Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
BMC Bioinformatics ; 19(1): 345, 2018 Oct 01.
Article in English | MEDLINE | ID: mdl-30285606

ABSTRACT

BACKGROUND: The Open Targets Platform integrates different data sources in order to facilitate identification of potential therapeutic drug targets to treat human diseases. It currently provides evidence for nearly 2.6 million potential target-disease pairs. G-protein coupled receptors are a drug target class of high interest because of the number of successful drugs being developed against them over many years. Here we describe a systematic approach utilizing the Open Targets Platform data to uncover and prioritize potential new disease indications for the G-protein coupled receptors and their ligands. RESULTS: Utilizing the data available in the Open Targets platform, potential G-protein coupled receptor and endogenous ligand disease association pairs were systematically identified. Intriguing examples such as GPR35 for inflammatory bowel disease and CXCR4 for viral infection are used as illustrations of how a systematic approach can aid in the prioritization of interesting drug discovery hypotheses. Combining evidences for G-protein coupled receptors and their corresponding endogenous peptidergic ligands increases confidence and provides supportive evidence for potential new target-disease hypotheses. Comparing such hypotheses to the global pharma drug discovery pipeline to validate the approach showed that more than 93% of G-protein coupled receptor-disease pairs with a high overall Open Targets score involved receptors with an existing drug discovery program. CONCLUSIONS: The Open Targets gene-disease score can be used to prioritize potential G-protein coupled receptors-indication hypotheses. In addition, availability of multiple different evidence types markedly increases confidence as does combining evidence from known receptor-ligand pairs. Comparing the top-ranked hypotheses to the current global pharma pipeline serves validation of our approach and identifies and prioritizes new therapeutic opportunities.


Subject(s)
Disease/genetics , Drug Discovery/methods , Ligands , Protein Binding/physiology , Receptors, G-Protein-Coupled/metabolism , Humans
2.
J Transl Med ; 15(1): 182, 2017 08 29.
Article in English | MEDLINE | ID: mdl-28851378

ABSTRACT

BACKGROUND: Target identification and validation is a pressing challenge in the pharmaceutical industry, with many of the programmes that fail for efficacy reasons showing poor association between the drug target and the disease. Computational prediction of successful targets could have a considerable impact on attrition rates in the drug discovery pipeline by significantly reducing the initial search space. Here, we explore whether gene-disease association data from the Open Targets platform is sufficient to predict therapeutic targets that are actively being pursued by pharmaceutical companies or are already on the market. METHODS: To test our hypothesis, we train four different classifiers (a random forest, a support vector machine, a neural network and a gradient boosting machine) on partially labelled data and evaluate their performance using nested cross-validation and testing on an independent set. We then select the best performing model and use it to make predictions on more than 15,000 genes. Finally, we validate our predictions by mining the scientific literature for proposed therapeutic targets. RESULTS: We observe that the data types with the best predictive power are animal models showing a disease-relevant phenotype, differential expression in diseased tissue and genetic association with the disease under investigation. On a test set, the neural network classifier achieves over 71% accuracy with an AUC of 0.76 when predicting therapeutic targets in a semi-supervised learning setting. We use this model to gain insights into current and failed programmes and to predict 1431 novel targets, of which a highly significant proportion has been independently proposed in the literature. CONCLUSIONS: Our in silico approach shows that data linking genes and diseases is sufficient to predict novel therapeutic targets effectively and confirms that this type of evidence is essential for formulating or strengthening hypotheses in the target discovery process. Ultimately, more rapid and automated target prioritisation holds the potential to reduce both the costs and the development times associated with bringing new medicines to patients.


Subject(s)
Computer Simulation , Genetic Predisposition to Disease , Molecular Targeted Therapy , Algorithms , Area Under Curve , Data Mining , Drug Discovery , Neural Networks, Computer , Reproducibility of Results
3.
Brief Bioinform ; 13(6): 751-68, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22445903

ABSTRACT

It is well known that microbes have an intricate role in human health and disease. However, targeted strategies for modulating human health through the modification of either human-associated microbial communities or associated human-host targets have yet to be realized. New knowledge about the role of microbial communities in the microbiota of the gastrointestinal tract (GIT) and their collective genomes, the GIT microbiome, in chronic diseases opens new opportunities for therapeutic interventions. GIT microbiota participation in drug metabolism is a further pharmaceutical consideration. In this review, we discuss how computational methods could lead to a systems-level understanding of the global physiology of the host-microbiota superorganism in health and disease. Such knowledge will provide a platform for the identification and development of new therapeutic strategies for chronic diseases possibly involving microbial as well as human-host targets that improve upon existing probiotics, prebiotics or antibiotics. In addition, integrative bioinformatics analysis will further our understanding of the microbial biotransformation of exogenous compounds or xenobiotics, which could lead to safer and more efficacious drugs.


Subject(s)
Data Mining , Gastrointestinal Tract/microbiology , Metagenome , Humans , Probiotics/therapeutic use , RNA, Ribosomal, 16S/genetics
4.
Toxicol Appl Pharmacol ; 270(2): 149-57, 2013 Jul 15.
Article in English | MEDLINE | ID: mdl-23602889

ABSTRACT

Improving drug attrition remains a challenge in pharmaceutical discovery and development. A major cause of early attrition is the demonstration of safety signals which can negate any therapeutic index previously established. Safety attrition needs to be put in context of clinical translation (i.e. human relevance) and is negatively impacted by differences between animal models and human. In order to minimize such an impact, an earlier assessment of pharmacological target homology across animal model species will enhance understanding of the context of animal safety signals and aid species selection during later regulatory toxicology studies. Here we sequenced the genomes of the Sus scrofa Göttingen minipig and the Canis familiaris beagle, two widely used animal species in regulatory safety studies. Comparative analyses of these new genomes with other key model organisms, namely mouse, rat, cynomolgus macaque, rhesus macaque, two related breeds (S. scrofa Duroc and C. familiaris boxer) and human reveal considerable variation in gene content. Key genes in toxicology and metabolism studies, such as the UGT2 family, CYP2D6, and SLCO1A2, displayed unique duplication patterns. Comparisons of 317 known human drug targets revealed surprising variation such as species-specific positive selection, duplication and higher occurrences of pseudogenized targets in beagle (41 genes) relative to minipig (19 genes). These data will facilitate the more effective use of animals in biomedical research.


Subject(s)
Dogs/genetics , Drug Discovery/methods , Genome , Models, Animal , Swine, Miniature/genetics , Animals , Base Sequence , Female , Molecular Sequence Data , Sequence Alignment , Sequence Analysis, DNA , Swine
5.
Nat Rev Drug Discov ; 18(1): 41-58, 2019 01.
Article in English | MEDLINE | ID: mdl-30310233

ABSTRACT

Given the high attrition rates, substantial costs and slow pace of new drug discovery and development, repurposing of 'old' drugs to treat both common and rare diseases is increasingly becoming an attractive proposition because it involves the use of de-risked compounds, with potentially lower overall development costs and shorter development timelines. Various data-driven and experimental approaches have been suggested for the identification of repurposable drug candidates; however, there are also major technological and regulatory challenges that need to be addressed. In this Review, we present approaches used for drug repurposing (also known as drug repositioning), discuss the challenges faced by the repurposing community and recommend innovative ways by which these challenges could be addressed to help realize the full potential of drug repurposing.


Subject(s)
Drug Discovery , Drug Industry , Drug Repositioning/standards , Humans
6.
Sci Rep ; 9(1): 9386, 2019 06 28.
Article in English | MEDLINE | ID: mdl-31253830

ABSTRACT

Support from human genetics increases the probability of success in drug development. However, few examples exist of successful genomically-driven drug repositioning. Given that a Mendelian form of severe enterocolitis is due to up-regulation of the interleukin-18 (IL18) signaling pathway, and pharmacologic inhibition of IL18 has been shown to reverse this enterocolitis, we undertook a Mendelian randomization study to test the causal effect of elevated IL18 levels on inflammatory bowel disease susceptibility (IBD) in 12,882 cases and 21,770 controls. Mendelian randomization is an established method to assess the role of biomarkers in disease etiology in a manner that minimizes confounding and prevents reverse causation. Using three SNPs that explained almost 7% of the variance in IL18 level, we found that each genetically predicted standard deviation increase in IL18 was associated with an increase in IBD susceptibility (odds ratio = 1.22, 95% CI = 1.11-1.34, P-value = 6 × 10-5). This association was further validated in 25,042 IBD cases and 34,915 controls (odds ratio = 1.13, 95% CI = 1.05-1.20). Recently, an anti-IL18 monoclonal antibody, which decreased free IL18 levels, was found to be safe, yet ineffective in a phase II trial for type 2 diabetes. Taken together, these genomic findings implicated IBD as an alternative indication for anti-IL18 therapy, which should be tested in randomized controlled trials.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Drug Repositioning , Inflammatory Bowel Diseases/drug therapy , Interleukin-18/therapeutic use , Alleles , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Anti-Inflammatory Agents, Non-Steroidal/adverse effects , Biomarkers , Genetic Predisposition to Disease , Humans , Inflammatory Bowel Diseases/diagnosis , Inflammatory Bowel Diseases/etiology , Interleukin-18/blood , Mendelian Randomization Analysis , Odds Ratio , Polymorphism, Single Nucleotide , Receptors, Interleukin-18/genetics , Receptors, Interleukin-18/metabolism , Severity of Illness Index , Treatment Outcome
7.
BMC Evol Biol ; 8: 273, 2008 Oct 06.
Article in English | MEDLINE | ID: mdl-18837980

ABSTRACT

BACKGROUND: Related species, such as humans and chimpanzees, often experience the same disease with varying degrees of pathology, as seen in the cases of Alzheimer's disease, or differing symptomatology as in AIDS. Furthermore, certain diseases such as schizophrenia, epithelial cancers and autoimmune disorders are far more frequent in humans than in other species for reasons not associated with lifestyle. Genes that have undergone positive selection during species evolution are indicative of functional adaptations that drive species differences. Thus we investigate whether biomedical disease differences between species can be attributed to positively selected genes. RESULTS: We identified genes that putatively underwent positive selection during the evolution of humans and four mammals which are often used to model human diseases (mouse, rat, chimpanzee and dog). We show that genes predicted to have been subject to positive selection pressure during human evolution are implicated in diseases such as epithelial cancers, schizophrenia, autoimmune diseases and Alzheimer's disease, all of which differ in prevalence and symptomatology between humans and their mammalian relatives. In agreement with previous studies, the chimpanzee lineage was found to have more genes under positive selection than any of the other lineages. In addition, we found new evidence to support the hypothesis that genes that have undergone positive selection tend to interact with each other. This is the first such evidence to be detected widely among mammalian genes and may be important in identifying molecular pathways causative of species differences. CONCLUSION: Our dataset of genes predicted to have been subject to positive selection in five species serves as an informative resource that can be consulted prior to selecting appropriate animal models during drug target validation. We conclude that studying the evolution of functional and biomedical disease differences between species is an important way to gain insight into their molecular causes and may provide a method to predict when animal models do not mirror human biology.


Subject(s)
Disease , Evolution, Molecular , Selection, Genetic , Algorithms , Animals , Base Sequence , Cluster Analysis , Computational Biology/methods , Dogs , Genetic Variation , Humans , Mice , Pan troglodytes/genetics , Rats , Sequence Alignment , Species Specificity
9.
Drug Discov Today ; 12(19-20): 826-32, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17933683

ABSTRACT

Drug discovery remains a difficult business with a very high level of attrition. Many steps in this long process use data generated from various species. One key challenge is to successfully translate the pre-clinical findings of target validation and safety studies in animal models to diverse human beings in the clinic. Advanced computational evolutionary analysis techniques combined with the increasing availability of sequence information enable the application of systematic evolutionary approaches to targets and pathways of interest to drug discovery. These analyses have the potential to increase our understanding of experimental differences observed between species.


Subject(s)
Computational Biology/methods , Drug Design , Evolution, Molecular , Genomics/methods , Research Design , Animals , Humans , Phylogeny , Species Specificity
10.
Expert Opin Drug Discov ; 12(7): 687-693, 2017 07.
Article in English | MEDLINE | ID: mdl-28494630

ABSTRACT

INTRODUCTION: Discovering, developing and validating new disease treatments is a challenging and time-consuming endeavor. Successful drug discovery hinges on selecting the best drug targets with relevance to human disease and evidence that modulating them will be beneficial for patients. Open data initiatives are increasingly placing such knowledge into the public domain. Areas covered: In this review, the authors discuss emerging resources such as Open Targets which integrate key information to prioritize target-disease connections. Researchers can use it, along with other resources, to select potential new therapeutic targets to initiate drug discovery projects. They also discuss public resources such as DrugBank and ChEMBL that offer potential tools to interrogate these targets. Expert opinion: In our opinion, publically available resources are democratizing and connecting information, enabling disease experts to access and prioritize targets of interest in ways that were not possible a few years ago. Moreover, there are several modalities in addition to small molecule perturbation to modulate a target's activity. Drug discovery scientists can now utilize these new resources to simultaneously evaluate a much larger number of targets than previously possible.


Subject(s)
Databases, Pharmaceutical , Drug Discovery/methods , Molecular Targeted Therapy , Animals , Computational Biology , Databases, Chemical , Drug Design , Humans
11.
Drug Discov Today ; 22(12): 1800-1807, 2017 12.
Article in English | MEDLINE | ID: mdl-28919242

ABSTRACT

The recently developed Open Targets platform consolidates a wide range of comprehensive evidence associating known and potential drug targets with human diseases. We have harnessed the integrated data from this platform for novel drug repositioning opportunities. Our computational workflow systematically mines data from various evidence categories and presents potential repositioning opportunities for drugs that are marketed or being investigated in ongoing human clinical trials, based on evidence strength on target-disease pairing. We classified these novel target-disease opportunities in several ways: (i) number of independent counts of evidence; (ii) broad therapy area of origin; and (iii) repositioning within or across therapy areas. Finally, we elaborate on one example that was identified by this approach.


Subject(s)
Computational Biology/methods , Drug Repositioning , Animals , Humans , Rare Diseases/drug therapy , Receptor, Melanocortin, Type 1/metabolism , Vitiligo/drug therapy , Vitiligo/metabolism
12.
Physiol Rep ; 4(10)2016 May.
Article in English | MEDLINE | ID: mdl-27207783

ABSTRACT

The gastrointestinal (GI) tract can have significant impact on the regulation of the whole-body metabolism and may contribute to the development of obesity and diabetes. To systemically elucidate the role of the GI tract in obesity, we performed a transcriptomic analysis in different parts of the GI tract of two obese mouse models: ob/ob and high-fat diet (HFD) fed mice. Compared to their lean controls, significant changes in the gene expression were observed in both obese mouse groups in the stomach (ob/ob: 959; HFD: 542). In addition, these changes were quantitatively much higher than in the intestine. Despite the difference in genetic background, the two mouse models shared 296 similar gene expression changes in the stomach. Among those genes, some had known associations to obesity, diabetes, and insulin resistance. In addition, the gene expression profiles strongly suggested an increased gastric acid secretion in both obese mouse models, probably through an activation of the gastrin pathway. In conclusion, our data reveal a previously unknown dominant connection between the stomach and obesity in murine models extensively used in research.


Subject(s)
Gastric Mucosa/metabolism , Gene Expression Profiling , Intestinal Mucosa/metabolism , Obesity/genetics , Obesity/metabolism , Adipose Tissue/metabolism , Animals , Diet, High-Fat/adverse effects , Gene Expression Profiling/methods , Gene Expression Regulation , Male , Mice , Mice, Inbred C57BL , Mice, Obese
13.
Sci Rep ; 6: 36205, 2016 11 08.
Article in English | MEDLINE | ID: mdl-27824084

ABSTRACT

It is commonly assumed that drug targets are expressed in tissues relevant to their indicated diseases, even under normal conditions. While multiple anecdotal cases support this hypothesis, a comprehensive study has not been performed to verify it. We conducted a systematic analysis to assess gene and protein expression for all targets of marketed and phase III drugs across a diverse collection of normal human tissues. For 87% of gene-disease pairs, the target is expressed in a disease-affected tissue under healthy conditions. This result validates the importance of confirming expression of a novel drug target in an appropriate tissue for each disease indication and strengthens previous findings showing that targets of efficacious drugs should be expressed in relevant tissues under normal conditions. Further characterization of the remaining 13% of gene-disease pairs revealed that most genes are expressed in a different tissue linked to another disease. Our analysis demonstrates the value of extensive tissue specific expression resources.both in terms of tissue and cell diversity as well as techniques used to measure gene expression.


Subject(s)
Gene Expression Profiling/methods , Genetic Predisposition to Disease/genetics , Proteomics/methods , Clinical Trials, Phase III as Topic , Gene Regulatory Networks , Humans , Molecular Targeted Therapy , Oligonucleotide Array Sequence Analysis , Organ Specificity
14.
Drug Discov Today ; 10(8): 595-601, 2005 Apr 15.
Article in English | MEDLINE | ID: mdl-15837603

ABSTRACT

Small non-coding RNAs called microRNAs have been shown to play important roles in gene regulation across a broad range of metazoans from plants to humans. In this review, the nature and function of microRNAs will be discussed, with special emphasis on the computational tools and databases available to predict microRNAs and the genes they target.


Subject(s)
Gene Targeting/methods , MicroRNAs/pharmacology , MicroRNAs/physiology , Animals , Gene Targeting/trends , Humans , Oligonucleotide Array Sequence Analysis/methods
15.
Nat Genet ; 47(8): 856-60, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26121088

ABSTRACT

Over a quarter of drugs that enter clinical development fail because they are ineffective. Growing insight into genes that influence human disease may affect how drug targets and indications are selected. However, there is little guidance about how much weight should be given to genetic evidence in making these key decisions. To answer this question, we investigated how well the current archive of genetic evidence predicts drug mechanisms. We found that, among well-studied indications, the proportion of drug mechanisms with direct genetic support increases significantly across the drug development pipeline, from 2.0% at the preclinical stage to 8.2% among mechanisms for approved drugs, and varies dramatically among disease areas. We estimate that selecting genetically supported targets could double the success rate in clinical development. Therefore, using the growing wealth of human genetic data to select the best targets and indications should have a measurable impact on the successful development of new drugs.


Subject(s)
Drug Approval/statistics & numerical data , Genetic Predisposition to Disease/genetics , Genome-Wide Association Study/statistics & numerical data , Polymorphism, Single Nucleotide , Chromosome Mapping , Databases, Genetic/statistics & numerical data , Genetic Association Studies/statistics & numerical data , Genetics, Medical/methods , Genetics, Medical/statistics & numerical data , Humans , Linkage Disequilibrium , Medical Subject Headings/statistics & numerical data , Molecular Targeted Therapy/statistics & numerical data
16.
Drug Discov Today ; 7(11): S64-9, 2002 Jun 01.
Article in English | MEDLINE | ID: mdl-12047882

ABSTRACT

The availability of the human genome sequence is a 'once in a lifetime' opportunity for scientists to uncover all possible human drug-targets. As the sequence is very large, the best way to identify new genes rapidly is by computational (in silico) methods. There are now many examples in which pharmaceutical companies have identified genes of interest initially by in silico analysis. High-throughput data-generation techniques, such as microarray analysis, are key to the generation of human genome data. Bioinformatics techniques are therefore certain to play an increasingly important role in drug discovery.


Subject(s)
Computational Biology/methods , Drug Delivery Systems/methods , Drug Discovery/methods , Microarray Analysis/methods , Genome, Human , Humans
17.
BMC Evol Biol ; 4: 39, 2004 Oct 12.
Article in English | MEDLINE | ID: mdl-15476560

ABSTRACT

BACKGROUND: As key regulators of mitotic chromosome segregation, the Aurora family of serine/threonine kinases play an important role in cell division. Abnormalities in Aurora kinases have been strongly linked with cancer, which has lead to the recent development of new classes of anti-cancer drugs that specifically target the ATP-binding domain of these kinases. From an evolutionary perspective, the species distribution of the Aurora kinase family is complex. Mammals uniquely have three Aurora kinases, Aurora-A, Aurora-B, and Aurora-C, while for other metazoans, including the frog, fruitfly and nematode, only Aurora-A and Aurora-B kinases are known. The fungi have a single Aurora-like homolog. Based on the tacit assumption of orthology to human counterparts, model organism studies have been central to the functional characterization of Aurora kinases. However, the ortholog and paralog relationships of these kinases across various species have not been rigorously examined. Here, we present comprehensive evolutionary analyses of the Aurora kinase family. RESULTS: Phylogenetic trees suggest that all three vertebrate Auroras evolved from a single urochordate ancestor. Specifically, Aurora-A is an orthologous lineage in cold-blooded vertebrates and mammals, while structurally similar Aurora-B and Aurora-C evolved more recently in mammals from a duplication of an ancestral Aurora-B/C gene found in cold-blooded vertebrates. All so-called Aurora-A and Aurora-B kinases of non-chordates are ancestral to the clade of chordate Auroras and, therefore, are not strictly orthologous to vertebrate counterparts. Comparisons of human Aurora-B and Aurora-C sequences to the resolved 3D structure of human Aurora-A lends further support to the evolutionary scenario that vertebrate Aurora-B and Aurora-C are closely related paralogs. Of the 26 residues lining the ATP-binding active site, only three were variant and all were specific to Aurora-A. CONCLUSIONS: In this study, we found that invertebrate Aurora-A and Aurora-B kinases are highly divergent protein families from their chordate counterparts. Furthermore, while the Aurora-A family is ubiquitous among all vertebrates, the Aurora-B and Aurora-C families in humans arose from a gene duplication event in mammals. These findings show the importance of understanding evolutionary relationships in the interpretation and transference of knowledge from studies of model organism systems to human cellular biology. In addition, given the important role of Aurora kinases in cancer, evolutionary analysis and comparisons of ATP-binding domains suggest a rationale for designing dual action anti-tumor drugs that inhibit both Aurora-B and Aurora-C kinases.


Subject(s)
Antineoplastic Agents/pharmacology , Evolution, Molecular , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/classification , Amino Acid Sequence , Animals , Aurora Kinase B , Aurora Kinase C , Aurora Kinases , Catalytic Domain , Chordata , Enzyme Inhibitors/pharmacology , Humans , Models, Animal , Molecular Sequence Data , Phylogeny , Protein Serine-Threonine Kinases/chemistry , Sequence Alignment
18.
Sci Rep ; 4: 7160, 2014 Nov 24.
Article in English | MEDLINE | ID: mdl-25418113

ABSTRACT

Drug co-prescription (or drug combination) is a therapeutic strategy widely used as it may improve efficacy and reduce side-effect (SE). Since it is impractical to screen all possible drug combinations for every indication, computational methods have been developed to predict new combinations. In this study, we describe a novel approach that utilizes clinical SEs from post-marketing surveillance and the drug label to predict 1,508 novel drug-drug combinations. It outperforms other prediction methods, achieving an AUC of 0.92 compared to an AUC of 0.69 in a previous method, on a much larger drug combination set (245 drug combinations in our dataset compared to 75 in previous work.). We further found from the feature selection that three FDA black-box warned serious SEs, namely pneumonia, haemorrhage rectum, and retinal bleeding, contributed mostly to the predictions and a model only using these three SEs can achieve an average area under curve (AUC) at 0.80 and accuracy at 0.91, potentially with its simplicity being recognized as a practical rule-of-three in drug co-prescription or making fixed-dose drug combination. We also demonstrate this performance is less likely to be influenced by confounding factors such as biased disease indications or chemical structures.


Subject(s)
Drug Combinations , Drug-Related Side Effects and Adverse Reactions , Models, Theoretical , Area Under Curve , Databases, Factual , Drug Interactions , Gastrointestinal Hemorrhage/etiology , Humans , Pneumonia/etiology , Product Surveillance, Postmarketing , ROC Curve , Retinal Hemorrhage/etiology
19.
Drug Discov Today ; 19(9): 1364-71, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24662034

ABSTRACT

Psoriasis is a chronic inflammatory skin disease with complex pathological features and unmet pharmacotherapy needs. Here, we present a framework for developing new therapeutic intervention strategies for psoriasis by utilizing publicly available clinical transcriptomics data sets. By exploring the underlying molecular mechanisms of psoriasis, the effects of subsequent perturbation of these mechanisms by drugs and an integrative analysis, we propose a psoriasis disease signature, identify potential drug repurposing opportunities and present novel target selection methodologies. We anticipate that the outlined methodology or similar approaches will further support biomarker discovery and the development of new drugs for psoriasis.


Subject(s)
Dermatologic Agents/pharmacology , Drug Design , Psoriasis/drug therapy , Biomarkers/metabolism , Drug Repositioning , Humans , Molecular Targeted Therapy , Psoriasis/genetics , Psoriasis/physiopathology , Transcriptome
SELECTION OF CITATIONS
SEARCH DETAIL