51
|
Cheng F, Desai RJ, Handy DE, Wang R, Schneeweiss S, Barabási AL, Loscalzo J. Network-based approach to prediction and population-based validation of in silico drug repurposing. Nat Commun 2018; 9:2691. [PMID: 30002366 PMCID: PMC6043492 DOI: 10.1038/s41467-018-05116-5] [Citation(s) in RCA: 336] [Impact Index Per Article: 48.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Accepted: 06/08/2018] [Indexed: 12/21/2022] Open
Abstract
Here we identify hundreds of new drug-disease associations for over 900 FDA-approved drugs by quantifying the network proximity of disease genes and drug targets in the human (protein–protein) interactome. We select four network-predicted associations to test their causal relationship using large healthcare databases with over 220 million patients and state-of-the-art pharmacoepidemiologic analyses. Using propensity score matching, two of four network-based predictions are validated in patient-level data: carbamazepine is associated with an increased risk of coronary artery disease (CAD) [hazard ratio (HR) 1.56, 95% confidence interval (CI) 1.12–2.18], and hydroxychloroquine is associated with a decreased risk of CAD (HR 0.76, 95% CI 0.59–0.97). In vitro experiments show that hydroxychloroquine attenuates pro-inflammatory cytokine-mediated activation in human aortic endothelial cells, supporting mechanistically its potential beneficial effect in CAD. In summary, we demonstrate that a unique integration of protein-protein interaction network proximity and large-scale patient-level longitudinal data complemented by mechanistic in vitro studies can facilitate drug repurposing. Repurposing approved drugs could accelerate treatment options for various diseases. Here, the authors use network proximity of disease gene products and drug targets in the human protein interactome to identify drug-disease associations for cardiovascular disease, and validate these using longitudinal healthcare data.
Collapse
Affiliation(s)
- Feixiong Cheng
- Center for Complex Networks Research and Department of Physics, Northeastern University, Boston, MA, 02115, USA.,Center for Cancer Systems Biology and Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA
| | - Rishi J Desai
- Division of Pharmacoepidemiology and Pharmacoeconomics, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Diane E Handy
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Ruisheng Wang
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Sebastian Schneeweiss
- Division of Pharmacoepidemiology and Pharmacoeconomics, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Albert-László Barabási
- Center for Complex Networks Research and Department of Physics, Northeastern University, Boston, MA, 02115, USA.,Center for Cancer Systems Biology and Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA.,Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA.,Center for Network Science, Central European University, Budapest, 1051, Hungary
| | - Joseph Loscalzo
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA.
| |
Collapse
|
52
|
Jiang A, Jegga AG. Characterizing drug-related adverse events by joint analysis of biomedical and genomic data: A case study of drug-induced pulmonary fibrosis. AMIA JOINT SUMMITS ON TRANSLATIONAL SCIENCE PROCEEDINGS. AMIA JOINT SUMMITS ON TRANSLATIONAL SCIENCE 2018; 2017:91-97. [PMID: 29888048 PMCID: PMC5961825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Spontaneous reporting systems such as the FDA's adverse event reporting system (FAERS) present a great resource to mine for and analyze real-world medication usage. Our study is based on a central premise that FAERS captures unsuspected drug-related adverse events (AEs). Since drug-related AEs result for several reasons, no single approach will be able to predict the entire gamut of AEs. A fundamental premise of systems biology is that a full understanding of a biological process or phenotype (e.g., drug-related AE) requires that all the individual elements be studied in conjunction with one another. We therefore hypothesize that integrative analysis of FAERS-based drug-related AEs with the transcriptional signatures from disease models and drug treatments can lead to the generation of unbiased hypotheses for drug-induced AE-modulating mechanisms of action as well as drug combinations that may target those mechanisms. We test this hypothesis using drug-induced pulmonary fibrosis (DIPF) as a proof-of-concept study.
Collapse
Affiliation(s)
- Alex Jiang
- Division of Biomedical Informatics, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA,Comell University, Ithaca, New York, USA
| | - Anil G. Jegga
- Division of Biomedical Informatics, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA,Department of Computer Science, University of Cincinnati College of Engineering, Cincinnati, Ohio, USA
| |
Collapse
|
53
|
Toki T, Ono S. Spontaneous Reporting on Adverse Events by Consumers in the United States: An Analysis of the Food and Drug Administration Adverse Event Reporting System Database. Drugs Real World Outcomes 2018; 5:117-128. [PMID: 29725886 PMCID: PMC5984610 DOI: 10.1007/s40801-018-0134-0] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022] Open
Abstract
BACKGROUND Voluntary reports on adverse events (AEs) submitted by consumers have been facilitated through the MedWatch program in the United States (US), but few studies have described the characteristics of voluntary reports. OBJECTIVE The aim of this study was to reveal the characteristics of current voluntary reports on AEs reported by consumers and healthcare professionals. METHODS We performed analysis on voluntary reports of AEs in the US Food and Drug Administration AE Reporting System (FAERS) database submitted in 2016. We compared reports by consumers with those by healthcare professionals. RESULTS The number of voluntary reports by consumers has increased since 2013 in the US. Reports by consumers were different from ones by health professionals in several important aspects such as demographics and outcomes of patients, AEs, and suspect drugs. The proportion of reports on female patients and on disability as a patient outcome were higher in reports by consumers than in those by healthcare professionals. Consumers more frequently reported concomitant drugs compared with healthcare professionals. Time to report varied among the occupations and depending on seriousness of outcomes. CONCLUSIONS Our analysis of voluntary AE reports in the US FAERS database has shown that voluntary reports tended to include AEs related to subjective symptoms, as in some previous studies on patient reporting in the EU. Voluntary reports by consumers seemed to be different from ones by healthcare professionals in important aspects including demographics and reporting behaviors. These findings suggest that the heterogeneities should be addressed appropriately in using spontaneous reports.
Collapse
Affiliation(s)
- Tadashi Toki
- Laboratory of Pharmaceutical Regulation and Sciences, Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Shunsuke Ono
- Laboratory of Pharmaceutical Regulation and Sciences, Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan.
| |
Collapse
|
54
|
Abstract
Pharmaceutical cocrystals belong to a sub-class of cocrystals wherein one of the components is a drug molecule (or an active pharmaceutical ingredient, API) and the second is a benign food or drug grade additive (generally regarded as safe, GRAS). The two components are hydrogen-bonded in a fixed stoichiometric ratio in the crystal lattice. In the past decade, pharmaceutical cocrystals have demonstrated significant promise in their ability to modify the physicochemical and pharmacokinetic properties of drug substances, such as the solubility and dissolution rate, bioavailability, particle morphology and size, tableting and compaction, melting point, physical form, biochemical and hydration stability, and permeability. In this feature review, we highlight some prominent examples of drug cocrystals which exhibit variable hardness/softness and elasticity/plasticity depending on coformer selection, improvement of solubility and permeability in the same cocrystal, increase of the melting point for solid formulation, enhanced color performance, photostability and hydration stability, and a longer half-life. Cocrystals of flavanoids and polyphenols can make improved pharmaceuticals and also extend to the larger class of nutraceuticals. The application of crystal engineering to assemble ternary cocrystals expands this field to drug-drug cocrystals which may be useful in multi-drug resistance, mitigating side effects of drugs, or attenuating/enhancing drug action synergistically by rational selection. The advent of new techniques for structural characterization beyond the standard X-ray diffraction will provide a better understanding of drug phases which are at the borderline of crystalline-amorphous nature and even newer opportunities in the future.
Collapse
Affiliation(s)
- Geetha Bolla
- School of Chemistry, University of Hyderabad, Prof. C. R. Rao Road, Gachibowli, Hyderabad 500 046, India.
| | - Ashwini Nangia
- School of Chemistry, University of Hyderabad, Prof. C. R. Rao Road, Gachibowli, Hyderabad 500 046, India. and CSIR-National Chemical Laboratory, Dr. Homi Bhabha Road, Pune 411 008, India.
| |
Collapse
|
55
|
Sarangdhar M, Tabar S, Schmidt C, Kushwaha A, Shah K, Dahlquist JE, Jegga AG, Aronow BJ. Data mining differential clinical outcomes associated with drug regimens using adverse event reporting data. Nat Biotechnol 2018; 34:697-700. [PMID: 27404875 DOI: 10.1038/nbt.3623] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Affiliation(s)
- Mayur Sarangdhar
- Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Scott Tabar
- Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Charles Schmidt
- Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Akash Kushwaha
- Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Krish Shah
- Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Jeanine E Dahlquist
- Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Anil G Jegga
- Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Bruce J Aronow
- Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA.,Clinical and Translational Sciences Program, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| |
Collapse
|
56
|
Mechanick JI, Zhao S, Garvey WT. Leptin, An Adipokine With Central Importance in the Global Obesity Problem. Glob Heart 2017; 13:113-127. [PMID: 29248361 DOI: 10.1016/j.gheart.2017.10.003] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Accepted: 10/25/2017] [Indexed: 02/08/2023] Open
Abstract
Leptin has central importance in the global obesity and cardiovascular disease problem. Leptin is principally secreted by adipocytes and acts in the hypothalamus to suppress appetite and food intake, increase energy expenditure, and regulate body weight. Based on clinical translation of specific and networked actions, leptin affects the cardiovascular system and may be a marker and driver of cardiometabolic risk factors with interventions that are actionable by cardiologists. Leptin subnetwork analysis demonstrates a statistically significant role for ethnoculturally and socioeconomically appropriate lifestyle intervention in cardiovascular disease. Emergent mechanistic components and potential diagnostic or therapeutic targets include hexokinase 3, urocortins, clusterin, sialic acid-binding immunoglobulin-like lectin 6, C-reactive protein, platelet glycoprotein VI, albumin, pentraxin 3, ghrelin, obestatin prepropeptide, leptin receptor, neuropeptide Y, and corticotropin-releasing factor receptor 1. Emergent associated symptoms include weight change, eating disorders, vascular necrosis, chronic fatigue, and chest pain. Leptin-targeted therapies are reported for lipodystrophy and leptin deficiency, but they are investigational for leptin resistance, obesity, and other chronic diseases.
Collapse
Affiliation(s)
- Jeffrey I Mechanick
- Division of Cardiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Division of Endocrinology, Diabetes, and Bone Disease, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| | - Shan Zhao
- Basepaws Inc., Redondo Beach, CA, USA
| | - W Timothy Garvey
- Department of Nutritional Sciences and Diabetes Research Center, University of Alabama at Birmingham, Birmingham, AL, USA; Geriatric Research Education and Clinical Center, Birmingham Veterans Affairs Medical Center, Birmingham, AL, USA
| |
Collapse
|
57
|
Blucher AS, Choonoo G, Kulesz-Martin M, Wu G, McWeeney SK. Evidence-Based Precision Oncology with the Cancer Targetome. Trends Pharmacol Sci 2017; 38:1085-1099. [PMID: 28964549 PMCID: PMC5759325 DOI: 10.1016/j.tips.2017.08.006] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Revised: 08/17/2017] [Accepted: 08/29/2017] [Indexed: 11/16/2022]
Abstract
A core tenet of precision oncology is the rational choice of drugs to interact with patient-specific biological targets of interest, but it is currently difficult for researchers to obtain consistent and well-supported target information for pharmaceutical drugs. We review current drug-target interaction resources and critically assess how supporting evidence is handled. We introduce the concept of a unified Cancer Targetome to aggregate drug-target interactions in an evidence-based framework. We discuss current unmet needs and the implications for evidence-based clinical omics. The focus of this review is precision oncology but the discussion is highly relevant to targeted therapies in any area.
Collapse
Affiliation(s)
- Aurora S Blucher
- Division of Bioinformatics and Computational Biology, Department of Medical Informatics and Clinical Epidemiology, Oregon Health & Science University, Portland, OR, USA
| | - Gabrielle Choonoo
- Division of Bioinformatics and Computational Biology, Department of Medical Informatics and Clinical Epidemiology, Oregon Health & Science University, Portland, OR, USA; Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
| | - Molly Kulesz-Martin
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA; Department of Dermatology and Department of Cell and Developmental Biology, Oregon Health & Science University, Portland, OR, USA
| | - Guanming Wu
- Division of Bioinformatics and Computational Biology, Department of Medical Informatics and Clinical Epidemiology, Oregon Health & Science University, Portland, OR, USA
| | - Shannon K McWeeney
- Division of Bioinformatics and Computational Biology, Department of Medical Informatics and Clinical Epidemiology, Oregon Health & Science University, Portland, OR, USA; Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA.
| |
Collapse
|
58
|
Benson HE, Watterson S, Sharman JL, Mpamhanga CP, Parton A, Southan C, Harmar AJ, Ghazal P. Is systems pharmacology ready to impact upon therapy development? A study on the cholesterol biosynthesis pathway. Br J Pharmacol 2017; 174:4362-4382. [PMID: 28910500 PMCID: PMC5715582 DOI: 10.1111/bph.14037] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Revised: 08/10/2017] [Accepted: 08/30/2017] [Indexed: 12/22/2022] Open
Abstract
Background and Purpose An ever‐growing wealth of information on current drugs and their pharmacological effects is available from online databases. As our understanding of systems biology increases, we have the opportunity to predict, model and quantify how drug combinations can be introduced that outperform conventional single‐drug therapies. Here, we explore the feasibility of such systems pharmacology approaches with an analysis of the mevalonate branch of the cholesterol biosynthesis pathway. Experimental Approach Using open online resources, we assembled a computational model of the mevalonate pathway and compiled a set of inhibitors directed against targets in this pathway. We used computational optimization to identify combination and dose options that show not only maximal efficacy of inhibition on the cholesterol producing branch but also minimal impact on the geranylation branch, known to mediate the side effects of pharmaceutical treatment. Key Results We describe serious impediments to systems pharmacology studies arising from limitations in the data, incomplete coverage and inconsistent reporting. By curating a more complete dataset, we demonstrate the utility of computational optimization for identifying multi‐drug treatments with high efficacy and minimal off‐target effects. Conclusion and Implications We suggest solutions that facilitate systems pharmacology studies, based on the introduction of standards for data capture that increase the power of experimental data. We propose a systems pharmacology workflow for the refinement of data and the generation of future therapeutic hypotheses.
Collapse
Affiliation(s)
- Helen E Benson
- Centre for Integrative Physiology, University of Edinburgh, Edinburgh, UK
| | - Steven Watterson
- Northern Ireland Centre for Stratified Medicine, University of Ulster, C-Tric, Derry, UK
| | - Joanna L Sharman
- Centre for Integrative Physiology, University of Edinburgh, Edinburgh, UK
| | - Chido P Mpamhanga
- Centre for Cardiovascular Science, University of Edinburgh, The Queen's Medical Research Institute, Edinburgh, UK
| | - Andrew Parton
- Northern Ireland Centre for Stratified Medicine, University of Ulster, C-Tric, Derry, UK
| | | | - Anthony J Harmar
- Centre for Cardiovascular Science, University of Edinburgh, The Queen's Medical Research Institute, Edinburgh, UK
| | - Peter Ghazal
- Division of Infection and Pathway Medicine, University of Edinburgh Medical School, Edinburgh, UK.,Centre for Synthetic and Systems Biology, CH Waddington Building, King's Buildings, Edinburgh, UK
| |
Collapse
|
59
|
Xu D, Ham AG, Tivis RD, Caylor ML, Tao A, Flynn ST, Economen PJ, Dang HK, Johnson RW, Culbertson VL. MSBIS: A Multi-Step Biomedical Informatics Screening Approach for Identifying Medications that Mitigate the Risks of Metoclopramide-Induced Tardive Dyskinesia. EBioMedicine 2017; 26:132-137. [PMID: 29191560 PMCID: PMC5832625 DOI: 10.1016/j.ebiom.2017.11.015] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Revised: 11/07/2017] [Accepted: 11/20/2017] [Indexed: 02/02/2023] Open
Abstract
In 2009 the U.S. Food and Drug Administration (FDA) placed a black box warning on metoclopramide (MCP) due to the increased risks and prevalence of tardive dyskinesia (TD). In this study, we developed a multi-step biomedical informatics screening (MSBIS) approach leveraging publicly available bioactivity and drug safety data to identify concomitant drugs that mitigate the risks of MCP-induced TD. MSBIS includes (1) TargetSearch (http://dxulab.org/software) bioinformatics scoring for drug anticholinergic activity using CHEMBL bioactivity data; (2) unadjusted odds ratio (UOR) scoring for indications of TD-mitigating effects using the FDA Adverse Event Reporting System (FAERS); (3) adjusted odds ratio (AOR) re-scoring by removing the effect of cofounding factors (age, gender, reporting year); (4) logistic regression (LR) coefficient scoring for confirming the best TD-mitigating drug candidates. Drugs with increasing TD protective potential and statistical significance were obtained at each screening step. Fentanyl is identified as the most promising drug against MCP-induced TD (coefficient: −2.68; p-value < 0.01). The discovery is supported by clinical reports that patients fully recovered from MCP-induced TD after fentanyl-induced general anesthesia. Loperamide is identified as a potent mitigating drug against a broader range of drug-induced movement disorders through pharmacokinetic modifications. Using drug-induced TD as an example, we demonstrated that MSBIS is an efficient in silico tool for unknown drug-drug interaction detection, drug repurposing, and combination therapy design. An in silico MSBIS approach was developed to identify concomitant drugs that mitigate the side effects of a primary drug. Fentanyl and loperamide were identified to protect patients against drug-induced tardive dyskinesia (TD). MSBIS can be generalized to provide rational starting points for drug repurposing and combination therapy design.
Many dopamine antagonists cause TD and other severe, irreversible movement disorders. 120 drugs were screened using the MSBIS approach, leading to the discovery of fentanyl and loperamide. When co-administered with dopamine antagonists such as metoclopramide, they may protect patients against drug-induced movement disorders including TD. Their mechanisms of toxicity-mitigating action are remarkably different. The statistical significance of the findings was established using the FDA Adverse Event Reporting System and supported by published clinical reports and pharmacologic data. This study demonstrated the feasibility of the MSBIS approach and its applicability in identifying unknown drug-drug interactions that either mitigate undesirable toxicities or synergize therapeutic effects.
Collapse
Affiliation(s)
- Dong Xu
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, Kasiska Division of Health Sciences, Idaho State University, Meridian, ID 83642, USA.
| | - Alexandrea G Ham
- College of Pharmacy, Kasiska Division of Health Sciences, Idaho State University, Meridian, ID 83642, USA
| | - Rickey D Tivis
- Idaho Center for Health Research, Kasiska Division of Health Sciences, Idaho State University, Meridian, ID 83642, USA
| | - Matthew L Caylor
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, Kasiska Division of Health Sciences, Idaho State University, Meridian, ID 83642, USA
| | - Aoxiang Tao
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, Kasiska Division of Health Sciences, Idaho State University, Meridian, ID 83642, USA
| | - Steve T Flynn
- College of Pharmacy, Kasiska Division of Health Sciences, Idaho State University, Meridian, ID 83642, USA
| | - Peter J Economen
- College of Pharmacy, Kasiska Division of Health Sciences, Idaho State University, Meridian, ID 83642, USA
| | - Hung K Dang
- College of Pharmacy, Kasiska Division of Health Sciences, Idaho State University, Meridian, ID 83642, USA
| | - Royal W Johnson
- College of Pharmacy, Kasiska Division of Health Sciences, Idaho State University, Meridian, ID 83642, USA
| | - Vaughn L Culbertson
- Department of Pharmacy Practice and Administrative Sciences, College of Pharmacy, Kasiska Division of Health Sciences, Idaho State University, Meridian, ID 83642, USA
| |
Collapse
|
60
|
Wong H, Bohnert T, Damian-Iordache V, Gibson C, Hsu CP, Krishnatry AS, Liederer BM, Lin J, Lu Q, Mettetal JT, Mudra DR, Nijsen MJ, Schroeder P, Schuck E, Suryawanshi S, Trapa P, Tsai A, Wang H, Wu F. Translational pharmacokinetic-pharmacodynamic analysis in the pharmaceutical industry: an IQ Consortium PK-PD Discussion Group perspective. Drug Discov Today 2017; 22:1447-1459. [DOI: 10.1016/j.drudis.2017.04.015] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Revised: 04/03/2017] [Accepted: 04/25/2017] [Indexed: 02/06/2023]
|
61
|
Systems pharmacology-based identification of pharmacogenomic determinants of adverse drug reactions using human iPSC-derived cell lines. ACTA ACUST UNITED AC 2017. [DOI: 10.1016/j.coisb.2017.05.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
62
|
Translational learning from clinical studies predicts drug pharmacokinetics across patient populations. NPJ Syst Biol Appl 2017. [PMID: 28649438 PMCID: PMC5460240 DOI: 10.1038/s41540-017-0012-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Early indication of late-stage failure of novel candidate drugs could be facilitated by continuous integration, assessment, and transfer of knowledge acquired along pharmaceutical development programs. We here present a translational systems pharmacology workflow that combines drug cocktail probing in a specifically designed clinical study, physiologically based pharmacokinetic modeling, and Bayesian statistics to identify and transfer (patho-)physiological and drug-specific knowledge across distinct patient populations. Our work builds on two clinical investigations, one with 103 healthy volunteers and one with 79 diseased patients from which we systematically derived physiological information from pharmacokinetic data for a reference probe drug (midazolam) at the single-patient level. Taking into account the acquired knowledge describing (patho-)physiological alterations in the patient cohort allowed the successful prediction of the population pharmacokinetics of a second, candidate probe drug (torsemide) in the patient population. In addition, we identified significant relations of the acquired physiological processes to patient metadata from liver biopsies. The presented prototypical systems pharmacology approach is a proof of concept for model-based translation across different stages of pharmaceutical development programs. Applied consistently, it has the potential to systematically improve predictivity of pharmacokinetic simulations by incorporating the results of clinical trials and translating them to subsequent studies. Physiologically based modeling together with Bayesian statistics allows the prediction of drug pharmacokinetics in specific patient populations. An interdisciplinary group of clinicians and computational scientists led by Dr. Lars Kuepfer from Bayer developed a generic workflow consisting of several consecutive learning steps where knowledge about both individual physiology as well as drug physicochemistry can be efficiently derived from plasma concentration profiles. The acquired information is then be used for the prediction of the pharmacokinetic behavior of a new drug candidate in a diseased population. This allows to simulate the variability in drug exposure virtually before starting clinical investigation in real patients in order to evaluate drug safety or efficacy through the simulation of virtual populations. Further development of this workflow could improve the safety of clinical development programs to assess the risk-benefit ratio of novel drug candidates in silico.
Collapse
|
63
|
Lopez JS, Banerji U. Combine and conquer: challenges for targeted therapy combinations in early phase trials. Nat Rev Clin Oncol 2017; 14:57-66. [PMID: 27377132 PMCID: PMC6135233 DOI: 10.1038/nrclinonc.2016.96] [Citation(s) in RCA: 258] [Impact Index Per Article: 32.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Our increasing understanding of cancer biology has led to the development of molecularly targeted anticancer drugs. The full potential of these agents has not, however, been realised, owing to the presence of de novo (intrinsic) resistance, often resulting from compensatory signalling pathways, or the development of acquired resistance in cancer cells via clonal evolution under the selective pressures of treatment. Combinations of targeted treatments can circumvent some mechanisms of resistance to yield a clinical benefit. We explore the challenges in identifying the best drug combinations and the best combination strategies, as well as the complexities of delivering these treatments to patients. Recognizing treatment-induced toxicity and the inability to use continuous pharmacodynamically effective doses of many targeted treatments necessitates creative intermittent scheduling. Serial tumour profiling and the use of parallel co-clinical trials can contribute to understanding mechanisms of resistance, and will guide the development of adaptive clinical trial designs that can accommodate hypothesis testing, in order to realize the full potential of combination therapies.
Collapse
Affiliation(s)
- Juanita S Lopez
- Drug Development Unit, The Institute of Cancer Research and The Royal Marsden NHS Foundation Trust, Sycamore House, Downs Road, London SM2 5PT, UK
| | - Udai Banerji
- Drug Development Unit, The Institute of Cancer Research and The Royal Marsden NHS Foundation Trust, Sycamore House, Downs Road, London SM2 5PT, UK
| |
Collapse
|
64
|
Rationale and strategies for formulation development of oral fixed dose combination drug products. JOURNAL OF PHARMACEUTICAL INVESTIGATION 2016. [DOI: 10.1007/s40005-016-0286-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
65
|
Abstract
Systems pharmacology aims to holistically understand mechanisms of drug actions to support drug discovery and clinical practice. Systems pharmacology modeling (SPM) is data driven. It integrates an exponentially growing amount of data at multiple scales (genetic, molecular, cellular, organismal, and environmental). The goal of SPM is to develop mechanistic or predictive multiscale models that are interpretable and actionable. The current explosions in genomics and other omics data, as well as the tremendous advances in big data technologies, have already enabled biologists to generate novel hypotheses and gain new knowledge through computational models of genome-wide, heterogeneous, and dynamic data sets. More work is needed to interpret and predict a drug response phenotype, which is dependent on many known and unknown factors. To gain a comprehensive understanding of drug actions, SPM requires close collaborations between domain experts from diverse fields and integration of heterogeneous models from biophysics, mathematics, statistics, machine learning, and semantic webs. This creates challenges in model management, model integration, model translation, and knowledge integration. In this review, we discuss several emergent issues in SPM and potential solutions using big data technology and analytics. The concurrent development of high-throughput techniques, cloud computing, data science, and the semantic web will likely allow SPM to be findable, accessible, interoperable, reusable, reliable, interpretable, and actionable.
Collapse
Affiliation(s)
- Lei Xie
- Department of Computer Science, Hunter College, The City University of New York, New York, NY 10065; .,The Graduate Center, The City University of New York, New York, NY 10016
| | - Eli J Draizen
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, Maryland 20894; .,Program in Bioinformatics, Boston University, Boston, Massachusetts 02215
| | - Philip E Bourne
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, Maryland 20894; .,Office of the Director, National Institutes of Health, Bethesda, Maryland 20894
| |
Collapse
|
66
|
Mechanick JI, Zhao S, Garvey WT. The Adipokine-Cardiovascular-Lifestyle Network. J Am Coll Cardiol 2016; 68:1785-1803. [DOI: 10.1016/j.jacc.2016.06.072] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2016] [Revised: 06/29/2016] [Accepted: 06/29/2016] [Indexed: 12/17/2022]
|
67
|
Chen Y, Palczewska G, Masuho I, Gao S, Jin H, Dong Z, Gieser L, Brooks MJ, Kiser PD, Kern TS, Martemyanov KA, Swaroop A, Palczewski K. Synergistically acting agonists and antagonists of G protein-coupled receptors prevent photoreceptor cell degeneration. Sci Signal 2016; 9:ra74. [PMID: 27460988 DOI: 10.1126/scisignal.aag0245] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Photoreceptor cell degeneration leads to visual impairment and blindness in several types of retinal disease. However, the discovery of safe and effective therapeutic strategies conferring photoreceptor cell protection remains challenging. Targeting distinct cellular pathways with low doses of different drugs that produce a functionally synergistic effect could provide a strategy for preventing or treating retinal dystrophies. We took a systems pharmacology approach to identify potential combination therapies using a mouse model of light-induced retinal degeneration. We showed that a combination of U.S. Food and Drug Administration-approved drugs that act on different G protein (heterotrimeric guanine nucleotide-binding protein)-coupled receptors (GPCRs) exhibited synergistic activity that protected retinas from light-induced degeneration even when each drug was administered at a low dose. In functional assays, the combined effects of these drugs were stimulation of Gi/o signaling by activating the dopamine receptors D2R and D4R, as well as inhibition of Gs and Gq signaling by antagonizing D1R and the α1A-adrenergic receptor ADRA1A, respectively. Moreover, transcriptome analyses demonstrated that such combined GPCR-targeted treatments preserved patterns of retinal gene expression that were more similar to those of the normal retina than did higher-dose monotherapy. Our study thus supports a systems pharmacology approach to identify treatments for retinopathies, an approach that could extend to other complex disorders.
Collapse
Affiliation(s)
- Yu Chen
- Yueyang Hospital and Clinical Research Institute of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China. Department of Pharmacology, School of Medicine, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106, USA.
| | | | - Ikuo Masuho
- Department of Neuroscience, The Scripps Research Institute, 130 Scripps Way, Jupiter, FL 33458, USA
| | - Songqi Gao
- Department of Pharmacology, School of Medicine, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106, USA
| | - Hui Jin
- Department of Pharmacology, School of Medicine, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106, USA
| | | | - Linn Gieser
- Neurobiology-Neurodegeneration and Repair Laboratory, National Eye Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Matthew J Brooks
- Neurobiology-Neurodegeneration and Repair Laboratory, National Eye Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Philip D Kiser
- Department of Pharmacology, School of Medicine, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106, USA. Research Service, Louis Stokes Cleveland VA Medical Center, Cleveland, OH 44106, USA
| | - Timothy S Kern
- Department of Pharmacology, School of Medicine, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106, USA. Research Service, Louis Stokes Cleveland VA Medical Center, Cleveland, OH 44106, USA. Department of Medicine, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Kirill A Martemyanov
- Department of Neuroscience, The Scripps Research Institute, 130 Scripps Way, Jupiter, FL 33458, USA
| | - Anand Swaroop
- Neurobiology-Neurodegeneration and Repair Laboratory, National Eye Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Krzysztof Palczewski
- Department of Pharmacology, School of Medicine, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106, USA. Polgenix Inc., Cleveland, OH 44106, USA.
| |
Collapse
|
68
|
Nagashima T, Shirakawa H, Nakagawa T, Kaneko S. Prevention of antipsychotic-induced hyperglycaemia by vitamin D: a data mining prediction followed by experimental exploration of the molecular mechanism. Sci Rep 2016; 6:26375. [PMID: 27199286 PMCID: PMC4873813 DOI: 10.1038/srep26375] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Accepted: 04/29/2016] [Indexed: 01/24/2023] Open
Abstract
Atypical antipsychotics are associated with an increased risk of hyperglycaemia, thus limiting their clinical use. This study focused on finding the molecular mechanism underlying antipsychotic-induced hyperglycaemia. First, we searched for drug combinations in the FDA Adverse Event Reporting System (FAERS) database wherein a coexisting drug reduced the hyperglycaemia risk of atypical antipsychotics, and found that a combination with vitamin D analogues significantly decreased the occurrence of quetiapine-induced adverse events relating diabetes mellitus in FAERS. Experimental validation using mice revealed that quetiapine acutely caused insulin resistance, which was mitigated by dietary supplementation with cholecalciferol. Further database analysis of the relevant signalling pathway and gene expression predicted quetiapine-induced downregulation of Pik3r1, a critical gene acting downstream of insulin receptor. Focusing on the phosphatidylinositol 3-kinase (PI3K) signalling pathway, we found that the reduced expression of Pik3r1 mRNA was reversed by cholecalciferol supplementation in skeletal muscle, and that insulin-stimulated glucose uptake into C2C12 myotube was inhibited in the presence of quetiapine, which was reversed by concomitant calcitriol in a PI3K-dependent manner. Taken together, these results suggest that vitamin D coadministration prevents antipsychotic-induced hyperglycaemia and insulin resistance by upregulation of PI3K function.
Collapse
Affiliation(s)
- Takuya Nagashima
- Department of Molecular Pharmacology, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Yoshida-Shimoadachi-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Hisashi Shirakawa
- Department of Molecular Pharmacology, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Yoshida-Shimoadachi-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Takayuki Nakagawa
- Department of Clinical Pharmacology and Therapeutics, Kyoto University Hospital, 54 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan
| | - Shuji Kaneko
- Department of Molecular Pharmacology, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Yoshida-Shimoadachi-cho, Sakyo-ku, Kyoto 606-8501, Japan
| |
Collapse
|
69
|
Large-Scale Prediction of Beneficial Drug Combinations Using Drug Efficacy and Target Profiles. J Chem Inf Model 2015; 55:2705-16. [PMID: 26624799 DOI: 10.1021/acs.jcim.5b00444] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
The identification of beneficial drug combinations is a challenging issue in pharmaceutical and clinical research toward combinatorial drug therapy. In the present study, we developed a novel computational method for large-scale prediction of beneficial drug combinations using drug efficacy and target profiles. We designed an informative descriptor for each drug-drug pair based on multiple drug profiles representing drug-targeted proteins and Anatomical Therapeutic Chemical Classification System codes. Then, we constructed a predictive model by learning a sparsity-induced classifier based on known drug combinations from the Orange Book and KEGG DRUG databases. Our results show that the proposed method outperforms the previous methods in terms of the accuracy of high-confidence predictions, and the extracted features are biologically meaningful. Finally, we performed a comprehensive prediction of novel drug combinations for 2,639 approved drugs, which predicted 142,988 new potentially beneficial drug-drug pairs. We showed several examples of successfully predicted drug combinations for a variety of diseases.
Collapse
|
70
|
Kell DB. The transporter-mediated cellular uptake of pharmaceutical drugs is based on their metabolite-likeness and not on their bulk biophysical properties: Towards a systems pharmacology. ACTA ACUST UNITED AC 2015. [DOI: 10.1016/j.pisc.2015.06.004] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
|
71
|
Boland MR, Jacunski A, Lorberbaum T, Romano JD, Moskovitch R, Tatonetti NP. Systems biology approaches for identifying adverse drug reactions and elucidating their underlying biological mechanisms. WILEY INTERDISCIPLINARY REVIEWS-SYSTEMS BIOLOGY AND MEDICINE 2015; 8:104-22. [PMID: 26559926 DOI: 10.1002/wsbm.1323] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/29/2015] [Revised: 09/30/2015] [Accepted: 10/01/2015] [Indexed: 01/06/2023]
Abstract
Small molecules are indispensable to modern medical therapy. However, their use may lead to unintended, negative medical outcomes commonly referred to as adverse drug reactions (ADRs). These effects vary widely in mechanism, severity, and populations affected, making ADR prediction and identification important public health concerns. Current methods rely on clinical trials and postmarket surveillance programs to find novel ADRs; however, clinical trials are limited by small sample size, whereas postmarket surveillance methods may be biased and inherently leave patients at risk until sufficient clinical evidence has been gathered. Systems pharmacology, an emerging interdisciplinary field combining network and chemical biology, provides important tools to uncover and understand ADRs and may mitigate the drawbacks of traditional methods. In particular, network analysis allows researchers to integrate heterogeneous data sources and quantify the interactions between biological and chemical entities. Recent work in this area has combined chemical, biological, and large-scale observational health data to predict ADRs in both individual patients and global populations. In this review, we explore the rapid expansion of systems pharmacology in the study of ADRs. We enumerate the existing methods and strategies and illustrate progress in the field with a model framework that incorporates crucial data elements, such as diet and comorbidities, known to modulate ADR risk. Using this framework, we highlight avenues of research that may currently be underexplored, representing opportunities for future work.
Collapse
Affiliation(s)
- Mary Regina Boland
- Department of Biomedical Informatics, Columbia University, New York, NY, USA.,Department of Systems Biology, Columbia University, New York, NY, USA.,Department of Medicine, Columbia University, New York, NY, USA.,Observational Health Data Science and Informatics (OHDSI), New York, NY, USA
| | - Alexandra Jacunski
- Department of Biomedical Informatics, Columbia University, New York, NY, USA.,Department of Systems Biology, Columbia University, New York, NY, USA.,Department of Medicine, Columbia University, New York, NY, USA.,Integrated Program in Cellular, Molecular and Biomedical Studies, Columbia University, New York, NY, USA
| | - Tal Lorberbaum
- Department of Biomedical Informatics, Columbia University, New York, NY, USA.,Department of Systems Biology, Columbia University, New York, NY, USA.,Department of Medicine, Columbia University, New York, NY, USA.,Department of Physiology and Cellular Biophysics, Columbia University, New York, NY, USA
| | - Joseph D Romano
- Department of Biomedical Informatics, Columbia University, New York, NY, USA.,Department of Systems Biology, Columbia University, New York, NY, USA.,Department of Medicine, Columbia University, New York, NY, USA
| | - Robert Moskovitch
- Department of Biomedical Informatics, Columbia University, New York, NY, USA.,Department of Systems Biology, Columbia University, New York, NY, USA.,Department of Medicine, Columbia University, New York, NY, USA
| | - Nicholas P Tatonetti
- Department of Biomedical Informatics, Columbia University, New York, NY, USA.,Department of Systems Biology, Columbia University, New York, NY, USA.,Department of Medicine, Columbia University, New York, NY, USA.,Observational Health Data Science and Informatics (OHDSI), New York, NY, USA
| |
Collapse
|
72
|
Personalized Whole-Cell Kinetic Models of Metabolism for Discovery in Genomics and Pharmacodynamics. Cell Syst 2015; 1:283-92. [DOI: 10.1016/j.cels.2015.10.003] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Revised: 08/13/2015] [Accepted: 10/07/2015] [Indexed: 01/07/2023]
|
73
|
Systems Medicine: The Application of Systems Biology Approaches for Modern Medical Research and Drug Development. Mol Biol Int 2015; 2015:698169. [PMID: 26357572 PMCID: PMC4556074 DOI: 10.1155/2015/698169] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2015] [Revised: 07/27/2015] [Accepted: 07/29/2015] [Indexed: 12/21/2022] Open
Abstract
The exponential development of highly advanced scientific and medical research technologies throughout the past 30 years has arrived to the point where the high number of characterized molecular agents related to pathogenesis cannot be readily integrated or processed by conventional analytical approaches. Indeed, the realization that several moieties are signatures of disease has partly led to the increment of complex diseases being characterized. Scientists and clinicians can now investigate and analyse any individual dysregulations occurring within the genomic, transcriptomic, miRnomic, proteomic, and metabolomic levels thanks to currently available advanced technologies. However, there are drawbacks within this scientific brave new age in that only isolated molecular levels are individually investigated for their influence in affecting any particular health condition. Since their conception in 1992, systems biology/medicine focuses mainly on the perturbations of overall pathway kinetics for the consequent onset and/or deterioration of the investigated condition/s. Systems medicine approaches can therefore be employed for shedding light in multiple research scenarios, ultimately leading to the practical result of uncovering novel dynamic interaction networks that are critical for influencing the course of medical conditions. Consequently, systems medicine also serves to identify clinically important molecular targets for diagnostic and therapeutic measures against such a condition.
Collapse
|
74
|
Silva AJ, Müller KR. The need for novel informatics tools for integrating and planning research in molecular and cellular cognition. ACTA ACUST UNITED AC 2015; 22:494-8. [PMID: 26286658 PMCID: PMC4561409 DOI: 10.1101/lm.029355.112] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Accepted: 07/09/2015] [Indexed: 11/24/2022]
Abstract
The sheer volume and complexity of publications in the biological sciences are straining traditional approaches to research planning. Nowhere is this problem more serious than in molecular and cellular cognition, since in this neuroscience field, researchers routinely use approaches and information from a variety of areas in neuroscience and other biology fields. Additionally, the multilevel integration process characteristic of this field involves the establishment of experimental connections between molecular, electrophysiological, behavioral, and even cognitive data. This multidisciplinary integration process requires strategies and approaches that originate in several different fields, which greatly increases the complexity and demands of this process. Although causal assertions, where phenomenon A is thought to contribute or relate to B, are at the center of this integration process and key to research in biology, there are currently no tools to help scientists keep track of the increasingly more complex network of causal connections they use when making research decisions. Here, we propose the development of semiautomated graphical and interactive tools to help neuroscientists and other biologists, including those working in molecular and cellular cognition, to track, map, and weight causal evidence in research papers. There is a great need for a concerted effort by biologists, computer scientists, and funding institutions to develop maps of causal information that would aid in integration of research findings and in experiment planning.
Collapse
Affiliation(s)
- Alcino J Silva
- Department of Neurobiology, Department of Psychiatry, Department of Psychology, Integrative Center for Learning and Memory, Brain Research Institute, University of California at Los Angeles, Los Angeles, California 90095-1761, USA
| | - Klaus-Robert Müller
- Machine Learning Group, Technische Universität Berlin, 10587, Berlin, Germany Berlin Center for Big Data and Department of Brain and Cognitive Engineering, Korea University, Seoul, 136-701 Korea
| |
Collapse
|
75
|
van Hasselt JGC, van der Graaf PH. Towards integrative systems pharmacology models in oncology drug development. DRUG DISCOVERY TODAY. TECHNOLOGIES 2015; 15:1-8. [PMID: 26464083 DOI: 10.1016/j.ddtec.2015.06.004] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Revised: 05/31/2015] [Accepted: 06/12/2015] [Indexed: 02/02/2023]
Abstract
Quantitative systems pharmacology (QSP) modeling represents an emerging area of value to further streamline knowledge integration and to better inform decision making processes in drug development. QSP models reside at the interface between systems biology models and pharmacological models, yet their concrete implementation still needs to be established further. This review outlines key modeling techniques in both of these areas and to subsequently discuss challenges and opportunities for further integration, in oncology drug development.
Collapse
Affiliation(s)
- J G Coen van Hasselt
- Division of Pharmacology, Cluster Systems Pharmacology, Leiden Academic Centre for Drug Research, Leiden University, Einsteinweg 55, Leiden, The Netherlands.
| | - Piet H van der Graaf
- Division of Pharmacology, Cluster Systems Pharmacology, Leiden Academic Centre for Drug Research, Leiden University, Einsteinweg 55, Leiden, The Netherlands.
| |
Collapse
|
76
|
Lee JA, Shinn P, Jaken S, Oliver S, Willard FS, Heidler S, Peery RB, Oler J, Chu S, Southall N, Dexheimer TS, Smallwood J, Huang R, Guha R, Jadhav A, Cox K, Austin CP, Simeonov A, Sittampalam GS, Husain S, Franklin N, Wild DJ, Yang JJ, Sutherland JJ, Thomas CJ. Novel Phenotypic Outcomes Identified for a Public Collection of Approved Drugs from a Publicly Accessible Panel of Assays. PLoS One 2015; 10:e0130796. [PMID: 26177200 PMCID: PMC4503722 DOI: 10.1371/journal.pone.0130796] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2015] [Accepted: 05/26/2015] [Indexed: 12/17/2022] Open
Abstract
Phenotypic assays have a proven track record for generating leads that become first-in-class therapies. Whole cell assays that inform on a phenotype or mechanism also possess great potential in drug repositioning studies by illuminating new activities for the existing pharmacopeia. The National Center for Advancing Translational Sciences (NCATS) pharmaceutical collection (NPC) is the largest reported collection of approved small molecule therapeutics that is available for screening in a high-throughput setting. Via a wide-ranging collaborative effort, this library was analyzed in the Open Innovation Drug Discovery (OIDD) phenotypic assay modules publicly offered by Lilly. The results of these tests are publically available online at www.ncats.nih.gov/expertise/preclinical/pd2 and via the PubChem Database (https://pubchem.ncbi.nlm.nih.gov/) (AID 1117321). Phenotypic outcomes for numerous drugs were confirmed, including sulfonylureas as insulin secretagogues and the anti-angiogenesis actions of multikinase inhibitors sorafenib, axitinib and pazopanib. Several novel outcomes were also noted including the Wnt potentiating activities of rotenone and the antifolate class of drugs, and the anti-angiogenic activity of cetaben.
Collapse
Affiliation(s)
- Jonathan A. Lee
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana, United States of America
| | - Paul Shinn
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Susan Jaken
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana, United States of America
| | - Sarah Oliver
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana, United States of America
| | - Francis S. Willard
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana, United States of America
| | - Steven Heidler
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana, United States of America
| | - Robert B. Peery
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana, United States of America
| | - Jennifer Oler
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana, United States of America
| | - Shaoyou Chu
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana, United States of America
| | - Noel Southall
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Thomas S. Dexheimer
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Jeffrey Smallwood
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana, United States of America
| | - Ruili Huang
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Rajarshi Guha
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Ajit Jadhav
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Karen Cox
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana, United States of America
| | - Christopher P. Austin
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Anton Simeonov
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland, United States of America
| | - G. Sitta Sittampalam
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Saba Husain
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana, United States of America
| | - Natalie Franklin
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana, United States of America
| | - David J. Wild
- Indiana University School of Informatics and Computing, Bloomington, Indiana, United States of America
| | - Jeremy J. Yang
- Indiana University School of Informatics and Computing, Bloomington, Indiana, United States of America
| | - Jeffrey J. Sutherland
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana, United States of America
- * E-mail: (JJS); (CJT)
| | - Craig J. Thomas
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland, United States of America
- * E-mail: (JJS); (CJT)
| |
Collapse
|
77
|
Webb JT, Behar M. Topology, dynamics, and heterogeneity in immune signaling. WILEY INTERDISCIPLINARY REVIEWS-SYSTEMS BIOLOGY AND MEDICINE 2015; 7:285-300. [DOI: 10.1002/wsbm.1306] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/02/2015] [Revised: 04/14/2015] [Accepted: 04/21/2015] [Indexed: 12/28/2022]
Affiliation(s)
- J. Taylor Webb
- Department of Biomedical Engineering; The University of Texas at Austin; Austin TX USA
| | - Marcelo Behar
- Department of Biomedical Engineering; The University of Texas at Austin; Austin TX USA
| |
Collapse
|
78
|
Wang RS, Maron BA, Loscalzo J. Systems medicine: evolution of systems biology from bench to bedside. WILEY INTERDISCIPLINARY REVIEWS-SYSTEMS BIOLOGY AND MEDICINE 2015; 7:141-61. [PMID: 25891169 DOI: 10.1002/wsbm.1297] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Revised: 03/04/2015] [Accepted: 03/06/2015] [Indexed: 12/11/2022]
Abstract
High-throughput experimental techniques for generating genomes, transcriptomes, proteomes, metabolomes, and interactomes have provided unprecedented opportunities to interrogate biological systems and human diseases on a global level. Systems biology integrates the mass of heterogeneous high-throughput data and predictive computational modeling to understand biological functions as system-level properties. Most human diseases are biological states caused by multiple components of perturbed pathways and regulatory networks rather than individual failing components. Systems biology not only facilitates basic biological research but also provides new avenues through which to understand human diseases, identify diagnostic biomarkers, and develop disease treatments. At the same time, systems biology seeks to assist in drug discovery, drug optimization, drug combinations, and drug repositioning by investigating the molecular mechanisms of action of drugs at a system's level. Indeed, systems biology is evolving to systems medicine as a new discipline that aims to offer new approaches for addressing the diagnosis and treatment of major human diseases uniquely, effectively, and with personalized precision.
Collapse
Affiliation(s)
- Rui-Sheng Wang
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Bradley A Maron
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA.,Department of Cardiology, Veterans Affairs Boston Healthcare System, West Roxbury, MA, USA
| | - Joseph Loscalzo
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| |
Collapse
|
79
|
Chen Y, Palczewski K. Systems Pharmacology Links GPCRs with Retinal Degenerative Disorders. Annu Rev Pharmacol Toxicol 2015; 56:273-98. [PMID: 25839098 DOI: 10.1146/annurev-pharmtox-010715-103033] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
In most biological systems, second messengers and their key regulatory and effector proteins form links between multiple cellular signaling pathways. Such signaling nodes can integrate the deleterious effects of genetic aberrations, environmental stressors, or both in complex diseases, leading to cell death by various mechanisms. Here we present a systems (network) pharmacology approach that, together with transcriptomics analyses, was used to identify different G protein-coupled receptors that experimentally protected against cellular stress and death caused by linked signaling mechanisms. We describe the application of this concept to degenerative and diabetic retinopathies in appropriate mouse models as an example. Systems pharmacology also provides an attractive framework for devising strategies to combat complex diseases by using (repurposing) US Food and Drug Administration-approved pharmacological agents.
Collapse
Affiliation(s)
- Yu Chen
- Yueyang Hospital and.,Clinical Research Institute of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China
| | - Krzysztof Palczewski
- Department of Pharmacology, School of Medicine, Case Western Reserve University, Cleveland, Ohio 44106;
| |
Collapse
|
80
|
Sutherland JJ, Daly TM, Liu X, Goldstein K, Johnston JA, Ryan TP. Co-prescription trends in a large cohort of subjects predict substantial drug-drug interactions. PLoS One 2015; 10:e0118991. [PMID: 25739022 PMCID: PMC4349653 DOI: 10.1371/journal.pone.0118991] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2014] [Accepted: 01/08/2015] [Indexed: 11/18/2022] Open
Abstract
Pharmaceutical prescribing and drug-drug interaction data underlie recommendations on drug combinations that should be avoided or closely monitored by prescribers. Because the number of patients taking multiple medications is increasing, a comprehensive view of prescribing patterns in patients is important to better assess real world pharmaceutical response and evaluate the potential for multi-drug interactions. We obtained self-reported prescription data from NHANES surveys between 1999 and 2010, and confirm the previously reported finding of increasing drug use in the elderly. We studied co-prescription drug trends by focusing on the 2009-2010 survey, which contains prescription data on 690 drugs used by 10,537 subjects. We found that medication profiles were unique for individuals aged 65 years or more, with ≥98 unique drug regimens encountered per 100 subjects taking 3 or more medications. When drugs were viewed by therapeutic class, it was found that the most commonly prescribed drugs were not the most commonly co-prescribed drugs for any of the 16 drug classes investigated. We cross-referenced these medication lists with drug interaction data from Drugs.com to evaluate the potential for drug interactions. The number of drug alerts rose proportionally with the number of co-prescribed medications, rising from 3.3 alerts for individuals prescribed 5 medications to 11.7 alerts for individuals prescribed 10 medications. We found 22% of elderly subjects taking both a substrate and inhibitor of a given cytochrome P450 enzyme, and 4% taking multiple inhibitors of the same enzyme simultaneously. By examining drug pairs prescribed in 0.1% of the population or more, we found low agreement between co-prescription rate and co-discussion in the literature. These data show that prescribing trends in treatment could drive a large extent of individual variability in drug response, and that current pairwise approaches to assessing drug-drug interactions may be inadequate for predicting real world outcomes.
Collapse
Affiliation(s)
- Jeffrey J. Sutherland
- Lilly Research Labs IT, Eli Lilly and Company, Indianapolis, Indiana, United States of America
- * E-mail: (JJS); (TPR)
| | - Thomas M. Daly
- Pathology and Laboratory Medicine Institute, Cleveland Clinic, Cleveland, Ohio, United States of America
| | - Xiong Liu
- Lilly Research Labs IT, Eli Lilly and Company, Indianapolis, Indiana, United States of America
| | - Keith Goldstein
- Drug Disposition and Toxicology, Eli Lilly and Company, Indianapolis, Indiana, United States of America
| | - Joseph A. Johnston
- Global Patient Outcomes and Real World Evidence, Eli Lilly and Company, Indianapolis, Indiana, United States of America
| | - Timothy P. Ryan
- Sano Informed Prescribing, Brentwood, Tennessee, United States of America
- * E-mail: (JJS); (TPR)
| |
Collapse
|
81
|
Knowledge retrieval from PubMed abstracts and electronic medical records with the Multiple Sclerosis Ontology. PLoS One 2015; 10:e0116718. [PMID: 25665127 PMCID: PMC4321837 DOI: 10.1371/journal.pone.0116718] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2014] [Accepted: 12/13/2014] [Indexed: 12/03/2022] Open
Abstract
Background In order to retrieve useful information from scientific literature and electronic medical records (EMR) we developed an ontology specific for Multiple Sclerosis (MS). Methods The MS Ontology was created using scientific literature and expert review under the Protégé OWL environment. We developed a dictionary with semantic synonyms and translations to different languages for mining EMR. The MS Ontology was integrated with other ontologies and dictionaries (diseases/comorbidities, gene/protein, pathways, drug) into the text-mining tool SCAIView. We analyzed the EMRs from 624 patients with MS using the MS ontology dictionary in order to identify drug usage and comorbidities in MS. Testing competency questions and functional evaluation using F statistics further validated the usefulness of MS ontology. Results Validation of the lexicalized ontology by means of named entity recognition-based methods showed an adequate performance (F score = 0.73). The MS Ontology retrieved 80% of the genes associated with MS from scientific abstracts and identified additional pathways targeted by approved disease-modifying drugs (e.g. apoptosis pathways associated with mitoxantrone, rituximab and fingolimod). The analysis of the EMR from patients with MS identified current usage of disease modifying drugs and symptomatic therapy as well as comorbidities, which are in agreement with recent reports. Conclusion The MS Ontology provides a semantic framework that is able to automatically extract information from both scientific literature and EMR from patients with MS, revealing new pathogenesis insights as well as new clinical information.
Collapse
|
82
|
Li P, Huang C, Fu Y, Wang J, Wu Z, Ru J, Zheng C, Guo Z, Chen X, Zhou W, Zhang W, Li Y, Chen J, Lu A, Wang Y. Large-scale exploration and analysis of drug combinations. Bioinformatics 2015; 31:2007-16. [PMID: 25667546 DOI: 10.1093/bioinformatics/btv080] [Citation(s) in RCA: 104] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2014] [Accepted: 02/03/2015] [Indexed: 12/12/2022] Open
Abstract
MOTIVATION Drug combinations are a promising strategy for combating complex diseases by improving the efficacy and reducing corresponding side effects. Currently, a widely studied problem in pharmacology is to predict effective drug combinations, either through empirically screening in clinic or pure experimental trials. However, the large-scale prediction of drug combination by a systems method is rarely considered. RESULTS We report a systems pharmacology framework to predict drug combinations (PreDCs) on a computational model, termed probability ensemble approach (PEA), for analysis of both the efficacy and adverse effects of drug combinations. First, a Bayesian network integrating with a similarity algorithm is developed to model the combinations from drug molecular and pharmacological phenotypes, and the predictions are then assessed with both clinical efficacy and adverse effects. It is illustrated that PEA can predict the combination efficacy of drugs spanning different therapeutic classes with high specificity and sensitivity (AUC = 0.90), which was further validated by independent data or new experimental assays. PEA also evaluates the adverse effects (AUC = 0.95) quantitatively and detects the therapeutic indications for drug combinations. Finally, the PreDC database includes 1571 known and 3269 predicted optimal combinations as well as their potential side effects and therapeutic indications. AVAILABILITY AND IMPLEMENTATION The PreDC database is available at http://sm.nwsuaf.edu.cn/lsp/predc.php.
Collapse
Affiliation(s)
- Peng Li
- Lab of Systems Pharmacology, Center of Bioinformatics, College of Life Science, Northwest A&F University, Yangling, Shaanxi, China, School of Chemical engineering, Dalian University of Technology, Dalian, Liaoning, China, Beijing University of Chinese Medicine, ChaoYang District, Beijing, China and School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong
| | - Chao Huang
- Lab of Systems Pharmacology, Center of Bioinformatics, College of Life Science, Northwest A&F University, Yangling, Shaanxi, China, School of Chemical engineering, Dalian University of Technology, Dalian, Liaoning, China, Beijing University of Chinese Medicine, ChaoYang District, Beijing, China and School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong
| | - Yingxue Fu
- Lab of Systems Pharmacology, Center of Bioinformatics, College of Life Science, Northwest A&F University, Yangling, Shaanxi, China, School of Chemical engineering, Dalian University of Technology, Dalian, Liaoning, China, Beijing University of Chinese Medicine, ChaoYang District, Beijing, China and School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong
| | - Jinan Wang
- Lab of Systems Pharmacology, Center of Bioinformatics, College of Life Science, Northwest A&F University, Yangling, Shaanxi, China, School of Chemical engineering, Dalian University of Technology, Dalian, Liaoning, China, Beijing University of Chinese Medicine, ChaoYang District, Beijing, China and School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong
| | - Ziyin Wu
- Lab of Systems Pharmacology, Center of Bioinformatics, College of Life Science, Northwest A&F University, Yangling, Shaanxi, China, School of Chemical engineering, Dalian University of Technology, Dalian, Liaoning, China, Beijing University of Chinese Medicine, ChaoYang District, Beijing, China and School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong
| | - Jinlong Ru
- Lab of Systems Pharmacology, Center of Bioinformatics, College of Life Science, Northwest A&F University, Yangling, Shaanxi, China, School of Chemical engineering, Dalian University of Technology, Dalian, Liaoning, China, Beijing University of Chinese Medicine, ChaoYang District, Beijing, China and School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong
| | - Chunli Zheng
- Lab of Systems Pharmacology, Center of Bioinformatics, College of Life Science, Northwest A&F University, Yangling, Shaanxi, China, School of Chemical engineering, Dalian University of Technology, Dalian, Liaoning, China, Beijing University of Chinese Medicine, ChaoYang District, Beijing, China and School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong
| | - Zihu Guo
- Lab of Systems Pharmacology, Center of Bioinformatics, College of Life Science, Northwest A&F University, Yangling, Shaanxi, China, School of Chemical engineering, Dalian University of Technology, Dalian, Liaoning, China, Beijing University of Chinese Medicine, ChaoYang District, Beijing, China and School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong
| | - Xuetong Chen
- Lab of Systems Pharmacology, Center of Bioinformatics, College of Life Science, Northwest A&F University, Yangling, Shaanxi, China, School of Chemical engineering, Dalian University of Technology, Dalian, Liaoning, China, Beijing University of Chinese Medicine, ChaoYang District, Beijing, China and School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong
| | - Wei Zhou
- Lab of Systems Pharmacology, Center of Bioinformatics, College of Life Science, Northwest A&F University, Yangling, Shaanxi, China, School of Chemical engineering, Dalian University of Technology, Dalian, Liaoning, China, Beijing University of Chinese Medicine, ChaoYang District, Beijing, China and School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong
| | - Wenjuan Zhang
- Lab of Systems Pharmacology, Center of Bioinformatics, College of Life Science, Northwest A&F University, Yangling, Shaanxi, China, School of Chemical engineering, Dalian University of Technology, Dalian, Liaoning, China, Beijing University of Chinese Medicine, ChaoYang District, Beijing, China and School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong
| | - Yan Li
- Lab of Systems Pharmacology, Center of Bioinformatics, College of Life Science, Northwest A&F University, Yangling, Shaanxi, China, School of Chemical engineering, Dalian University of Technology, Dalian, Liaoning, China, Beijing University of Chinese Medicine, ChaoYang District, Beijing, China and School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong
| | - Jianxin Chen
- Lab of Systems Pharmacology, Center of Bioinformatics, College of Life Science, Northwest A&F University, Yangling, Shaanxi, China, School of Chemical engineering, Dalian University of Technology, Dalian, Liaoning, China, Beijing University of Chinese Medicine, ChaoYang District, Beijing, China and School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong
| | - Aiping Lu
- Lab of Systems Pharmacology, Center of Bioinformatics, College of Life Science, Northwest A&F University, Yangling, Shaanxi, China, School of Chemical engineering, Dalian University of Technology, Dalian, Liaoning, China, Beijing University of Chinese Medicine, ChaoYang District, Beijing, China and School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong
| | - Yonghua Wang
- Lab of Systems Pharmacology, Center of Bioinformatics, College of Life Science, Northwest A&F University, Yangling, Shaanxi, China, School of Chemical engineering, Dalian University of Technology, Dalian, Liaoning, China, Beijing University of Chinese Medicine, ChaoYang District, Beijing, China and School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong
| |
Collapse
|
83
|
Liu Y, Wei Q, Yu G, Gai W, Li Y, Chen X. DCDB 2.0: a major update of the drug combination database. DATABASE-THE JOURNAL OF BIOLOGICAL DATABASES AND CURATION 2014; 2014:bau124. [PMID: 25539768 PMCID: PMC4275564 DOI: 10.1093/database/bau124] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Experience in clinical practice and research in systems pharmacology suggested the limitations of the current one-drug-one-target paradigm in new drug discovery. Single-target drugs may not always produce desired physiological effects on the entire biological system, even if they have successfully regulated the activities of their designated targets. On the other hand, multicomponent therapy, in which two or more agents simultaneously interact with multiple targets, has attracted growing attention. Many drug combinations consisting of multiple agents have already entered clinical practice, especially in treating complex and refractory diseases. Drug combination database (DCDB), launched in 2010, is the first available database that collects and organizes information on drug combinations, with an aim to facilitate systems-oriented new drug discovery. Here, we report the second major release of DCDB (Version 2.0), which includes 866 new drug combinations (1363 in total), consisting of 904 distinctive components. These drug combinations are curated from ∼140,000 clinical studies and the food and drug administration (FDA) electronic orange book. In this update, DCDB collects 237 unsuccessful drug combinations, which may provide a contrast for systematic discovery of the patterns in successful drug combinations. Database URL: http://www.cls.zju.edu.cn/dcdb/
Collapse
Affiliation(s)
- Yanbin Liu
- Department of Bioinformatics, College of Life Sciences and Institute of Biochemistry, College of Life Sciences, Zhejiang University, Hangzhou 310058, P.R. China Department of Bioinformatics, College of Life Sciences and Institute of Biochemistry, College of Life Sciences, Zhejiang University, Hangzhou 310058, P.R. China
| | - Qiang Wei
- Department of Bioinformatics, College of Life Sciences and Institute of Biochemistry, College of Life Sciences, Zhejiang University, Hangzhou 310058, P.R. China
| | - Guisheng Yu
- Department of Bioinformatics, College of Life Sciences and Institute of Biochemistry, College of Life Sciences, Zhejiang University, Hangzhou 310058, P.R. China
| | - Wanxia Gai
- Department of Bioinformatics, College of Life Sciences and Institute of Biochemistry, College of Life Sciences, Zhejiang University, Hangzhou 310058, P.R. China
| | - Yongquan Li
- Department of Bioinformatics, College of Life Sciences and Institute of Biochemistry, College of Life Sciences, Zhejiang University, Hangzhou 310058, P.R. China
| | - Xin Chen
- Department of Bioinformatics, College of Life Sciences and Institute of Biochemistry, College of Life Sciences, Zhejiang University, Hangzhou 310058, P.R. China Department of Bioinformatics, College of Life Sciences and Institute of Biochemistry, College of Life Sciences, Zhejiang University, Hangzhou 310058, P.R. China
| |
Collapse
|
84
|
Mignani S, Bryszewska M, Klajnert-Maculewicz B, Zablocka M, Majoral JP. Advances in combination therapies based on nanoparticles for efficacious cancer treatment: an analytical report. Biomacromolecules 2014; 16:1-27. [PMID: 25426779 DOI: 10.1021/bm501285t] [Citation(s) in RCA: 97] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
The main objective of nanomedicine research is the development of nanoparticles as drug delivery systems or drugs per se to tackle diseases as cancer, which are a leading cause of death with developed nations. Targeted treatments against solid tumors generally lead to dramatic regressions, but, unfortunately, the responses are often short-lived due to resistant cancer cells. In addition, one of the major challenges of combination drug therapy (called "cocktail") is the crucial optimization of different drug parameters. This issue can be solved using combination nanotherapy. Nanoparticles developed in oncology based on combination nanotherapy are either (a) those designed to combat multidrug resistance or (b) those used to circumvent resistance to clinical cancer drugs. This review provides an overview of the different nanoparticles currently used in clinical treatments in oncology. We analyze in detail the development of combinatorial nanoparticles including dendrimers for dual drug delivery via two strategic approaches: (a) use of chemotherapeutics and chemosensitizers to combat multidrug resistance and (b) use of multiple cytotoxic drugs. Finally, in this review, we discuss the challenges, clinical outlook, and perspectives of the nanoparticle-based combination therapy in cancer.
Collapse
Affiliation(s)
- Serge Mignani
- Université Paris Descartes, PRES Sorbonne Paris Cité, CNRS UMR 860, Laboratoire de Chimie et de Biochimie pharmacologiques et toxicologique, 45, rue des Saints Pères, 75006 Paris, France
| | | | | | | | | |
Collapse
|
85
|
Systems biology: unlocking the complexities of disease to enhance medicine. Future Med Chem 2014; 6:1727-9. [PMID: 25407366 DOI: 10.4155/fmc.14.124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|
86
|
A community computational challenge to predict the activity of pairs of compounds. Nat Biotechnol 2014; 32:1213-22. [PMID: 25419740 DOI: 10.1038/nbt.3052] [Citation(s) in RCA: 206] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2014] [Accepted: 09/25/2014] [Indexed: 12/26/2022]
Abstract
Recent therapeutic successes have renewed interest in drug combinations, but experimental screening approaches are costly and often identify only small numbers of synergistic combinations. The DREAM consortium launched an open challenge to foster the development of in silico methods to computationally rank 91 compound pairs, from the most synergistic to the most antagonistic, based on gene-expression profiles of human B cells treated with individual compounds at multiple time points and concentrations. Using scoring metrics based on experimental dose-response curves, we assessed 32 methods (31 community-generated approaches and SynGen), four of which performed significantly better than random guessing. We highlight similarities between the methods. Although the accuracy of predictions was not optimal, we find that computational prediction of compound-pair activity is possible, and that community challenges can be useful to advance the field of in silico compound-synergy prediction.
Collapse
|
87
|
Anand P, Chandra N. Characterizing the pocketome of Mycobacterium tuberculosis and application in rationalizing polypharmacological target selection. Sci Rep 2014; 4:6356. [PMID: 25220818 PMCID: PMC5376175 DOI: 10.1038/srep06356] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2014] [Accepted: 06/20/2014] [Indexed: 01/13/2023] Open
Abstract
Polypharmacology is beginning to emerge as an important concept in the field of drug discovery. However, there are no established approaches to either select appropriate target sets or design polypharmacological drugs. Here, we propose a structural-proteomics approach that utilizes the structural information of the binding sites at a genome-scale obtained through in-house algorithms to characterize the pocketome, yielding a list of ligands that can participate in various biochemical events in the mycobacterial cell. The pocket-type space is seen to be much larger than the sequence or fold-space, suggesting that variations at the site-level contribute significantly to functional repertoire of the organism. All-pair comparisons of binding sites within Mycobacterium tuberculosis (Mtb), pocket-similarity network construction and clustering result in identification of binding-site sets, each containing a group of similar binding sites, theoretically having a potential to interact with a common set of compounds. A polypharmacology index is formulated to rank targets by incorporating a measure of druggability and similarity to other pockets within the proteome. This study presents a rational approach to identify targets with polypharmacological potential along with possible drugs for repurposing, while simultaneously, obtaining clues on lead compounds for use in new drug-discovery pipelines.
Collapse
Affiliation(s)
- Praveen Anand
- Department of Biochemistry, Indian Institute of Science, Bangalore-560012, India
| | - Nagasuma Chandra
- Department of Biochemistry, Indian Institute of Science, Bangalore-560012, India
| |
Collapse
|
88
|
Kotelnikova E, Bernardo-Faura M, Silberberg G, Kiani NA, Messinis D, Melas IN, Artigas L, Schwartz E, Mazo I, Masso M, Alexopoulos LG, Mas JM, Olsson T, Tegner J, Martin R, Zamora A, Paul F, Saez-Rodriguez J, Villoslada P. Signaling networks in MS: a systems-based approach to developing new pharmacological therapies. Mult Scler 2014; 21:138-46. [PMID: 25112814 DOI: 10.1177/1352458514543339] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The pathogenesis of multiple sclerosis (MS) involves alterations to multiple pathways and processes, which represent a significant challenge for developing more-effective therapies. Systems biology approaches that study pathway dysregulation should offer benefits by integrating molecular networks and dynamic models with current biological knowledge for understanding disease heterogeneity and response to therapy. In MS, abnormalities have been identified in several cytokine-signaling pathways, as well as those of other immune receptors. Among the downstream molecules implicated are Jak/Stat, NF-Kb, ERK1/3, p38 or Jun/Fos. Together, these data suggest that MS is likely to be associated with abnormalities in apoptosis/cell death, microglia activation, blood-brain barrier functioning, immune responses, cytokine production, and/or oxidative stress, although which pathways contribute to the cascade of damage and can be modulated remains an open question. While current MS drugs target some of these pathways, others remain untouched. Here, we propose a pragmatic systems analysis approach that involves the large-scale extraction of processes and pathways relevant to MS. These data serve as a scaffold on which computational modeling can be performed to identify disease subgroups based on the contribution of different processes. Such an analysis, targeting these relevant MS-signaling pathways, offers the opportunity to accelerate the development of novel individual or combination therapies.
Collapse
Affiliation(s)
- Ekaterina Kotelnikova
- Institute Biomedical Research August Pi Sunyer (IDIBAPS) - Hospital Clinic of Barcelona, Spain/Personal Biomedicine ZAO, and A. A. Kharkevich Institute for Information Transmission Problems, Russian Academy of Sciences, Russia
| | | | - Gilad Silberberg
- Unit of Computational Medicine, Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital, Sweden
| | - Narsis A Kiani
- Unit of Computational Medicine, Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital, Sweden
| | | | - Ioannis N Melas
- European Molecular Biology Laboratory, European Bioinformatics Institute, UK/ProtATonce Ltd, Greece/National Technical University of Athens, Greece
| | | | | | | | | | | | | | | | - Jesper Tegner
- Unit of Computational Medicine, Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital, Sweden
| | | | | | - Friedemann Paul
- NeuroCure Clinical Research Center and Department of Neurology, Charité University Medicine Berlin, Germany
| | | | - Pablo Villoslada
- Institute Biomedical Research August Pi Sunyer (IDIBAPS) - Hospital Clinic of Barcelona, Spain/Personal Biomedicine ZAO, and A. A. Kharkevich Institute for Information Transmission Problems, Russian Academy of Sciences, Russia
| |
Collapse
|
89
|
Ferrario A, Baltezarević D, Novakovic T, Parker M, Samardzic J. Evidence-based decision making in healthcare in Central Eastern Europe. Expert Rev Pharmacoecon Outcomes Res 2014; 14:611-5. [PMID: 25090222 DOI: 10.1586/14737167.2014.946014] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
- Alessandra Ferrario
- London School of Economics and Political Science, LSE Health, Houghton Street, London, UK
| | | | | | | | | |
Collapse
|
90
|
Lanir Y. Mechanistic micro-structural theory of soft tissues growth and remodeling: tissues with unidirectional fibers. Biomech Model Mechanobiol 2014; 14:245-66. [DOI: 10.1007/s10237-014-0600-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2013] [Accepted: 05/23/2014] [Indexed: 10/25/2022]
|
91
|
Wilson C. Therapy: One plus one equals zero--drug combinations mitigate adverse effects. Nat Rev Endocrinol 2014; 10:3. [PMID: 24165997 DOI: 10.1038/nrendo.2013.213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
92
|
|