1
|
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
|
2
|
Koch E, Kämpe A, Alver M, Sigurðarson S, Einarsson G, Partanen J, Smith RL, Jaholkowski P, Taipale H, Lähteenvuo M, Steen NE, Smeland OB, Djurovic S, Molden E, Sigurdsson E, Stefánsson H, Stefánsson K, Palotie A, Milani L, O'Connell KS, Andreassen OA. Polygenic liability for antipsychotic dosage and polypharmacy - a real-world registry and biobank study. Neuropsychopharmacology 2024; 49:1113-1119. [PMID: 38184734 PMCID: PMC11109158 DOI: 10.1038/s41386-023-01792-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 11/10/2023] [Accepted: 12/19/2023] [Indexed: 01/08/2024]
Abstract
Genomic prediction of antipsychotic dose and polypharmacy has been difficult, mainly due to limited access to large cohorts with genetic and drug prescription data. In this proof of principle study, we investigated if genetic liability for schizophrenia is associated with high dose requirements of antipsychotics and antipsychotic polypharmacy, using real-world registry and biobank data from five independent Nordic cohorts of a total of N = 21,572 individuals with psychotic disorders (schizophrenia, bipolar disorder, and other psychosis). Within regression models, a polygenic risk score (PRS) for schizophrenia was studied in relation to standardized antipsychotic dose as well as antipsychotic polypharmacy, defined based on longitudinal prescription registry data as well as health records and self-reported data. Meta-analyses across the five cohorts showed that PRS for schizophrenia was significantly positively associated with prescribed (standardized) antipsychotic dose (beta(SE) = 0.0435(0.009), p = 0.0006) and antipsychotic polypharmacy defined as taking ≥2 antipsychotics (OR = 1.10, CI = 1.05-1.21, p = 0.0073). The direction of effect was similar in all five independent cohorts. These findings indicate that genotypes may aid clinically relevant decisions on individual patients´ antipsychotic treatment. Further, the findings illustrate how real-world data have the potential to generate results needed for future precision medicine approaches in psychiatry.
Collapse
Affiliation(s)
- 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.
| | - Anders Kämpe
- Institute for Molecular Medicine, Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland
| | - Maris Alver
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | | | | | - Juulia Partanen
- Institute for Molecular Medicine, Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland
| | - Robert L 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
| | - 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
| | - Heidi Taipale
- Niuvanniemi Hospital, Kuopio, Finland
- Department of Clinical Neuroscience, Division of Insurance Medicine, Karolinska Institutet, Stockholm, Sweden
- School of Pharmacy, University of Eastern Finland, Kuopio, Finland
| | | | - 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
| | - 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
| | - Srdjan Djurovic
- Department of Medical Genetics, Oslo University Hospital, Oslo, Norway
- NORMENT Centre, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Espen Molden
- Center for Psychopharmacology, Diakonhjemmet Hospital, Oslo, Norway
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, University of Oslo, Oslo, Norway
| | - Engilbert Sigurdsson
- Faculty of Medicine, University of Iceland and Department of Psychiatry, Landspitali, National University Hospital, Reykjavík, Iceland
| | | | | | - Aarno Palotie
- Institute for Molecular Medicine, Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland
| | - Lili Milani
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
- Genetics and Personalized Medicine Clinic, Tartu University Hospital, Tartu, Estonia
| | - 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
| | - 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
|
3
|
Zhai S, Mehrotra DV, Shen J. Applying polygenic risk score methods to pharmacogenomics GWAS: challenges and opportunities. Brief Bioinform 2023; 25:bbad470. [PMID: 38152980 PMCID: PMC10782924 DOI: 10.1093/bib/bbad470] [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: 07/14/2023] [Revised: 11/20/2023] [Accepted: 11/28/2023] [Indexed: 12/29/2023] Open
Abstract
Polygenic risk scores (PRSs) have emerged as promising tools for the prediction of human diseases and complex traits in disease genome-wide association studies (GWAS). Applying PRSs to pharmacogenomics (PGx) studies has begun to show great potential for improving patient stratification and drug response prediction. However, there are unique challenges that arise when applying PRSs to PGx GWAS beyond those typically encountered in disease GWAS (e.g. Eurocentric or trans-ethnic bias). These challenges include: (i) the lack of knowledge about whether PGx or disease GWAS/variants should be used in the base cohort (BC); (ii) the small sample sizes in PGx GWAS with corresponding low power and (iii) the more complex PRS statistical modeling required for handling both prognostic and predictive effects simultaneously. To gain insights in this landscape about the general trends, challenges and possible solutions, we first conduct a systematic review of both PRS applications and PRS method development in PGx GWAS. To further address the challenges, we propose (i) a novel PRS application strategy by leveraging both PGx and disease GWAS summary statistics in the BC for PRS construction and (ii) a new Bayesian method (PRS-PGx-Bayesx) to reduce Eurocentric or cross-population PRS prediction bias. Extensive simulations are conducted to demonstrate their advantages over existing PRS methods applied in PGx GWAS. Our systematic review and methodology research work not only highlights current gaps and key considerations while applying PRS methods to PGx GWAS, but also provides possible solutions for better PGx PRS applications and future research.
Collapse
Affiliation(s)
- Song Zhai
- Biostatistics and Research Decision Sciences, Merck & Co., Inc., Rahway, NJ 07065, USA
| | - Devan V Mehrotra
- Biostatistics and Research Decision Sciences, Merck & Co., Inc., North Wales, PA 19454, USA
| | - Judong Shen
- Biostatistics and Research Decision Sciences, Merck & Co., Inc., Rahway, NJ 07065, USA
| |
Collapse
|
4
|
Sandhu AK, Naderi E, Wijninga MJ, Liemburg EJ, Cath D, Bruggeman R, Alizadeh BZ. Pharmacogenetics of Long-Term Outcomes of Schizophrenia Spectrum Disorders: The Functional Role of CYP2D6 and CYP2C19. J Pers Med 2023; 13:1354. [PMID: 37763122 PMCID: PMC10532576 DOI: 10.3390/jpm13091354] [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: 07/27/2023] [Revised: 08/25/2023] [Accepted: 08/27/2023] [Indexed: 09/29/2023] Open
Abstract
Schizophrenia spectrum disorders (SSD) are complex mental disorders, and while treatment with antipsychotics is important, many patients do not respond or develop serious side effects. Genetic variation has been shown to play a considerable role in determining an individual's response to antipsychotic medication. However, previous pharmacogenetic (PGx) studies have been limited by small sample sizes, lack of consensus regarding relevant genetic variants, and cross-sectional designs. The current study aimed to investigate the association between PGx variants and long-term clinical outcomes in 691 patients of European ancestry with SSD. Using evidence from the literature on candidate genes involved in antipsychotic pharmacodynamics, we created a polygenic risk score (PRS) to investigate its association with clinical outcomes. We also created PRS using core variants of psychotropic drug metabolism enzymes CYP2D6 and CYP2C19. Furthermore, the CYP2D6 and CYP2C19 functional activity scores were calculated to determine the relationship between metabolism and clinical outcomes. We found no association for PGx PRSs and clinical outcomes; however, an association was found with CYP2D6 activity scores by the traditional method. Higher CYP2D6 metabolism was associated with high positive and high cognitive impairment groups relative to low symptom severity groups. These findings highlight the need to test PGx efficacy with different symptom domains. More evidence is needed before pharmacogenetic variation can contribute to personalized treatment plans.
Collapse
Affiliation(s)
- Amrit K. Sandhu
- Department of Epidemiology, University Medical Centre Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands
| | - Elnaz Naderi
- Department of Epidemiology, University Medical Centre Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands
- Centre for Statistical Genetics, Gertude H. Sergiesky Centre, Department of Neurology, Columbia University Medical Centre, New York, NY 10032, USA
| | - Morenika J. Wijninga
- Department of Epidemiology, University Medical Centre Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands
| | - Edith J. Liemburg
- Department of Psychiatry, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands
| | | | - Danielle Cath
- Department of Psychiatry, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands
- GGZ Drenthe, Department of Specialist Trainings, 9704 LA Assen, The Netherlands
| | - Richard Bruggeman
- Department of Psychiatry, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands
| | - Behrooz Z. Alizadeh
- Department of Epidemiology, University Medical Centre Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands
- Department of Psychiatry, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands
| |
Collapse
|
5
|
Vasiliu O. The pharmacogenetics of the new-generation antipsychotics - A scoping review focused on patients with severe psychiatric disorders. Front Psychiatry 2023; 14:1124796. [PMID: 36873203 PMCID: PMC9978195 DOI: 10.3389/fpsyt.2023.1124796] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 01/30/2023] [Indexed: 02/18/2023] Open
Abstract
Exploring the possible correlations between gene variations and the clinical effects of the new-generation antipsychotics is considered essential in the framework of personalized medicine. It is expected that pharmacogenetic data will be useful for increasing the treatment efficacy, tolerability, therapeutic adherence, functional recovery, and quality of life in patients with severe psychiatric disorders (SPD). This scoping review investigated the available evidence about the pharmacokinetics, pharmacodynamics, and pharmacogenetics of five new-generation antipsychotics, i.e., cariprazine, brexpiprazole, aripiprazole, lumateperone, and pimavanserin. Based on the analysis of 25 primary and secondary sources and the review of these agents' summaries of product characteristics, aripiprazole benefits from the most relevant data about the impact of gene variability on its pharmacokinetics and pharmacodynamics, with significant consequences on this antipsychotic's efficacy and tolerability. The determination of the CYP2D6 metabolizer status is important when administering aripiprazole, either as monotherapy or associated with other pharmacological agents. Allelic variability in genes encoding dopamine D2, D3, and serotonin, 5HT2A, 5HT2C receptors, COMT, BDNF, and dopamine transporter DAT1 was also associated with different adverse events or variations in the clinical efficacy of aripiprazole. Brexpiprazole also benefits from specific recommendations regarding the CYP2D6 metabolizer status and the risks of associating this antipsychotic with strong/moderate CYP2D6 or CYP3A4 inhibitors. US Food and Drug Administration (FDA) and European Medicines Agency (EMA) recommendations about cariprazine refer to possible pharmacokinetic interactions with strong CYP3A4 inhibitors or inducers. Pharmacogenetic data about cariprazine is sparse, and relevant information regarding gene-drug interactions for lumateperone and pimavanserin is yet lacking. In conclusion, more studies are needed to detect the influence of gene variations on the pharmacokinetics and pharmacodynamics of new-generation antipsychotics. This type of research could increase the ability of clinicians to predict favorable responses to specific antipsychotics and to improve the tolerability of the treatment regimen in patients with SPD.
Collapse
Affiliation(s)
- Octavian Vasiliu
- Department of Psychiatry, Dr. Carol Davila Central Military Emergency University Hospital, Bucharest, Romania
| |
Collapse
|
6
|
Shen T, Pu JL, Jiang YS, Yue YM, He TT, Qu BY, Zhao S, Yan YP, Lai HY, Zhang BR. Impact of cognition-related single nucleotide polymorphisms on brain imaging phenotype in Parkinson's disease. Neural Regen Res 2022; 18:1154-1160. [PMID: 36255006 PMCID: PMC9827791 DOI: 10.4103/1673-5374.355764] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Multiple single nucleotide polymorphisms may contribute to cognitive decline in Parkinson's disease. However, the mechanism by which these single nucleotide polymorphisms modify brain imaging phenotype remains unclear. The aim of this study was to investigate the potential effects of multiple single nucleotide polymorphisms on brain imaging phenotype in Parkinson's disease. Forty-eight Parkinson's disease patients and 39 matched healthy controls underwent genotyping and 7T magnetic resonance imaging. A cognitive-weighted polygenic risk score model was designed, in which the effect sizes were determined individually for 36 single nucleotide polymorphisms. The correlations between polygenic risk score, neuroimaging features, and clinical data were analyzed. Furthermore, individual single nucleotide polymorphism analysis was performed to explore the main effects of genotypes and their interactive effects with Parkinson's disease diagnosis. We found that, in Parkinson's disease, the polygenic risk score was correlated with the neural activity of the hippocampus, parahippocampus, and fusiform gyrus, and with hippocampal-prefrontal and fusiform-temporal connectivity, as well as with gray matter alterations in the orbitofrontal cortex. In addition, we found that single nucleotide polymorphisms in α-synuclein (SNCA) were associated with white matter microstructural changes in the superior corona radiata, corpus callosum, and external capsule. A single nucleotide polymorphism in catechol-O-methyltransferase was associated with the neural activities of the lingual, fusiform, and occipital gyri, which are involved in visual cognitive dysfunction. Furthermore, DRD3 was associated with frontal and temporal lobe function and structure. In conclusion, imaging genetics is useful for providing a better understanding of the genetic pathways involved in the pathophysiologic processes underlying Parkinson's disease. This study provides evidence of an association between genetic factors, cognitive functions, and multi-modality neuroimaging biomarkers in Parkinson's disease.
Collapse
Affiliation(s)
- Ting Shen
- Department of Neurology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, China,Department of Neurology of the Second Affiliated Hospital, Interdisciplinary Institute of Neuroscience and Technology, Key Laboratory of Medical Neurobiology of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| | - Jia-Li Pu
- Department of Neurology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, China
| | - Ya-Si Jiang
- Department of Neurology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, China,Department of Neurology of the Second Affiliated Hospital, Interdisciplinary Institute of Neuroscience and Technology, Key Laboratory of Medical Neurobiology of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| | - Yu-Mei Yue
- Department of Neurology of Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, China
| | - Ting-Ting He
- Department of Neurology of the Second Affiliated Hospital, Interdisciplinary Institute of Neuroscience and Technology, Key Laboratory of Medical Neurobiology of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China,College of Biomedical Engineering and Instrument Science, Key Laboratory for Biomedical Engineering of Ministry of Education, Zhejiang University, Hangzhou, Zhejiang Province, China
| | - Bo-Yi Qu
- Department of Neurology of the Second Affiliated Hospital, Interdisciplinary Institute of Neuroscience and Technology, Key Laboratory of Medical Neurobiology of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China,College of Biomedical Engineering and Instrument Science, Key Laboratory for Biomedical Engineering of Ministry of Education, Zhejiang University, Hangzhou, Zhejiang Province, China
| | - Shuai Zhao
- Department of Neurology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, China
| | - Ya-Ping Yan
- Department of Neurology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, China
| | - Hsin-Yi Lai
- Department of Neurology of the Second Affiliated Hospital, Interdisciplinary Institute of Neuroscience and Technology, Key Laboratory of Medical Neurobiology of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China,Department of Neurology of Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, China,College of Biomedical Engineering and Instrument Science, Key Laboratory for Biomedical Engineering of Ministry of Education, Zhejiang University, Hangzhou, Zhejiang Province, China,Correspondence to: Bao-Rong Zhang, ; Hsin-Yi Lai, .
| | - Bao-Rong Zhang
- Department of Neurology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, China,Correspondence to: Bao-Rong Zhang, ; Hsin-Yi Lai, .
| |
Collapse
|
7
|
Lanfear DE, Luzum JA, She R, Gui H, Donahue MP, O'Connor CM, Adams KF, Sanders-van Wijk S, Zeld N, Maeder MT, Sabbah HN, Kraus WE, Brunner-LaRocca HP, Li J, Williams LK. Polygenic Score for β-Blocker Survival Benefit in European Ancestry Patients With Reduced Ejection Fraction Heart Failure. Circ Heart Fail 2020; 13:e007012. [PMID: 33012170 DOI: 10.1161/circheartfailure.119.007012] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
BACKGROUND β-Blockers (BBs) are mainstay therapy for heart failure with reduced ejection fraction. However, individual patient responses to BB vary, which may be partially due to genetic variation. The goal of this study was to derive and validate the first polygenic response predictor (PRP) for BB survival benefit in heart failure with reduced ejection fraction patients. METHODS Derivation and validation analyses were performed in n=1436 total HF patients of European descent and with ejection fraction <50%. The PRP was derived in a random subset of the Henry Ford Heart Failure Pharmacogenomic Registry (n=248) and then validated in a meta-analysis of the remaining patients from Henry Ford Heart Failure Pharmacogenomic Registry (n=247), the TIME-CHF (Trial of Intensified Versus Standard Medical Therapy in Elderly Patients With Congestive Heart Failure; n=431), and HF-ACTION trial (Heart Failure: a Controlled Trial Investigating Outcomes of Exercise Training; n=510). The PRP was constructed from a genome-wide analysis of BB×genotype interaction predicting time to all-cause mortality, adjusted for Meta-Analysis Global Group in Chronic Heart Failure score, genotype, level of BB exposure, and BB propensity score. RESULTS Five-fold cross-validation summaries out to 1000 single-nucleotide polymorphisms identified optimal prediction with a 44 single-nucleotide polymorphism score and cutoff at the 30th percentile. In validation testing (n=1188), greater BB exposure was associated with reduced all-cause mortality in patients with low PRP score (n=251; hazard ratio, 0.19 [95% CI, 0.04-0.51]; P=0.0075) but not high PRP score (n=937; hazard ratio, 0.84 [95% CI, 0.53-1.3]; P=0.448)-a difference that was statistically significant (P interaction, 0.0235). Results were consistent regardless of atrial fibrillation, ejection fraction (≤40% versus 41%-50%), or when examining cardiovascular death. CONCLUSIONS Among patients of European ancestry with heart failure with reduced ejection fraction, a PRP distinguished patients who derived substantial survival benefit from BB exposure from a larger group that did not. Additional work is needed to prospectively test clinical utility and to develop PRPs for other population groups and other medications.
Collapse
Affiliation(s)
- David E Lanfear
- Department of Internal Medicine, Center for Individualized and Genomic Medicine Research (D.E.L., J.A.L., R.S., H.G., N.Z., J.L., L.K.W.), Henry Ford Hospital, Detroit, MI.,Heart and Vascular Institute (D.E.L., H.N.S., J.L.), Henry Ford Hospital, Detroit, MI
| | - Jasmine A Luzum
- Department of Internal Medicine, Center for Individualized and Genomic Medicine Research (D.E.L., J.A.L., R.S., H.G., N.Z., J.L., L.K.W.), Henry Ford Hospital, Detroit, MI.,Department of Clinical Pharmacy, University of Michigan College of Pharmacy, Ann Arbor (J.A.L.)
| | - Ruicong She
- Department of Internal Medicine, Center for Individualized and Genomic Medicine Research (D.E.L., J.A.L., R.S., H.G., N.Z., J.L., L.K.W.), Henry Ford Hospital, Detroit, MI.,Department of Public Health Sciences (R.S.), Henry Ford Hospital, Detroit, MI
| | - Hongsheng Gui
- Department of Internal Medicine, Center for Individualized and Genomic Medicine Research (D.E.L., J.A.L., R.S., H.G., N.Z., J.L., L.K.W.), Henry Ford Hospital, Detroit, MI
| | - Mark P Donahue
- Division of Cardiology, Duke University, Durham, NC (M.P.D., W.E.K.)
| | | | - Kirkwood F Adams
- Division of Cardiology, University of North Carolina, Chapel Hill (K.F.A.)
| | | | - Nicole Zeld
- Department of Internal Medicine, Center for Individualized and Genomic Medicine Research (D.E.L., J.A.L., R.S., H.G., N.Z., J.L., L.K.W.), Henry Ford Hospital, Detroit, MI
| | - Micha T Maeder
- Cardiology Department, Kantonsspital St. Gallen, Switzerland (M.T.M.)
| | - Hani N Sabbah
- Heart and Vascular Institute (D.E.L., H.N.S., J.L.), Henry Ford Hospital, Detroit, MI
| | - William E Kraus
- Division of Cardiology, Duke University, Durham, NC (M.P.D., W.E.K.)
| | | | - Jia Li
- Department of Internal Medicine, Center for Individualized and Genomic Medicine Research (D.E.L., J.A.L., R.S., H.G., N.Z., J.L., L.K.W.), Henry Ford Hospital, Detroit, MI.,Heart and Vascular Institute (D.E.L., H.N.S., J.L.), Henry Ford Hospital, Detroit, MI
| | - L Keoki Williams
- Department of Internal Medicine, Center for Individualized and Genomic Medicine Research (D.E.L., J.A.L., R.S., H.G., N.Z., J.L., L.K.W.), Henry Ford Hospital, Detroit, MI
| |
Collapse
|
8
|
Aragam KG, Natarajan P. Polygenic Scores to Assess Atherosclerotic Cardiovascular Disease Risk: Clinical Perspectives and Basic Implications. Circ Res 2020; 126:1159-1177. [PMID: 32324503 PMCID: PMC7926201 DOI: 10.1161/circresaha.120.315928] [Citation(s) in RCA: 91] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
An individual's susceptibility to atherosclerotic cardiovascular disease is influenced by numerous clinical and lifestyle factors, motivating the multifaceted approaches currently endorsed for primary and secondary cardiovascular disease prevention. With growing knowledge of the genetic basis of atherosclerotic cardiovascular disease-in particular, coronary artery disease-and its contribution to disease pathogenesis, there is increased interest in understanding the potential clinical utility of a genetic predictor that might further refine the assessment and management of atherosclerotic cardiovascular disease risk. Rapid scientific and technological advances have enabled widespread genotyping efforts and dynamic research in the field of coronary artery disease genetic risk prediction. In this review, we describe how genomic analyses of coronary artery disease have been leveraged to create polygenic risk scores. We then discuss evaluations of the clinical utility of these scores, pertinent mechanistic insights gleaned, and practical considerations relevant to the implementation of polygenic risk scores in the health care setting.
Collapse
Affiliation(s)
- Krishna G. Aragam
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA
- Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA
- Department of Medicine, Harvard Medical School, Boston, MA
| | - Pradeep Natarajan
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA
- Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA
- Department of Medicine, Harvard Medical School, Boston, MA
| |
Collapse
|
9
|
Yoshida K, Müller DJ. Pharmacogenetics of Antipsychotic Drug Treatment: Update and Clinical Implications. MOLECULAR NEUROPSYCHIATRY 2020; 5:1-26. [PMID: 32399466 PMCID: PMC7206586 DOI: 10.1159/000492332] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Accepted: 07/20/2018] [Indexed: 12/24/2022]
Abstract
Numerous genetic variants have been shown to be associated with antipsychotic response and adverse effects of schizophrenia treatment. However, the clinical application of these findings is limited. The aim of this narrative review is to summarize the most recent publications and recommendations related to the genetics of antipsychotic treatment and shed light on the clinical utility of pharmacogenetics/pharmacogenomics (PGx). We reviewed the literature on PGx studies with antipsychotic drugs (i.e., antipsychotic response and adverse effects) and commonly used commercial PGx tools for clinical practice. Publications and reviews were included with emphasis on articles published between January 2015 and April 2018. We found 44 studies focusing on antipsychotic response and 45 studies on adverse effects (e.g., antipsychotic-induced weight gain, movement disorders, hormonal abnormality, and clozapine-induced agranulocytosis/granulocytopenia), albeit with mixed results. Overall, several gene variants related to antipsychotic response and adverse effects in the treatment of patients with schizophrenia have been reported, and several commercial pharmacogenomic tests have become available. However, further well-designed investigations and replication studies in large and well-characterized samples are needed to facilitate the application of PGx findings to clinical practice.
Collapse
Affiliation(s)
- Kazunari Yoshida
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo, Japan
| | - Daniel J. Müller
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
- Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
- Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
10
|
Polygenic Risk Scores for Psychiatric Disorders Reveal Novel Clues About the Genetics of Disordered Gambling. Twin Res Hum Genet 2019; 22:283-289. [PMID: 31608857 DOI: 10.1017/thg.2019.90] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Disordered gambling (DG) is a rare but serious condition that results in considerable financial and interpersonal harms. Twin studies indicate that DG is heritable but are silent with respect to specific genes or pathways involved. Existing genomewide association studies (GWAS) of DG have been substantially underpowered. Larger GWAS of other psychiatric disorders now permit calculation of polygenic risk scores (PRSs) that reflect the aggregated effects of common genetic variants contributing risk for the target condition. The current study investigated whether gambling and DG are associated with PRSs for four psychiatric conditions found to be comorbid with DG in epidemiologic surveys: major depressive disorder (MDD), attention-deficit hyperactivity disorder (ADHD), bipolar disorder (BD) and schizophrenia (SCZ). Genotype data and survey responses were analyzed from the Wave IV assessment (conducted in 2008) of the National Longitudinal Study of Adolescent to Adult Health, a representative sample of adolescents recruited in 1994-1995 and followed into adulthood. Among participants classified as having European ancestry based on genetic analysis (N = 5215), 78.4% reported ever having gambled, and 1.3% reported lifetime DG. Polygenic risk for BD was associated with decreased odds of lifetime gambling, OR = 0.93 [0.87, 0.99], p = .045, pseudo-R2(%) = .12. The SCZ PRS was associated with increased odds of DG, OR = 1.54 [1.07, 2.21], p = .02, pseudo-R2(%) = .85. Polygenic risk scores for MDD and ADHD were not related to either gambling outcome. Investigating features common to both SCZ and DG might generate valuable clues about the genetically influenced liabilities to DG.
Collapse
|
11
|
Pergola G, Di Carlo P, Jaffe AE, Papalino M, Chen Q, Hyde TM, Kleinman JE, Shin JH, Rampino A, Blasi G, Weinberger DR, Bertolino A. Prefrontal Coexpression of Schizophrenia Risk Genes Is Associated With Treatment Response in Patients. Biol Psychiatry 2019; 86:45-55. [PMID: 31126695 DOI: 10.1016/j.biopsych.2019.03.981] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Revised: 03/13/2019] [Accepted: 03/14/2019] [Indexed: 10/27/2022]
Abstract
BACKGROUND Gene coexpression networks are relevant to functional and clinical translation of schizophrenia risk genes. We hypothesized that schizophrenia risk genes converge into coexpression pathways that may be associated with gene regulation mechanisms and with response to treatment in patients with schizophrenia. METHODS We identified gene coexpression networks in two prefrontal cortex postmortem RNA sequencing datasets (n = 688) and replicated them in four more datasets (n = 1295). We identified and replicated (p values < .001) a single module enriched for schizophrenia risk loci (13 risk genes in 10 loci). In silico screening of potential regulators of the schizophrenia risk module via bioinformatic analyses identified two transcription factors and three microRNAs associated with the risk module. To translate postmortem information into clinical phenotypes, we identified polymorphisms predicting coexpression and combined them to obtain an index approximating module coexpression (Polygenic Coexpression Index [PCI]). RESULTS The PCI-coexpression association was successfully replicated in two independent brain transcriptome datasets (n = 131; p values < .05). Finally, we tested the association between the PCI and short-term treatment response in two independent samples of patients with schizophrenia treated with olanzapine (n = 167). The PCI was associated with treatment response in the positive symptom domain in both clinical cohorts (p values < .05). CONCLUSIONS In summary, our findings in 1983 samples of human postmortem prefrontal cortex show that coexpression of a set of genes enriched for schizophrenia risk genes is relevant to treatment response. This coexpression pathway may be coregulated by transcription factors and microRNA associated with it.
Collapse
Affiliation(s)
- Giulio Pergola
- Group of Psychiatric Neuroscience, Department of Basic Medical Sciences, Neuroscience and Sense Organs, University of Bari Aldo Moro, Bari, Italy; Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, Maryland.
| | - Pasquale Di Carlo
- Group of Psychiatric Neuroscience, Department of Basic Medical Sciences, Neuroscience and Sense Organs, University of Bari Aldo Moro, Bari, Italy; Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, Maryland
| | - Andrew E Jaffe
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, Maryland; Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland; Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland; Center for Computational Biology, Johns Hopkins University, Baltimore, Maryland
| | - Marco Papalino
- Group of Psychiatric Neuroscience, Department of Basic Medical Sciences, Neuroscience and Sense Organs, University of Bari Aldo Moro, Bari, Italy
| | - Qiang Chen
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, Maryland
| | - Thomas M Hyde
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, Maryland; Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland; Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Joel E Kleinman
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, Maryland; Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Joo Heon Shin
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, Maryland
| | - Antonio Rampino
- Group of Psychiatric Neuroscience, Department of Basic Medical Sciences, Neuroscience and Sense Organs, University of Bari Aldo Moro, Bari, Italy; Azienda Ospedaliero-Universitaria Consorziale Policlinico, Bari, Italy
| | - Giuseppe Blasi
- Group of Psychiatric Neuroscience, Department of Basic Medical Sciences, Neuroscience and Sense Organs, University of Bari Aldo Moro, Bari, Italy; Azienda Ospedaliero-Universitaria Consorziale Policlinico, Bari, Italy
| | - Daniel R Weinberger
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, Maryland; Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland; McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Alessandro Bertolino
- Group of Psychiatric Neuroscience, Department of Basic Medical Sciences, Neuroscience and Sense Organs, University of Bari Aldo Moro, Bari, Italy; Azienda Ospedaliero-Universitaria Consorziale Policlinico, Bari, Italy.
| |
Collapse
|
12
|
Vita A, Minelli A, Barlati S, Deste G, Giacopuzzi E, Valsecchi P, Turrina C, Gennarelli M. Treatment-Resistant Schizophrenia: Genetic and Neuroimaging Correlates. Front Pharmacol 2019; 10:402. [PMID: 31040787 PMCID: PMC6476957 DOI: 10.3389/fphar.2019.00402] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2018] [Accepted: 04/01/2019] [Indexed: 12/11/2022] Open
Abstract
Schizophrenia is a severe neuropsychiatric disorder that affects approximately 0.5–1% of the population. Response to antipsychotic therapy is highly variable, and it is not currently possible to predict those patients who will or will not respond to antipsychotic medication. Furthermore, a high percentage of patients, approximately 30%, are classified as treatment-resistant (treatment-resistant schizophrenia; TRS). TRS is defined as a non-response to at least two trials of antipsychotic medication of adequate dose and duration. These patients are usually treated with clozapine, the only evidence-based pharmacotherapy for TRS. However, clozapine is associated with severe adverse events. For these reasons, there is an increasing interest to identify better targets for drug development of new compounds and to establish better biomarkers for existing medications. The ability of antipsychotics to improve psychotic symptoms is dependent on their antagonist and reverse agonist activities at different neuroreceptors, and some genetic association studies of TRS have focused on different pharmacodynamic factors. Some genetic studies have shown an association between antipsychotic response or TRS and neurodevelopment candidate genes, antipsychotic mechanisms of action (such as dopaminergic, serotonergic, GABAergic, and glutamatergic) or pharmacokinetic factors (i.e., differences in the cytochrome families). Moreover, there is a growing body of literature on the structural and functional neuroimaging research into TRS. Neuroimaging studies can help to uncover the underlying neurobiological reasons for such resistance and identify resistant patients earlier. Studies examining the neuropharmacological mechanisms of antipsychotics, including clozapine, can help to improve our knowledge of their action on the central nervous system, with further implications for the discovery of biomarkers and the development of new treatments. The identification of the underlying mechanisms of TRS is a major challenge for developing personalized medicine in the psychiatric field for schizophrenia treatment. The main goal of precision medicine is to use genetic and brain-imaging information to improve the safety, effectiveness, and health outcomes of patients via more efficiently targeted risk stratification, prevention, and tailored medication and treatment management approaches. The aim of this review is to summarize the state of art of pharmacogenetic, pharmacogenomic and neuroimaging studies in TRS.
Collapse
Affiliation(s)
- Antonio Vita
- Department of Mental Health and Addiction Services, ASST Spedali Civili, Brescia, Italy.,Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Alessandra Minelli
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Stefano Barlati
- Department of Mental Health and Addiction Services, ASST Spedali Civili, Brescia, Italy.,Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Giacomo Deste
- Department of Mental Health and Addiction Services, ASST Spedali Civili, Brescia, Italy
| | - Edoardo Giacopuzzi
- Genetic Unit, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| | - Paolo Valsecchi
- Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Cesare Turrina
- Department of Mental Health and Addiction Services, ASST Spedali Civili, Brescia, Italy.,Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Massimo Gennarelli
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy.,Genetic Unit, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| |
Collapse
|
13
|
Ho DSW, Schierding W, Wake M, Saffery R, O’Sullivan J. Machine Learning SNP Based Prediction for Precision Medicine. Front Genet 2019; 10:267. [PMID: 30972108 PMCID: PMC6445847 DOI: 10.3389/fgene.2019.00267] [Citation(s) in RCA: 104] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Accepted: 03/11/2019] [Indexed: 12/17/2022] Open
Abstract
In the past decade, precision genomics based medicine has emerged to provide tailored and effective healthcare for patients depending upon their genetic features. Genome Wide Association Studies have also identified population based risk genetic variants for common and complex diseases. In order to meet the full promise of precision medicine, research is attempting to leverage our increasing genomic understanding and further develop personalized medical healthcare through ever more accurate disease risk prediction models. Polygenic risk scoring and machine learning are two primary approaches for disease risk prediction. Despite recent improvements, the results of polygenic risk scoring remain limited due to the approaches that are currently used. By contrast, machine learning algorithms have increased predictive abilities for complex disease risk. This increase in predictive abilities results from the ability of machine learning algorithms to handle multi-dimensional data. Here, we provide an overview of polygenic risk scoring and machine learning in complex disease risk prediction. We highlight recent machine learning application developments and describe how machine learning approaches can lead to improved complex disease prediction, which will help to incorporate genetic features into future personalized healthcare. Finally, we discuss how the future application of machine learning prediction models might help manage complex disease by providing tissue-specific targets for customized, preventive interventions.
Collapse
Affiliation(s)
| | | | - Melissa Wake
- Murdoch Children Research Institute, Melbourne, VIC, Australia
| | - Richard Saffery
- Murdoch Children Research Institute, Melbourne, VIC, Australia
| | | |
Collapse
|
14
|
Alemany-Navarro M, Costas J, Real E, Segalàs C, Bertolín S, Domènech L, Rabionet R, Carracedo Á, Menchón JM, Alonso P. Do polygenic risk and stressful life events predict pharmacological treatment response in obsessive compulsive disorder? A gene-environment interaction approach. Transl Psychiatry 2019; 9:70. [PMID: 30718812 PMCID: PMC6362161 DOI: 10.1038/s41398-019-0410-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Revised: 08/18/2018] [Accepted: 11/08/2018] [Indexed: 12/13/2022] Open
Abstract
The rate of response to pharmacological treatment in Obsessive-compulsive disorder (OCD) oscillates between 40 and 70%. Genetic and environmental factors have been associated with treatment response in OCD. This study analyzes the predictive ability of a polygenic risk score (PRS) built from OCD-risk variants, for treatment response in OCD, and the modulation role of stressful life events (SLEs) at the onset of the disorder. PRSs were calculated for a sample of 103 patients. Yale-Brown Obsessive Compulsive Scale (YBOCS) scores were obtained before and after a 12-week treatment. Regression analyses were performed to analyze the influence of the PRS and SLEs at onset on treatment response. PRS did not predict treatment response. The best predictive model for post-treatment YBOCS (post YBOCS) included basal YBOCS and age. PRS appeared as a predictor for basal and post YBOCS. SLEs at onset were not a predictor for treatment response when included in the regression model. No evidence for PRS predictive ability for treatment response was found. The best predictor for treatment response was age, agreeing with previous literature specific for SRI treatment. Suggestions are made on the possible role of neuroplasticity as a mediator on this association. PRS significantly predicted OCD severity independent on pharmacological treatment. SLE at onset modulation role was not evidenced. Further research is needed to elucidate the genetic and environmental bases of treatment response in OCD.
Collapse
Affiliation(s)
- María Alemany-Navarro
- Institut d' Investigació Biomèdica de Bellvitge (IDIBELL), L'Hospitalet de Llobregat (Barcelona), Spain. .,OCD Clinical and Research Unit, Psychiatry Department, Hospital Universitari de Bellvitge, L'Hospitalet de Llobregat (Barcelona), Spain.
| | - Javier Costas
- 0000 0000 9403 4738grid.420359.9Grupo de Xenética Psiquiátrica, Instituto de Investigación Sanitaria de Santiago, Complexo Hospitalario Universitario de Santiago de Compostela, Servizo Galego de Saúde, Santiago de Compostela, Spain
| | - Eva Real
- Institut d’ Investigació Biomèdica de Bellvitge (IDIBELL), L’Hospitalet de Llobregat (Barcelona), Spain ,0000 0000 8836 0780grid.411129.eOCD Clinical and Research Unit, Psychiatry Department, Hospital Universitari de Bellvitge, L’Hospitalet de Llobregat (Barcelona), Spain
| | - Cinto Segalàs
- Institut d’ Investigació Biomèdica de Bellvitge (IDIBELL), L’Hospitalet de Llobregat (Barcelona), Spain ,0000 0000 8836 0780grid.411129.eOCD Clinical and Research Unit, Psychiatry Department, Hospital Universitari de Bellvitge, L’Hospitalet de Llobregat (Barcelona), Spain
| | - Sara Bertolín
- 0000 0000 8836 0780grid.411129.eOCD Clinical and Research Unit, Psychiatry Department, Hospital Universitari de Bellvitge, L’Hospitalet de Llobregat (Barcelona), Spain
| | - Laura Domènech
- grid.473715.3Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Dr. Aiguader 88, Barcelona, 08003 Spain ,0000 0001 2172 2676grid.5612.0Universitat Pompeu Fabra (UPF), Barcelona, Spain ,CIBER in Epidemiology and Public Health (CIBERESP), Barcelona, Spain
| | - Raquel Rabionet
- grid.473715.3Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Dr. Aiguader 88, Barcelona, 08003 Spain ,0000 0001 2172 2676grid.5612.0Universitat Pompeu Fabra (UPF), Barcelona, Spain ,CIBER in Epidemiology and Public Health (CIBERESP), Barcelona, Spain
| | - Ángel Carracedo
- 0000 0000 9403 4738grid.420359.9Grupo de Xenética Psiquiátrica, Instituto de Investigación Sanitaria de Santiago, Complexo Hospitalario Universitario de Santiago de Compostela, Servizo Galego de Saúde, Santiago de Compostela, Spain ,0000000109410645grid.11794.3aGrupo de Medicina Xenómica, Universidade de Santiago de Compostela, Centro Nacional de Genotipado - Instituto Carlos III, Santiago de Compostela, Spain ,0000 0004 1791 1185grid.452372.5Centro de Investigación Biomédica en Red de Enfermedades Raras, Santiago de Compostela, Spain
| | - Jose M. Menchón
- Institut d’ Investigació Biomèdica de Bellvitge (IDIBELL), L’Hospitalet de Llobregat (Barcelona), Spain ,0000 0000 8836 0780grid.411129.eOCD Clinical and Research Unit, Psychiatry Department, Hospital Universitari de Bellvitge, L’Hospitalet de Llobregat (Barcelona), Spain ,0000 0000 9314 1427grid.413448.eCIBERSAM (Centro de Investigación en Red de Salud Mental), Instituto de Salud Carlos III, Madrid, Spain ,0000 0004 1937 0247grid.5841.8Department of Clinical Sciences, Bellvitge Campus, University of Barcelona, Barcelona, Spain
| | - Pino Alonso
- Institut d’ Investigació Biomèdica de Bellvitge (IDIBELL), L’Hospitalet de Llobregat (Barcelona), Spain ,0000 0000 8836 0780grid.411129.eOCD Clinical and Research Unit, Psychiatry Department, Hospital Universitari de Bellvitge, L’Hospitalet de Llobregat (Barcelona), Spain ,0000 0000 9314 1427grid.413448.eCIBERSAM (Centro de Investigación en Red de Salud Mental), Instituto de Salud Carlos III, Madrid, Spain ,0000 0004 1937 0247grid.5841.8Department of Clinical Sciences, Bellvitge Campus, University of Barcelona, Barcelona, Spain
| |
Collapse
|
15
|
Mistry S, Harrison JR, Smith DJ, Escott-Price V, Zammit S. The use of polygenic risk scores to identify phenotypes associated with genetic risk of schizophrenia: Systematic review. Schizophr Res 2018; 197:2-8. [PMID: 29129507 DOI: 10.1016/j.schres.2017.10.037] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Revised: 10/27/2017] [Accepted: 10/28/2017] [Indexed: 12/12/2022]
Abstract
Studying the phenotypic manifestations of increased genetic liability for schizophrenia can increase our understanding of this disorder. Specifically, information from alleles identified in genome-wide association studies can be collapsed into a polygenic risk score (PRS) to explore how genetic risk is manifest within different samples. In this systematic review, we provide a comprehensive assessment of studies examining associations between schizophrenia PRS (SZ-PRS) and several phenotypic measures. We searched EMBASE, Medline and PsycINFO (from August 2009-14th March 2016) plus references of included studies, following PRISMA guidelines. Study inclusion was based on predetermined criteria and data were extracted independently and in duplicate. Overall, SZ-PRS was associated with increased risk for psychiatric disorders such as depression and bipolar disorder, lower performance IQ and negative symptoms. SZ-PRS explained up to 6% of genetic variation in psychiatric phenotypes, compared to <0.7% in measures of cognition. Future gains from using the PRS approach may be greater if used for examining phenotypes that are more closely related to biological substrates, for scores based on gene-pathways, and where PRSs are used to stratify individuals for study of treatment response. As it was difficult to interpret findings across studies due to insufficient information provided by many studies, we propose a framework to guide robust reporting of PRS associations in the future.
Collapse
Affiliation(s)
- Sumit Mistry
- Institute of Psychological Medicine and Clinical Neurosciences, MRC Centre for Neuropsychiatric Genetics and Genomics, Cardiff University, UK.
| | - Judith R Harrison
- Institute of Psychological Medicine and Clinical Neurosciences, MRC Centre for Neuropsychiatric Genetics and Genomics, Cardiff University, UK
| | - Daniel J Smith
- Institute of Health and Wellbeing, 1 Lilybank Gardens, University of Glasgow, UK
| | - Valentina Escott-Price
- Institute of Psychological Medicine and Clinical Neurosciences, MRC Centre for Neuropsychiatric Genetics and Genomics, Cardiff University, UK
| | - Stanley Zammit
- Institute of Psychological Medicine and Clinical Neurosciences, MRC Centre for Neuropsychiatric Genetics and Genomics, Cardiff University, UK; Centre for Academic Mental Health, School of Social and Community Medicine, University of Bristol, UK
| |
Collapse
|
16
|
Bogdan R, Baranger DAA, Agrawal A. Polygenic Risk Scores in Clinical Psychology: Bridging Genomic Risk to Individual Differences. Annu Rev Clin Psychol 2018; 14:119-157. [PMID: 29579395 PMCID: PMC7772939 DOI: 10.1146/annurev-clinpsy-050817-084847] [Citation(s) in RCA: 90] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Genomewide association studies (GWASs) across psychiatric phenotypes have shown that common genetic variants generally confer risk with small effect sizes (odds ratio < 1.1) that additively contribute to polygenic risk. Summary statistics derived from large discovery GWASs can be used to generate polygenic risk scores (PRS) in independent, target data sets to examine correlates of polygenic disorder liability (e.g., does genetic liability to schizophrenia predict cognition?). The intuitive appeal and generalizability of PRS have led to their widespread use and new insights into mechanisms of polygenic liability. However, when currently applied across traits they account for small amounts of variance (<3%), are relatively uninformative for clinical treatment, and, in isolation, provide no insight into molecular mechanisms. Larger GWASs are needed to increase the precision of PRS, and novel approaches integrating various data sources (e.g., multitrait analysis of GWASs) may improve the utility of current PRS.
Collapse
Affiliation(s)
- Ryan Bogdan
- BRAINLab, Department of Psychological and Brain Sciences, and Division of Biology and Biomedical Sciences, Washington University in St. Louis, St. Louis, Missouri 63110, USA;
| | - David A A Baranger
- BRAINLab, Department of Psychological and Brain Sciences, and Division of Biology and Biomedical Sciences, Washington University in St. Louis, St. Louis, Missouri 63110, USA;
| | - Arpana Agrawal
- Department of Psychiatry, Washington University in St. Louis School of Medicine, St. Louis, Missouri 63110, USA
| |
Collapse
|
17
|
Sengupta SM, MacDonald K, Fathalli F, Yim A, Lepage M, Iyer S, Malla A, Joober R. Polygenic Risk Score associated with specific symptom dimensions in first-episode psychosis. Schizophr Res 2017; 184:116-121. [PMID: 27916287 DOI: 10.1016/j.schres.2016.11.039] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Revised: 11/21/2016] [Accepted: 11/24/2016] [Indexed: 12/14/2022]
Abstract
Recent Genome-Wide Association Studies (GWAS) have provided evidence for the involvement of a number of genetic variants in schizophrenia (SCZ). The objective of the current study was to examine the association between these variants and symptom dimensions, evaluated prospectively over a period of 24months, in a clinically well-characterized sample of individuals (n=241) with first-episode psychosis (FEP). The genetic variants were analyzed collectively as captured through a Polygenic Risk Score (PRS), calculated for each individual. At each evaluation time point (baseline, 1, 2, 6 and 24months), correlation analysis was conducted with PRS and symptom dimension scores assessed by the Positive and Negative Syndrome Scale (PANSS). We also examined the association of PRS with global symptom rating, depression, anxiety, social and occupational functioning as measured by widely used and well validated scales. At baseline, significant positive correlation was observed between PRS and the general psychopathology dimension of the PANSS but no associations were observed with the positive or negative symptom dimensions. Anxiety, assessed using the Hamilton Anxiety Rating Scale, was also significantly correlated with the PRS. No significant correlation was observed with other symptom dimensions or with the PANSS score at the later evaluations. These results provide novel evidence of an association between general psychopathology and PRS in young people with first episode psychosis. They also demonstrate that it is important to note the dynamic changes of symptoms over time when trying to refine the relationship between genetic factors and phenotypes.
Collapse
Affiliation(s)
- Sarojini M Sengupta
- Douglas Mental Health University Institute, 6875 LaSalle Blvd, Verdun, Quebec H4H 1R3, Canada; Department of Psychiatry, McGill University, Ludmer Research & Training Building, 1033 Pine Ave. West, Montreal, Quebec H3A 1A1, Canada.
| | - Kathleen MacDonald
- Douglas Mental Health University Institute, 6875 LaSalle Blvd, Verdun, Quebec H4H 1R3, Canada; Department of Psychiatry, McGill University, Ludmer Research & Training Building, 1033 Pine Ave. West, Montreal, Quebec H3A 1A1, Canada
| | - Ferid Fathalli
- Douglas Mental Health University Institute, 6875 LaSalle Blvd, Verdun, Quebec H4H 1R3, Canada; Department of Psychiatry, McGill University, Ludmer Research & Training Building, 1033 Pine Ave. West, Montreal, Quebec H3A 1A1, Canada
| | - Anita Yim
- Department of Medicine, Université de Sherbrooke, Local E5-1283, Sherbrooke, Québec J1K 2R1, Canada
| | - Martin Lepage
- Douglas Mental Health University Institute, 6875 LaSalle Blvd, Verdun, Quebec H4H 1R3, Canada; Department of Psychiatry, McGill University, Ludmer Research & Training Building, 1033 Pine Ave. West, Montreal, Quebec H3A 1A1, Canada; Integrated Program in Neuroscience, McGill University, Room 141, Montreal Neurological Institute, 3801 University Street, Montreal, Quebec H3A 2B4, Canada
| | - Srividya Iyer
- Douglas Mental Health University Institute, 6875 LaSalle Blvd, Verdun, Quebec H4H 1R3, Canada; Department of Psychiatry, McGill University, Ludmer Research & Training Building, 1033 Pine Ave. West, Montreal, Quebec H3A 1A1, Canada
| | - Ashok Malla
- Douglas Mental Health University Institute, 6875 LaSalle Blvd, Verdun, Quebec H4H 1R3, Canada; Department of Psychiatry, McGill University, Ludmer Research & Training Building, 1033 Pine Ave. West, Montreal, Quebec H3A 1A1, Canada
| | - Ridha Joober
- Douglas Mental Health University Institute, 6875 LaSalle Blvd, Verdun, Quebec H4H 1R3, Canada; Department of Psychiatry, McGill University, Ludmer Research & Training Building, 1033 Pine Ave. West, Montreal, Quebec H3A 1A1, Canada; Integrated Program in Neuroscience, McGill University, Room 141, Montreal Neurological Institute, 3801 University Street, Montreal, Quebec H3A 2B4, Canada; Department of Human Genetics, McGill University, Room N5-13, Stewart Biology Building, 1205 Dr. Penfield Ave., Montreal, Quebec H3A 1B1, Canada
| |
Collapse
|
18
|
Lally J, Gaughran F, Timms P, Curran SR. Treatment-resistant schizophrenia: current insights on the pharmacogenomics of antipsychotics. Pharmgenomics Pers Med 2016; 9:117-129. [PMID: 27853387 PMCID: PMC5106233 DOI: 10.2147/pgpm.s115741] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Up to 30% of people with schizophrenia do not respond to two (or more) trials of dopaminergic antipsychotics. They are said to have treatment-resistant schizophrenia (TRS). Clozapine is still the only effective treatment for TRS, although it is underused in clinical practice. Initial use is delayed, it can be hard for patients to tolerate, and clinicians can be uncertain as to when to use it. What if, at the start of treatment, we could identify those patients likely to respond to clozapine - and those likely to suffer adverse effects? It is likely that clinicians would feel less inhibited about using it, allowing clozapine to be used earlier and more appropriately. Genetic testing holds out the tantalizing possibility of being able to do just this, and hence the vital importance of pharmacogenomic studies. These can potentially identify genetic markers for both tolerance of and vulnerability to clozapine. We aim to summarize progress so far, possible clinical applications, limitations to the evidence, and problems in applying these findings to the management of TRS. Pharmacogenomic studies of clozapine response and tolerability have produced conflicting results. These are due, at least in part, to significant differences in the patient groups studied. The use of clinical pharmacogenomic testing - to personalize clozapine treatment and identify patients at high risk of treatment failure or of adverse events - has moved closer over the last 20 years. However, to develop such testing that could be used clinically will require larger, multicenter, prospective studies.
Collapse
Affiliation(s)
- John Lally
- Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, UK
- Department of Psychiatry, Royal College of Surgeons in Ireland, Beaumont Hospital, Dublin, Ireland
- National Psychosis Service
| | - Fiona Gaughran
- Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, UK
- National Psychosis Service
| | - Philip Timms
- START Team, South London and Maudsley NHS Foundation Trust
- King’s College London
| | - Sarah R Curran
- King’s College London
- South West London and St George’s Mental Health NHS Foundation Trust
- St George’s University of London, London, UK
| |
Collapse
|