciences and Learning, Linköping University, Linköping, Sweden.,Centre For Public Health, Institute Of Clinical Sciences, Queens University Belfast, Belfast, United Kingdom
| | - Raoul Jenni
- Unit for Research in Schizophrenia, Center for Psychiatric Neuroscience, Department of Psychiatry, Lausanne University Hospital, Lausanne, Switzerland
| | - Eileen M Joyce
- UCL Queen Square Institute of Neurology, University College London, London, United Kingdom
| | - Laura Kassoumeri
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, United Kingdom
| | - Inès Khadimallah
- Unit for Research in Schizophrenia, Center for Psychiatric Neuroscience, Department of Psychiatry, Lausanne University Hospital, Lausanne, Switzerland
| | - Ornella Lastrina
- Department of Biomedical and Neuro-motor Sciences, Psychiatry Unit, Alma Mater Studiorum Università di Bologna, Bologna, Italy
| | - Roberto Muratori
- Department of Medical and Surgical Sciences, Bologna Transcultural Psychosomatic Team, Alma Mater Studiorum, University of Bologna, Bologna, Italy
| | - Handan Noyan
- Faculty of Social Sciences, Department of Psychology, Beykoz University, Istanbul, Turkey
| | - Francis A O'Neill
- Centre For Public Health, Institute Of Clinical Sciences, Queens University Belfast, Belfast, United Kingdom
| | - Baptiste Pignon
- University Paris-Est Créteil, Institut national de la santé et de la recherche médicale, Mondor Institute for Biomedical Research, Translational Neuropsychiatry, Fondation FondaMental, Créteil, France.,Assistance Publique-Hôpitaux de Paris, Hôpitaux Universitaires HMondor, Département Médico-Universitaire de Psychiatrie et d'Addictologie, Fédération Hospitalo-Universitaire de Médecine de Précision, Créteil, France
| | - Romeo Restellini
- Treatment and Early Intervention in Psychosis Program, Service of General Psychiatry, Department of Psychiatry, Lausanne University Hospital, Lausanne, Switzerland.,Unit for Research in Schizophrenia, Center for Psychiatric Neuroscience, Department of Psychiatry, Lausanne University Hospital, Lausanne, Switzerland
| | - Jean-Romain Richard
- University Paris-Est Créteil, Institut national de la santé et de la recherche médicale, Mondor Institute for Biomedical Research, Translational Neuropsychiatry, Fondation FondaMental, Créteil, France
| | - Franck Schürhoff
- University Paris-Est Créteil, Institut national de la santé et de la recherche médicale, Mondor Institute for Biomedical Research, Translational Neuropsychiatry, Fondation FondaMental, Créteil, France.,Assistance Publique-Hôpitaux de Paris, Hôpitaux Universitaires HMondor, Département Médico-Universitaire de Psychiatrie et d'Addictologie, Fédération Hospitalo-Universitaire de Médecine de Précision, Créteil, France
| | - Filip Španiel
- Department of Applied Neuroscience and Neuroimaging, National Institute of Mental Health, Klecany, Czechia.,Department of Psychiatry and Medical Psychology, Third Faculty of Medicine, Charles University, Prague, Czechia
| | - Andrei Szöke
- University Paris-Est Créteil, Institut national de la santé et de la recherche médicale, Mondor Institute for Biomedical Research, Translational Neuropsychiatry, Fondation FondaMental, Créteil, France.,Assistance Publique-Hôpitaux de Paris, Hôpitaux Universitaires HMondor, Département Médico-Universitaire de Psychiatrie et d'Addictologie, Fédération Hospitalo-Universitaire de Médecine de Précision, Créteil, France
| | - Ilaria Tarricone
- Department of Medical and Surgical Sciences, Bologna Transcultural Psychosomatic Team, Alma Mater Studiorum, University of Bologna, Bologna, Italy
| | - Andrea Tortelli
- University Paris-Est Créteil, Institut national de la santé et de la recherche médicale, Mondor Institute for Biomedical Research, Translational Neuropsychiatry, Fondation FondaMental, Créteil, France.,Groupe Hospitalier Universitaire Psychiatrie Neurosciences Paris, Pôle Psychiatrie Précarité, Paris, France
| | - Alp Üçok
- Department of Psychiatry, Istanbul University, Istanbul, Turkey
| | - Javier Vázquez-Bourgon
- Department of Psychiatry, University Hospital Marques de Valdecilla-Instituto de Investigación Marques de Valdecilla, Santander, Spain.,Department of Medicine and Psychiatry, School of Medicine, University of Cantabria, Santander, Spain.,Centro de Investigacion Biomedica en Red de Salud Mental, Spanish Network for Research in Mental Health, Santander, Spain
| |
Collapse
|