1
|
Abdelhalim H, Berber A, Lodi M, Jain R, Nair A, Pappu A, Patel K, Venkat V, Venkatesan C, Wable R, Dinatale M, Fu A, Iyer V, Kalove I, Kleyman M, Koutsoutis J, Menna D, Paliwal M, Patel N, Patel T, Rafique Z, Samadi R, Varadhan R, Bolla S, Vadapalli S, Ahmed Z. Artificial Intelligence, Healthcare, Clinical Genomics, and Pharmacogenomics Approaches in Precision Medicine. Front Genet 2022; 13:929736. [PMID: 35873469 PMCID: PMC9299079 DOI: 10.3389/fgene.2022.929736] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Accepted: 05/25/2022] [Indexed: 12/13/2022] Open
Abstract
Precision medicine has greatly aided in improving health outcomes using earlier diagnosis and better prognosis for chronic diseases. It makes use of clinical data associated with the patient as well as their multi-omics/genomic data to reach a conclusion regarding how a physician should proceed with a specific treatment. Compared to the symptom-driven approach in medicine, precision medicine considers the critical fact that all patients do not react to the same treatment or medication in the same way. When considering the intersection of traditionally distinct arenas of medicine, that is, artificial intelligence, healthcare, clinical genomics, and pharmacogenomics—what ties them together is their impact on the development of precision medicine as a field and how they each contribute to patient-specific, rather than symptom-specific patient outcomes. This study discusses the impact and integration of these different fields in the scope of precision medicine and how they can be used in preventing and predicting acute or chronic diseases. Additionally, this study also discusses the advantages as well as the current challenges associated with artificial intelligence, healthcare, clinical genomics, and pharmacogenomics.
Collapse
Affiliation(s)
- Habiba Abdelhalim
- Rutgers Institute for Health, Health Care Policy and Aging Research, Rutgers University, New Brunswick, NJ, United States
| | - Asude Berber
- Rutgers Institute for Health, Health Care Policy and Aging Research, Rutgers University, New Brunswick, NJ, United States
| | - Mudassir Lodi
- Rutgers Institute for Health, Health Care Policy and Aging Research, Rutgers University, New Brunswick, NJ, United States
| | - Rihi Jain
- Rutgers Institute for Health, Health Care Policy and Aging Research, Rutgers University, New Brunswick, NJ, United States
| | - Achuth Nair
- Rutgers Institute for Health, Health Care Policy and Aging Research, Rutgers University, New Brunswick, NJ, United States
| | - Anirudh Pappu
- Rutgers Institute for Health, Health Care Policy and Aging Research, Rutgers University, New Brunswick, NJ, United States
| | - Kush Patel
- Rutgers Institute for Health, Health Care Policy and Aging Research, Rutgers University, New Brunswick, NJ, United States
| | - Vignesh Venkat
- Rutgers Institute for Health, Health Care Policy and Aging Research, Rutgers University, New Brunswick, NJ, United States
| | - Cynthia Venkatesan
- Rutgers Institute for Health, Health Care Policy and Aging Research, Rutgers University, New Brunswick, NJ, United States
| | - Raghu Wable
- Rutgers Institute for Health, Health Care Policy and Aging Research, Rutgers University, New Brunswick, NJ, United States
| | - Matthew Dinatale
- Rutgers Institute for Health, Health Care Policy and Aging Research, Rutgers University, New Brunswick, NJ, United States
| | - Allyson Fu
- Rutgers Institute for Health, Health Care Policy and Aging Research, Rutgers University, New Brunswick, NJ, United States
| | - Vikram Iyer
- Rutgers Institute for Health, Health Care Policy and Aging Research, Rutgers University, New Brunswick, NJ, United States
| | - Ishan Kalove
- Rutgers Institute for Health, Health Care Policy and Aging Research, Rutgers University, New Brunswick, NJ, United States
| | - Marc Kleyman
- Rutgers Institute for Health, Health Care Policy and Aging Research, Rutgers University, New Brunswick, NJ, United States
| | - Joseph Koutsoutis
- Rutgers Institute for Health, Health Care Policy and Aging Research, Rutgers University, New Brunswick, NJ, United States
| | - David Menna
- Rutgers Institute for Health, Health Care Policy and Aging Research, Rutgers University, New Brunswick, NJ, United States
| | - Mayank Paliwal
- Rutgers Institute for Health, Health Care Policy and Aging Research, Rutgers University, New Brunswick, NJ, United States
| | - Nishi Patel
- Rutgers Institute for Health, Health Care Policy and Aging Research, Rutgers University, New Brunswick, NJ, United States
| | - Thirth Patel
- Rutgers Institute for Health, Health Care Policy and Aging Research, Rutgers University, New Brunswick, NJ, United States
| | - Zara Rafique
- Rutgers Institute for Health, Health Care Policy and Aging Research, Rutgers University, New Brunswick, NJ, United States
| | - Rothela Samadi
- Rutgers Institute for Health, Health Care Policy and Aging Research, Rutgers University, New Brunswick, NJ, United States
| | - Roshan Varadhan
- Rutgers Institute for Health, Health Care Policy and Aging Research, Rutgers University, New Brunswick, NJ, United States
| | - Shreyas Bolla
- Rutgers Institute for Health, Health Care Policy and Aging Research, Rutgers University, New Brunswick, NJ, United States
| | - Sreya Vadapalli
- Rutgers Institute for Health, Health Care Policy and Aging Research, Rutgers University, New Brunswick, NJ, United States
| | - Zeeshan Ahmed
- Rutgers Institute for Health, Health Care Policy and Aging Research, Rutgers University, New Brunswick, NJ, United States.,Department of Medicine, Rutgers Robert Wood Johnson Medical School, Rutgers Biomedical and Health Sciences, New Brunswick, NJ, United States
| |
Collapse
|
2
|
Chambliss AB, Chan DW. Precision medicine: from pharmacogenomics to pharmacoproteomics. Clin Proteomics 2016; 13:25. [PMID: 27708556 PMCID: PMC5037608 DOI: 10.1186/s12014-016-9127-8] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Accepted: 09/17/2016] [Indexed: 12/31/2022] Open
Abstract
Disease progression and drug response may vary significantly from patient to patient. Fortunately, the rapid development of high-throughput ‘omics’ technologies has allowed for the identification of potential biomarkers that may aid in the understanding of the heterogeneities in disease development and treatment outcomes. However, mechanistic gaps remain when the genome or the proteome are investigated independently in response to drug treatment. In this article, we discuss the current status of pharmacogenomics in precision medicine and highlight the needs for concordant analysis at the proteome and metabolome levels via the more recently-evolved fields of pharmacoproteomics, toxicoproteomics, and pharmacometabolomics. Integrated ‘omics’ investigations will be critical in piecing together targetable mechanisms of action for both drug development and monitoring of therapy in order to fully apply precision medicine to the clinic.
Collapse
Affiliation(s)
- Allison B Chambliss
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21287 USA ; Department of Pathology, Keck School of Medicine of USC, Los Angeles, CA 90033 USA
| | - Daniel W Chan
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21287 USA
| |
Collapse
|
3
|
Li J, Lu Z. Systematic identification of pharmacogenomics information from clinical trials. J Biomed Inform 2012; 45:870-8. [PMID: 22546622 PMCID: PMC3760158 DOI: 10.1016/j.jbi.2012.04.005] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2011] [Revised: 03/13/2012] [Accepted: 04/11/2012] [Indexed: 11/23/2022]
Abstract
Recent progress in high-throughput genomic technologies has shifted pharmacogenomic research from candidate gene pharmacogenetics to clinical pharmacogenomics (PGx). Many clinical related questions may be asked such as 'what drug should be prescribed for a patient with mutant alleles?' Typically, answers to such questions can be found in publications mentioning the relationships of the gene-drug-disease of interest. In this work, we hypothesize that ClinicalTrials.gov is a comparable source rich in PGx related information. In this regard, we developed a systematic approach to automatically identify PGx relationships between genes, drugs and diseases from trial records in ClinicalTrials.gov. In our evaluation, we found that our extracted relationships overlap significantly with the curated factual knowledge through the literature in a PGx database and that most relationships appear on average 5 years earlier in clinical trials than in their corresponding publications, suggesting that clinical trials may be valuable for both validating known and capturing new PGx related information in a more timely manner. Furthermore, two human reviewers judged a portion of computer-generated relationships and found an overall accuracy of 74% for our text-mining approach. This work has practical implications in enriching our existing knowledge on PGx gene-drug-disease relationships as well as suggesting crosslinks between ClinicalTrials.gov and other PGx knowledge bases.
Collapse
Affiliation(s)
- Jiao Li
- National Library of Medicine, National Institutes of Health, Bethesda, MD 20894, United States
| | - Zhiyong Lu
- National Library of Medicine, National Institutes of Health, Bethesda, MD 20894, United States
| |
Collapse
|
4
|
|
5
|
Rice MJ. The institutional review board is an impediment to human research: the result is more animal-based research. Philos Ethics Humanit Med 2011; 6:12. [PMID: 21649895 PMCID: PMC3127833 DOI: 10.1186/1747-5341-6-12] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2010] [Accepted: 06/07/2011] [Indexed: 05/15/2023] Open
Abstract
Biomedical research today can be generally classified as human-based or nonhuman animal-based, each with separate and distinct review boards that must approve research protocols. Researchers wishing to work with humans or human tissues have become frustrated by the required burdensome approval panel, the Institutional Review Board. However, scientists have found it is much easier to work with the animal-based research review board, the Institutional Animal Care and Use Committee. Consequently, animals are used for investigations even when scientists believe these studies should be performed with humans or human tissue. This situation deserves attention from society and more specifically the animal protection and patient advocate communities, as neither patients nor animals are well served by the present situation.
Collapse
Affiliation(s)
- Mark J Rice
- Department of Anesthesiology, University of Florida College of Medicine, Gainesville, FL 32610-0254, USA.
| |
Collapse
|
6
|
Mishra L. Health care reform: how personalized medicine could help bundling of care for liver diseases. Hepatology 2011; 53:379-81. [PMID: 21274858 PMCID: PMC3444166 DOI: 10.1002/hep.24144] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Affiliation(s)
- Lopa Mishra
- Department of Gastroenterology, Hepatology, & Nutrition, The University of Texas M. D. Anderson Cancer Center, Houston, Texas 77030-4009, USA.
| |
Collapse
|
7
|
Pereira TV, Patsopoulos NA, Pereira AC, Krieger JE. Strategies for genetic model specification in the screening of genome-wide meta-analysis signals for further replication. Int J Epidemiol 2010; 40:457-69. [PMID: 21149279 DOI: 10.1093/ije/dyq203] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND Meta-analysis is increasingly being employed as a screening procedure in large-scale association studies to select promising variants for follow-up studies. However, standard methods for meta-analysis require the assumption of an underlying genetic model, which is typically unknown a priori. This drawback can introduce model misspecifications, causing power to be suboptimal, or the evaluation of multiple genetic models, which augments the number of false-positive associations, ultimately leading to waste of resources with fruitless replication studies. We used simulated meta-analyses of large genetic association studies to investigate naïve strategies of genetic model specification to optimize screenings of genome-wide meta-analysis signals for further replication. METHODS Different methods, meta-analytical models and strategies were compared in terms of power and type-I error. Simulations were carried out for a binary trait in a wide range of true genetic models, genome-wide thresholds, minor allele frequencies (MAFs), odds ratios and between-study heterogeneity (τ²). RESULTS Among the investigated strategies, a simple Bonferroni-corrected approach that fits both multiplicative and recessive models was found to be optimal in most examined scenarios, reducing the likelihood of false discoveries and enhancing power in scenarios with small MAFs either in the presence or in absence of heterogeneity. Nonetheless, this strategy is sensitive to τ² whenever the susceptibility allele is common (MAF ≥ 30%), resulting in an increased number of false-positive associations compared with an analysis that considers only the multiplicative model. CONCLUSION Invoking a simple Bonferroni adjustment and testing for both multiplicative and recessive models is fast and an optimal strategy in large meta-analysis-based screenings. However, care must be taken when examined variants are common, where specification of a multiplicative model alone may be preferable.
Collapse
Affiliation(s)
- Tiago V Pereira
- Laboratory of Genetics and Molecular Cardiology, Heart Institute (InCor), São Paulo University Medical School, University of São Paulo, São Paulo, Brazil
| | | | | | | |
Collapse
|
8
|
Greek R, Greek J. Is the use of sentient animals in basic research justifiable? Philos Ethics Humanit Med 2010; 5:14. [PMID: 20825676 PMCID: PMC2949619 DOI: 10.1186/1747-5341-5-14] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2009] [Accepted: 09/08/2010] [Indexed: 05/22/2023] Open
Abstract
Animals can be used in many ways in science and scientific research. Given that society values sentient animals and that basic research is not goal oriented, the question is raised: "Is the use of sentient animals in basic research justifiable?" We explore this in the context of funding issues, outcomes from basic research, and the position of society as a whole on using sentient animals in research that is not goal oriented. We conclude that the use of sentient animals in basic research cannot be justified in light of society's priorities.
Collapse
Affiliation(s)
- Ray Greek
- Americans For Medical Advancement, 2251 Refugio Rd, Goleta, CA 93117, USA
| | - Jean Greek
- Americans For Medical Advancement, 2251 Refugio Rd, Goleta, CA 93117, USA
| |
Collapse
|
9
|
Schulze TG, Alda M, Adli M, Akula N, Ardau R, Bui ET, Chillotti C, Cichon S, Czerski P, Del Zompo M, Detera-Wadleigh SD, Grof P, Gruber O, Hashimoto R, Hauser J, Hoban R, Iwata N, Kassem L, Kato T, Kittel-Schneider S, Kliwicki S, Kelsoe JR, Kusumi I, Laje G, Leckband SG, Manchia M, MacQueen G, Masui T, Ozaki N, Perlis RH, Pfennig A, Piccardi P, Richardson S, Rouleau G, Reif A, Rybakowski JK, Sasse J, Schumacher J, Severino G, Smoller JW, Squassina A, Turecki G, Young LT, Yoshikawa T, Bauer M, McMahon FJ. The International Consortium on Lithium Genetics (ConLiGen): an initiative by the NIMH and IGSLI to study the genetic basis of response to lithium treatment. Neuropsychobiology 2010; 62:72-8. [PMID: 20453537 PMCID: PMC2889682 DOI: 10.1159/000314708] [Citation(s) in RCA: 98] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
For more than half a decade, lithium has been successfully used to treat bipolar disorder. Worldwide, it is considered the first-line mood stabilizer. Apart from its proven antimanic and prophylactic effects, considerable evidence also suggests an antisuicidal effect in affective disorders. Lithium is also effectively used to augment antidepressant drugs in the treatment of refractory major depressive episodes and prevent relapses in recurrent unipolar depression. In contrast to many psychiatric drugs, lithium has outlasted various pharmacotherapeutic 'fashions', and remains an indispensable element in contemporary psychopharmacology. Nevertheless, data from pharmacogenetic studies of lithium are comparatively sparse, and these studies are generally characterized by small sample sizes and varying definitions of response. Here, we present an international effort to elucidate the genetic underpinnings of lithium response in bipolar disorder. Following an initiative by the International Group for the Study of Lithium-Treated Patients (www.IGSLI.org) and the Unit on the Genetic Basis of Mood and Anxiety Disorders at the National Institute of Mental Health,lithium researchers from around the world have formed the Consortium on Lithium Genetics (www.ConLiGen.org) to establish the largest sample to date for genome-wide studies of lithium response in bipolar disorder, currently comprising more than 1,200 patients characterized for response to lithium treatment. A stringent phenotype definition of response is one of the hallmarks of this collaboration. ConLiGen invites all lithium researchers to join its efforts.
Collapse
Affiliation(s)
- Thomas G. Schulze
- Genetic Basis of Mood and Anxiety Disorders, National Institute of Mental Health, National Institutes of Health, Department of Health and Human Services, Bethesda, Md, USA,Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health, Mannheim, Germany,The International Group for the Study of Lithium-Treated Patients (IGSLI), Research Center Juelich, Juelich, Germany,*Thomas G. Schulze, MD, Unit on the Genetic Basis of Mood and Anxiety Disorders, National Institute of Mental Health (NIMH), National Institutes of Health (NIH), 35 Convent Drive, Bldg. 35, Rm 1A205, MSC 3719, Bethesda, MD 20892-3719 (USA), Tel. +1 301 451 7213, Fax +1 301 402 9081, E-Mail
| | - Martin Alda
- Department of Psychiatry, Dalhousie University, Halifax, N.S., Canada,The International Group for the Study of Lithium-Treated Patients (IGSLI), Research Center Juelich, Juelich, Germany
| | - Mazda Adli
- Department of Psychiatry and Psychotherapy, Charité – Universitätsmedizin Berlin, Campus Charité Mitte, Berlin, Germany,The International Group for the Study of Lithium-Treated Patients (IGSLI), Research Center Juelich, Juelich, Germany
| | - Nirmala Akula
- Genetic Basis of Mood and Anxiety Disorders, National Institute of Mental Health, National Institutes of Health, Department of Health and Human Services, Bethesda, Md, USA
| | - Raffaella Ardau
- Unit of Clinical Pharmacology, Hospital University Agency, Cagliari, Italy
| | - Elise T. Bui
- Genetic Basis of Mood and Anxiety Disorders, National Institute of Mental Health, National Institutes of Health, Department of Health and Human Services, Bethesda, Md, USA
| | - Caterina Chillotti
- Unit of Clinical Pharmacology, Hospital University Agency, Cagliari, Italy
| | - Sven Cichon
- Department of Genomics, Life and Brain Center and Institute of Human Genetics, University of Bonn, Bonn, Germany,Institute of Neurosciences and Medicine (INM-1), Research Center Juelich, Juelich, Germany
| | - Piotr Czerski
- Psychiatric Genetic Unit, Poznan University of Medical Sciences, Poznan, Poland
| | - Maria Del Zompo
- Unit of Clinical Pharmacology, Hospital University Agency, Cagliari, Italy,Department of Neuroscience ‘B.B. Brodie’, University of Cagliari, Cagliari, Italy
| | - Sevilla D. Detera-Wadleigh
- Genetic Basis of Mood and Anxiety Disorders, National Institute of Mental Health, National Institutes of Health, Department of Health and Human Services, Bethesda, Md, USA
| | - Paul Grof
- Mood Disorders Center of Ottawa, Ottawa, Ont., Canada,Department of Psychiatry, University of Toronto, Toronto, Ont., Canada,The International Group for the Study of Lithium-Treated Patients (IGSLI), Research Center Juelich, Juelich, Germany
| | - Oliver Gruber
- Department of Psychiatry and Psychotherapy, Georg-August University, Göttingen, Dresden, Germany
| | - Ryota Hashimoto
- Osaka University Graduate School of Medicine, Osaka, Japan,The Japanese Collaborative Group on the Genetics of Lithium Response in Bipolar Disorder, Research Center Juelich, Juelich, Germany
| | - Joanna Hauser
- Psychiatric Genetic Unit, Poznan University of Medical Sciences, Poznan, Poland
| | - Rebecca Hoban
- Department of Psychiatry, University of California San Diego, USA,Department of Psychiatry, VA San Diego Healthcare System, La Jolla, Calif, USA
| | - Nakao Iwata
- Department of Psychiatry, Fujita Health University School of Medicine, Toyoake, Japan,The Japanese Collaborative Group on the Genetics of Lithium Response in Bipolar Disorder, Research Center Juelich, Juelich, Germany
| | - Layla Kassem
- Genetic Basis of Mood and Anxiety Disorders, National Institute of Mental Health, National Institutes of Health, Department of Health and Human Services, Bethesda, Md, USA
| | - Tadafumi Kato
- Laboratory for Molecular Dynamics of Mental Disorders, RIKEN Brain Science Institute, Saitama, Japan,The Japanese Collaborative Group on the Genetics of Lithium Response in Bipolar Disorder, Research Center Juelich, Juelich, Germany
| | - Sarah Kittel-Schneider
- Department of Psychiatry, Psychosomatics, and Psychotherapy, University of Würzburg, Würzburg, Dresden, Germany
| | - Sebastian Kliwicki
- Department of Adult Psychiatry, Poznan University of Medical Sciences, Poznan, Poland
| | - John R. Kelsoe
- Department of Psychiatry, University of California San Diego, USA,Department of Psychiatry, VA San Diego Healthcare System, La Jolla, Calif, USA
| | - Ichiro Kusumi
- Department of Psychiatry, Hokkaido University Graduate School of Medicine, Sapporo, Japan,The Japanese Collaborative Group on the Genetics of Lithium Response in Bipolar Disorder, Research Center Juelich, Juelich, Germany
| | - Gonzalo Laje
- Genetic Basis of Mood and Anxiety Disorders, National Institute of Mental Health, National Institutes of Health, Department of Health and Human Services, Bethesda, Md, USA
| | - Susan G. Leckband
- Department of Psychiatry, University of California San Diego, USA,Department of Pharmacy, VA San Diego Healthcare System, La Jolla, Calif, USA,Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, Calif, USA
| | - Mirko Manchia
- Department of Neuroscience ‘B.B. Brodie’, University of Cagliari, Cagliari, Italy
| | - Glenda MacQueen
- Department of Psychiatry, University of Calgary, Calgary, Alta., Canada
| | - Takuya Masui
- Department of Psychiatry, Hokkaido University Graduate School of Medicine, Sapporo, Japan,The Japanese Collaborative Group on the Genetics of Lithium Response in Bipolar Disorder, Research Center Juelich, Juelich, Germany
| | - Norio Ozaki
- Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Japan,The Japanese Collaborative Group on the Genetics of Lithium Response in Bipolar Disorder, Research Center Juelich, Juelich, Germany
| | - Roy H. Perlis
- Department of Psychiatry, Massachusetts General Hospital and Harvard Medical School, Boston, Mass., USA
| | - Andrea Pfennig
- Genetic Basis of Mood and Anxiety Disorders, National Institute of Mental Health, National Institutes of Health, Department of Health and Human Services, Bethesda, Md, USA,The International Group for the Study of Lithium-Treated Patients (IGSLI), Research Center Juelich, Juelich, Germany
| | - Paola Piccardi
- Department of Neuroscience ‘B.B. Brodie’, University of Cagliari, Cagliari, Italy
| | - Sara Richardson
- Genetic Basis of Mood and Anxiety Disorders, National Institute of Mental Health, National Institutes of Health, Department of Health and Human Services, Bethesda, Md, USA
| | - Guy Rouleau
- CHU Sainte-Justine Research Center, Department of Medicine, University of Montreal, Que, Canada
| | - Andreas Reif
- Department of Psychiatry, Psychosomatics, and Psychotherapy, University of Würzburg, Würzburg, Dresden, Germany
| | - Janusz K. Rybakowski
- Department of Adult Psychiatry, Poznan University of Medical Sciences, Poznan, Poland,The International Group for the Study of Lithium-Treated Patients (IGSLI), Research Center Juelich, Juelich, Germany
| | - Johanna Sasse
- Department of Psychiatry and Psychotherapy, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany,The International Group for the Study of Lithium-Treated Patients (IGSLI), Research Center Juelich, Juelich, Germany
| | - Johannes Schumacher
- Genetic Basis of Mood and Anxiety Disorders, National Institute of Mental Health, National Institutes of Health, Department of Health and Human Services, Bethesda, Md, USA,Department of Genomics, Life and Brain Center and Institute of Human Genetics, University of Bonn, Bonn, Germany
| | - Giovanni Severino
- Department of Neuroscience ‘B.B. Brodie’, University of Cagliari, Cagliari, Italy
| | - Jordan W. Smoller
- Department of Psychiatry, Massachusetts General Hospital and Harvard Medical School, Boston, Mass., USA
| | - Alessio Squassina
- Department of Neuroscience ‘B.B. Brodie’, University of Cagliari, Cagliari, Italy
| | - Gustavo Turecki
- Department of Psychiatry, Douglas Hospital Research Institute, McGill University, Montreal, Que, Canada
| | - L. Trevor Young
- Department of Psychiatry, University of British Columbia, Vancouver, B.C., Canada,The International Group for the Study of Lithium-Treated Patients (IGSLI), Research Center Juelich, Juelich, Germany
| | - Takeo Yoshikawa
- Laboratory for Molecular Psychiatry, RIKEN Brain Science Institute, Saitama, Japan,The Japanese Collaborative Group on the Genetics of Lithium Response in Bipolar Disorder, Research Center Juelich, Juelich, Germany
| | - Michael Bauer
- Department of Psychiatry and Psychotherapy, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany,The International Group for the Study of Lithium-Treated Patients (IGSLI), Research Center Juelich, Juelich, Germany
| | - Francis J. McMahon
- Genetic Basis of Mood and Anxiety Disorders, National Institute of Mental Health, National Institutes of Health, Department of Health and Human Services, Bethesda, Md, USA
| |
Collapse
|
10
|
Abstract
Modulators of G protein-coupled receptors (GPCRs) form a key area for the pharmaceutical industry, representing approximately 27% of all Food and Drug Administration (FDA)-approved drugs. Consequently, there are a wide variety of in vitro plate-based screening technologies that enable the measurement of compound affinity, potency, and efficacy for almost every type of GPCR. However, to maximize success it is prudent to ensure that (i) the most suitable assay formats are identified, (ii) they are configured optimally to detect the desired compound activity, and (iii) that they form a basis for predicting clinical effects. To achieve this, an understanding of the pathways and mechanisms of receptor activation relevant to the disease mechanism, as well as the benefits and/or limitations of the specific techniques, is key.
Collapse
|
11
|
Kelly P, Zhou Y, Whitehead J, Stallard N, Bowman C. Sequentially testing for a gene-drug interaction in a genomewide analysis. Stat Med 2008; 27:2022-34. [PMID: 17979181 DOI: 10.1002/sim.3059] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Assaying a large number of genetic markers from patients in clinical trials is now possible in order to tailor drugs with respect to efficacy. The statistical methodology for analysing such massive data sets is challenging. The most popular type of statistical analysis is to use a univariate test for each genetic marker, once all the data from a clinical study have been collected. This paper presents a sequential method for conducting an omnibus test for detecting gene-drug interactions across the genome, thus allowing informed decisions at the earliest opportunity and overcoming the multiple testing problems from conducting many univariate tests. We first propose an omnibus test for a fixed sample size. This test is based on combining F-statistics that test for an interaction between treatment and the individual single nucleotide polymorphism (SNP). As SNPs tend to be correlated, we use permutations to calculate a global p-value. We extend our omnibus test to the sequential case. In order to control the type I error rate, we propose a sequential method that uses permutations to obtain the stopping boundaries. The results of a simulation study show that the sequential permutation method is more powerful than alternative sequential methods that control the type I error rate, such as the inverse-normal method. The proposed method is flexible as we do not need to assume a mode of inheritance and can also adjust for confounding factors. An application to real clinical data illustrates that the method is computationally feasible for a large number of SNPs.
Collapse
Affiliation(s)
- Patrick Kelly
- School of Public Health, The University of Sydney, Sydney, NSW 2006, Australia.
| | | | | | | | | |
Collapse
|
12
|
Grossman I. Routine pharmacogenetic testing in clinical practice: dream or reality? Pharmacogenomics 2007; 8:1449-59. [DOI: 10.2217/14622416.8.10.1449] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Pharmacogenetics (PGx) has become progressively popular in recent years, thanks to growing anticipation among scientists, healthcare providers and the general public for the incorporation of genetic tests into the diagnostic arsenal at the physician’s disposal. Indeed, much research has been dedicated to elucidation of genetic determinants underlying interindividual variability in pharmacokinetic parameters, as well as drug safety and efficacy. However, few PGx applications have thus far been realized in healthcare management. This review uses examples from PGx research of psychiatric drugs to illustrate why the current published findings are inadequate and insufficient for utilization as routine clinical predictors of treatment safety, efficacy or dosing. I therefore suggest the necessary steps to demonstrate the validity, utility and cost–effectiveness of PGx. These recommendations include a whole range of aspects, starting from standardization of criteria and assessment of the technical quality of genotyping assays, up to design of prospective PGx studies, providing the basis for reimbursement programs to be recognized in routine clinical practice.
Collapse
Affiliation(s)
- Iris Grossman
- GlaxoSmithKline, Pharmacogenetics, Research and Development, 5 Moore Drive, Research Triangle Park, Durham 27709, NC, USA
- Duke University, IGSP Center for Population Genomics and Pharmacogenetics, Durham, NC, USA
| |
Collapse
|
13
|
Abstract
Personalized medicine introduces the promise to use molecular markers that signal the risk of disease or its presence before clinical signs and symptoms appear. This information underlies a new healthcare strategy focused on prevention and early intervention, rather than reaction to advanced stages of disease. Such a strategy can delay disease onset or minimize symptom severity. The molecular foundations that enable personalized medicine include detection of variation in nucleotide sequence of genes and in characteristic patterns of gene expression, proteins and metabolites. Genetic and molecular patterns are correlated with disease manifestations, drug responses, treatment prognosis, or prediction of predisposition to future disease states. However, the uncertainties for personalized medicine are considerable, including economic, ethical, legal, and societal questions. Although much of its promise remains unproven to date, the foundations of personalized medicine appear solid and evidence is accumulating rapidly pointing to its growing importance in healthcare (Fig. 1).
Collapse
Affiliation(s)
- Erwin P Bottinger
- Charles Bronfman Institute for Personalized Medicine, Department of Medicine, Mount Sinai School of Medicine, New York, New York 10029, USA.
| |
Collapse
|
14
|
Wheeler J, McHale M, Jackson V, Penny M. Assessing Theoretical Risk and Benefit suggested by Genetic Association Studies of CCR5: Experience in a Drug Development Programme for Maraviroc. Antivir Ther 2007. [DOI: 10.1177/135965350701200208] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
The proliferation of published gene association studies of the CCR5A32 mutation is of relevance to drug development of a CCR5 antagonist for HIV, in highlighting potential safety concerns. We conducted an initial review of all non-HIV gene association studies of CCR5-Δ32, followed by detailed meta-analyses in the three disease areas most commonly reported. Our review indicated no consistent evidence of increased risk of susceptibility to hepatitis C virus infection or multiple sclerosis among individuals with CCR5-Δ32 mutation, and suggested treatment with a CCR5 inhibitor is unlikely to have related adverse effects. There was, however, evidence to suggest rheumatoid arthritis as a potential therapeutic target for a CCR5 antagonist. Clinical evidence would be required to confirm these findings.
Collapse
Affiliation(s)
| | - Mary McHale
- Pfizer Research and Development, Sandwich, Kent, UK
| | | | | |
Collapse
|
15
|
Sams-Dodd F. Strategies to optimize the validity of disease models in the drug discovery process. Drug Discov Today 2007; 11:355-63. [PMID: 16580978 DOI: 10.1016/j.drudis.2006.02.005] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2005] [Revised: 12/06/2005] [Accepted: 02/17/2006] [Indexed: 11/23/2022]
Abstract
Models of human diseases are necessary for experimental research into the biological basis of disease and for the development of treatments. They have an enormous impact upon the success of biomedical research. However, in spite of this, a consistent system for evaluating, expressing and comparing the clinical validity of disease models is not available. The purpose of this paper is, therefore, to provide a theoretical discussion of the concepts behind disease models and to develop a terminology and a framework to analyze and express the clinical validity of disease models.
Collapse
Affiliation(s)
- Frank Sams-Dodd
- Bionomics Europe, Les Algorithmes, rue Jean Sapidus, Parc d'Innovation, 67400 Illkirch, France.
| |
Collapse
|
16
|
McConnell O, Bach A, Balibar C, Byrne N, Cai Y, Carter G, Chlenov M, Di L, Fan K, Goljer I, He Y, Herold D, Kagan M, Kerns E, Koehn F, Kraml C, Marathias V, Marquez B, McDonald L, Nogle L, Petucci C, Schlingmann G, Tawa G, Tischler M, Williamson RT, Sutherland A, Watts W, Young M, Zhang MY, Zhang Y, Zhou D, Ho D. Enantiomeric separation and determination of absolute stereochemistry of asymmetric molecules in drug discovery—Building chiral technology toolboxes. Chirality 2007; 19:658-82. [PMID: 17390370 DOI: 10.1002/chir.20399] [Citation(s) in RCA: 82] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
The application of Chiral Technology, or the (extensive) use of techniques or tools for the determination of absolute stereochemistry and the enantiomeric or chiral separation of racemic small molecule potential lead compounds, has been critical to successfully discovering and developing chiral drugs in the pharmaceutical industry. This has been due to the rapid increase over the past 10-15 years in potential drug candidates containing one or more asymmetric centers. Based on the experiences of one pharmaceutical company, a summary of the establishment of a Chiral Technology toolbox, including the implementation of known tools as well as the design, development, and implementation of new Chiral Technology tools, is provided.
Collapse
Affiliation(s)
- Oliver McConnell
- Wyeth Research, Chemical and Screening Sciences, Collegeville, PA 19426, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Günther S, Senger C, Michalsky E, Goede A, Preissner R. Representation of target-bound drugs by computed conformers: implications for conformational libraries. BMC Bioinformatics 2006; 7:293. [PMID: 16764718 PMCID: PMC1523373 DOI: 10.1186/1471-2105-7-293] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2006] [Accepted: 06/09/2006] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The increasing number of known protein structures provides valuable information about pharmaceutical targets. Drug binding sites are identifiable and suitable lead compounds can be proposed. The flexibility of ligands is a critical point for the selection of potential drugs. Since computed 3D structures of millions of compounds are available, the knowledge of their binding conformations would be a great benefit for the development of efficient screening methods. RESULTS Integration of two public databases allowed superposition of conformers for 193 approved drugs with 5507 crystallised target-bound counterparts. The generation of 9600 drug conformers using an atomic force field was carried out to obtain an optimal coverage of the conformational space. Bioactive conformations are best described by a conformational ensemble: half of all drugs exhibit multiple active states, distributed over the entire range of the reachable energy and conformational space.A number of up to 100 conformers per drug enabled us to reproduce the bound states within a similarity threshold of 1.0 angstroms in 70% of all cases. This fraction rises to about 90% for smaller or average sized drugs. CONCLUSION Single drugs adopt multiple bioactive conformations if they interact with different target proteins. Due to the structural diversity of binding sites they adopt conformations that are distributed over a broad conformational space and wide energy range. Since the majority of drugs is well represented by a predefined low number of conformers (up to 100) this procedure is a valuable method to compare compounds by three-dimensional features or for fast similarity searches starting with pharmacophores. The underlying 9600 generated drug conformers are downloadable from the Super Drug Web site 1. All superpositions are visualised at the same source. Additional conformers (110,000) of 2400 classified WHO-drugs are also available.
Collapse
Affiliation(s)
- Stefan Günther
- Institute for Biochemistry, Charité, Monbijoustr. 2, 10117 Berlin, Germany
| | - Christian Senger
- Institute for Biochemistry, Charité, Monbijoustr. 2, 10117 Berlin, Germany
| | - Elke Michalsky
- Institute for Biochemistry, Charité, Monbijoustr. 2, 10117 Berlin, Germany
| | - Andrean Goede
- Institute for Biochemistry, Charité, Monbijoustr. 2, 10117 Berlin, Germany
| | - Robert Preissner
- Institute for Biochemistry, Charité, Monbijoustr. 2, 10117 Berlin, Germany
| |
Collapse
|
18
|
Ginsburg GS, Angrist M. The future may be closer than you think: a response from the Personalized Medicine Coalition to the Royal Society's report on personalized medicine. Per Med 2006; 3:119-123. [DOI: 10.2217/17410541.3.2.119] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
A recent report from the British Royal Society on the prospects for personalized medicine provides a sobering assessment of the field and its prospects. The report contends that pharmacogenetics has little clinical relevance at the moment and will only progress with the completion of large, cumbersome clinical trials. The report goes on to note that the regulatory infrastructure, medical education initiatives and public deliberation necessary to make personalized medicine a reality are essentially nonexistent, at least so far. In our view, personalized medicine is much more than a hypothetical protocol designed to correlate genotypes with prescriptions. We argue that the development of personalized medicine is a broader phenomenon that is already being practiced in one form or another in many contexts. Both academic medicine and the pharmaceutical industry have a huge stake in bringing pharmacogenetic-based personalized medicine to fruition; we expect both entities to act as drivers of what will be a long-term, iterative process.
Collapse
Affiliation(s)
- Geoffrey S Ginsburg
- Duke University, Duke Institute for Genome Sciences & Policy, Durham, North Carolina, USA
- Personalized Medicine Coalition, 1401 H Street, NW Suite 650, Washington DC 20005, USA
| | - Misha Angrist
- Duke University, Duke Institute for Genome Sciences & Policy, Durham, North Carolina, USA
| |
Collapse
|
19
|
Abstract
The discipline of molecular biology has become increasingly important in recent times for the process of drug discovery. We describe the impact of molecular biology across the whole process of drug discovery and development, including (i) the identification and validation of new drug targets, (ii) the development of molecular screens to find new candidate drugs, and (iii) the generation of safety data and competences leading to enhanced clinical efficacy. We also speculate on emerging developments in drug discovery where it seems likely that molecular biology will play an even more vital role in the generation of future therapies.
Collapse
|
20
|
Bol D, Ebner R. Gene expression profiling in the discovery, optimization and development of novel drugs: one universal screening platform. Pharmacogenomics 2006; 7:227-35. [PMID: 16515402 DOI: 10.2217/14622416.7.2.227] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
With recent advances in robotics and high-content screening and analysis methods, transcriptional profiling can now be utilized as a comprehensive forward chemical genomics platform for drug discovery, lead selection and lead optimization. It can be used to define the state of a cell on the basis of gene networks, and to search for drugs that can shift cellular states in a manner predicted at the genome level to be therapeutically beneficial. The treatment of cells with compounds produces transcriptional 'fingerprints' that reveal mechanism-of-action, and enable discrimination between individual compounds based on drug behaviors important to all phases of drug discovery and development.
Collapse
Affiliation(s)
- David Bol
- Avalon Pharmaceuticals, Inc., 20358 Seneca Meadows Parkway, Germantown, MD 20876, USA.
| | | |
Collapse
|
21
|
&NA;. Pharmacogenomics has promising applications in a number of areas of the drug discovery pipeline. DRUGS & THERAPY PERSPECTIVES 2005. [DOI: 10.2165/00042310-200521110-00009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
|
22
|
Imbeaud S, Auffray C. 'The 39 steps' in gene expression profiling: critical issues and proposed best practices for microarray experiments. Drug Discov Today 2005; 10:1175-82. [PMID: 16182210 DOI: 10.1016/s1359-6446(05)03565-8] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Gene expression microarrays have been used widely to address increasingly complex biological questions and to produce an unprecedented amount of data, but have yet to realize their full potential. The interpretation of microarray data remains a major challenge because of the complexity of the underlying biological networks. To gather meaningful expression data, it is crucial to develop standardized approaches for vigilant study design, controlled annotation of resources, careful quality control of experiments, robust statistics, and data registration and storage. This article reviews the steps needed in the design and execution of valid microarray experiments so that global gene expression data can play a major role in the pursuit of future biological discoveries that will impact drug development.
Collapse
Affiliation(s)
- Sandrine Imbeaud
- Array s/IMAGE, Genexpress, Functional Genomics and Systems Biology for Health, LGN UMR 7091, CNRS and Pierre and Marie Curie University, Paris VI, Villejuif, France.
| | | |
Collapse
|
23
|
Fitzgerald GA. Anticipating change in drug development: the emerging era of translational medicine and therapeutics. Nat Rev Drug Discov 2005; 4:815-8. [PMID: 16224453 DOI: 10.1038/nrd1849] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Despite the large investments made in drug discovery in the past decade, there is still a dearth of new drugs. This highlights the persistence of a model of drug development that has not adapted to changes in science, public perception of drug companies or the marketplace. A high profit margin in the United States has shielded drug development from the usual economic pressures that would ordinarily drive reform. The strategy of merger, pursued by many companies to compensate for the failure to develop new drugs, has, in most cases, compounded the problem, imposing geographic and cultural segregation on an already inefficient process.
Collapse
Affiliation(s)
- Garret A Fitzgerald
- The Institute for Translational Medicine and Therapeutics, Room 805, Biomedical Research Building 2/3, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA.
| |
Collapse
|