1
|
Nguyen CD, Yoo J, Jeong SJ, Ha HA, Yang JH, Lee G, Shin JC, Kim JH. Melittin - the main component of bee venom: a promising therapeutic agent for neuroprotection through keap1/Nrf2/HO-1 pathway activation. Chin Med 2024; 19:166. [PMID: 39605070 PMCID: PMC11603938 DOI: 10.1186/s13020-024-01020-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Accepted: 10/07/2024] [Indexed: 11/29/2024] Open
Abstract
The Nuclear factor erythroid 2-related factor (Nrf2)/ Heme oxygenase-1 (HO-1) pathway, known for its significant role in regulating innate antioxidant defense mechanisms, is increasingly being recognized for its potential in neuroprotection studies. Derived from bee venom, melittin's neuroprotective effects have raised interest. This study confirmed that melittin specificity upregulated the weakened Nrf2/HO-1 signaling in mice brain. Interestingly, we also revealed melittin's efficient tactic, as it was suggested to first restore redox balance and then gradually stabilized other regulations of the mouse hippocampus. Using a neuro-stress-induced scopolamine model, chromatography and mass spectrometry analysis revealed that melittin crossed the compromised blood-brain barrier and accumulated in the hippocampus, which provided the chance to interact directly to weakened neurons. A wide range of improvements of melittin action were observed from various tests from behavior Morris water maze, Y maze test to immune florescent staining, western blots. As we need to find out what is the focus of melittin action, we conducted a careful observation in mice which showed that: the first signs of changes, in the hippocampus, within 5 h after melittin administration were the restoration of the Nrf2/HO-1 system and suppression of oxidative stress. After this event, from 7 to 12.5 h after administration, a diversity of conditions was all ameliorated: inflammation, apoptosis, neurotrophic factors, cholinergic function, and tissue ATP level. This chain reaction underscores that melittin focus was on redox balance's role, which revived multiple neuronal functions. Evidence of enhancement in the mouse hippocampus led to further exploration with hippocampal cell line HT22 model. Immunofluorescence analysis showed melittin-induced Nrf2 translocation to the nucleus, which would initiating the translation of antioxidant genes like HO-1. Pathway inhibitors pinpointed melittin's direct influence on the Nrf2/HO-1 pathway. 3D docking models and pull-down assays suggested melittin's direct interaction with Keap1, the regulator of the Nrf2/HO-1 pathway. Overall, this study not only highlighted melittin specifically effect on Nrf2/HO-1, thus rebalancing cellular redox, and also showed that this is an effective multi-faceted therapeutic strategy against neurodegeneration.
Collapse
Affiliation(s)
- Cong Duc Nguyen
- College of Korean Medicine, Dongshin University, Naju, 58245, Republic of Korea
| | - Jaehee Yoo
- College of Korean Medicine, Dongshin University, Naju, 58245, Republic of Korea
| | - Sang Jun Jeong
- College of Korean Medicine, Dongshin University, Naju, 58245, Republic of Korea
| | - Hai-Anh Ha
- Faculty of Pharmacy, College of Medicine and Pharmacy, Duy Tan University, Da Nang, 550000, Vietnam
| | - Ji Hye Yang
- College of Korean Medicine, Dongshin University, Naju, 58245, Republic of Korea
| | - Gihyun Lee
- College of Korean Medicine, Dongshin University, Naju, 58245, Republic of Korea
| | - Jeong Cheol Shin
- College of Korean Medicine, Dongshin University, Naju, 58245, Republic of Korea.
- Department of Acupuncture and Moxibustion Medicine, Dongshin University Gwangju Korean Medicine Hospital, 141, Wolsan-ro, Nam-gu, Gwangju City 61619, Republic of Korea , 141 Wolsan-Ro Nam-Gu, Gwangju, 61619, Republic of Korea.
| | - Jae-Hong Kim
- College of Korean Medicine, Dongshin University, Naju, 58245, Republic of Korea.
- Department of Acupuncture and Moxibustion Medicine, Dongshin University Gwangju Korean Medicine Hospital, 141, Wolsan-ro, Nam-gu, Gwangju City 61619, Republic of Korea , 141 Wolsan-Ro Nam-Gu, Gwangju, 61619, Republic of Korea.
| |
Collapse
|
2
|
A New Computational Approach to Evaluating Systemic Gene–Gene Interactions in a Pathway Affected by Drug LY294002. Processes (Basel) 2020. [DOI: 10.3390/pr8101230] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
In this study, we investigate how drugs systemically affect genes via pathways by integrating information from interactions between chemical compounds and molecular expression datasets, and from pathway information such as gene sets using mathematical models. First, we adopt drug-induced gene expression datasets; then, employ gene set enrichment analysis tools for selecting candidate enrichment pathways; and lastly, implement the inverse algorithm package for identifying gene–gene regulatory networks in a pathway. We tested LY294002-induced datasets of the MCF7 breast cancer cell lines, and found a CELL CYCLE pathway with 101 genes, ERBB signaling pathway consisting of 82 genes, and MTOR pathway consisting of 45 genes. We consider two interactions: quantity strength depending on number of interactions, and quality strength depending on weight of interaction as positive (+) and negative (−) interactions. Our methods revealed ANAPC1-CDK6 (−0.412) and ORC2L- CHEK1(0.951) for the CELL CYCLE pathway; INS-RPS6 (−3.125) and PRKAA2-PRKAA2 (+1.319) for the MTOR pathway; and CBLB-RPS6KB1 (−0.141), RPS6KB1-CBLC (+0.238) for the ERBB signaling pathway to be top quality interactions. Top quantity interactions discovered include 12; the CDC (−,+) gene family for the CELL CYCLE pathway, 20; PIK3 (−), 23; PIK3CG (+) for the MTOR pathway, 11; PAK (−), 10; PIK3 (+) for the ERBB signaling pathway.
Collapse
|
3
|
Identification of pathways associated with chemosensitivity through network embedding. PLoS Comput Biol 2019; 15:e1006864. [PMID: 30893303 PMCID: PMC6443184 DOI: 10.1371/journal.pcbi.1006864] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Revised: 04/01/2019] [Accepted: 02/09/2019] [Indexed: 12/27/2022] Open
Abstract
Basal gene expression levels have been shown to be predictive of cellular response to cytotoxic treatments. However, such analyses do not fully reveal complex genotype- phenotype relationships, which are partly encoded in highly interconnected molecular networks. Biological pathways provide a complementary way of understanding drug response variation among individuals. In this study, we integrate chemosensitivity data from a large-scale pharmacogenomics study with basal gene expression data from the CCLE project and prior knowledge of molecular networks to identify specific pathways mediating chemical response. We first develop a computational method called PACER, which ranks pathways for enrichment in a given set of genes using a novel network embedding method. It examines a molecular network that encodes known gene-gene as well as gene-pathway relationships, and determines a vector representation of each gene and pathway in the same low-dimensional vector space. The relevance of a pathway to the given gene set is then captured by the similarity between the pathway vector and gene vectors. To apply this approach to chemosensitivity data, we identify genes whose basal expression levels in a panel of cell lines are correlated with cytotoxic response to a compound, and then rank pathways for relevance to these response-correlated genes using PACER. Extensive evaluation of this approach on benchmarks constructed from databases of compound target genes and large collections of drug response signatures demonstrates its advantages in identifying compound-pathway associations compared to existing statistical methods of pathway enrichment analysis. The associations identified by PACER can serve as testable hypotheses on chemosensitivity pathways and help further study the mechanisms of action of specific cytotoxic drugs. More broadly, PACER represents a novel technique of identifying enriched properties of any gene set of interest while also taking into account networks of known gene-gene relationships and interactions. Gene expression levels have been used to study the cellular response to drug treatments. However, analysis of gene expression without considering gene interactions cannot fully reveal complex genotype-phenotype relationships. Biological pathways reveal the interactions among genes, thus providing a complementary way of understanding the drug response variation among individuals. In this paper, we aim to identify pathways that mediate the chemical response of each drug. We used the recently generated CTRP pharmacogenomics data and CCLE basal expression data to identify these pathways. We showed that using the prior knowledge encoded in molecular networks substantially improves pathway identification. In particular, we integrate genes and pathways into a large heterogeneous network in which links are protein-protein interactions and gene-pathway affiliations. We then project this heterogeneous network onto a low-dimensional space, which enables more precise similarity measurements between pathways and drug-response-correlated genes. Extensive experiments on two benchmarks show that our method substantially improved the pathway identification performance by using the molecular networks. More importantly, our method represents a novel technique of identifying enriched properties of any gene set of interest while also taking into account networks of known gene-gene relationships and interactions.
Collapse
|
4
|
Covell DG. A data mining approach for identifying pathway-gene biomarkers for predicting clinical outcome: A case study of erlotinib and sorafenib. PLoS One 2017; 12:e0181991. [PMID: 28792525 PMCID: PMC5549706 DOI: 10.1371/journal.pone.0181991] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Accepted: 07/10/2017] [Indexed: 12/28/2022] Open
Abstract
A novel data mining procedure is proposed for identifying potential pathway-gene biomarkers from preclinical drug sensitivity data for predicting clinical responses to erlotinib or sorafenib. The analysis applies linear ridge regression modeling to generate a small (N~1000) set of baseline gene expressions that jointly yield quality predictions of preclinical drug sensitivity data and clinical responses. Standard clustering of the pathway-gene combinations from gene set enrichment analysis of this initial gene set, according to their shared appearance in molecular function pathways, yields a reduced (N~300) set of potential pathway-gene biomarkers. A modified method for quantifying pathway fitness is used to determine smaller numbers of over and under expressed genes that correspond with favorable and unfavorable clinical responses. Detailed literature-based evidence is provided in support of the roles of these under and over expressed genes in compound efficacy. RandomForest analysis of potential pathway-gene biomarkers finds average treatment prediction errors of 10% and 22%, respectively, for patients receiving erlotinib or sorafenib that had a favorable clinical response. Higher errors were found for both compounds when predicting an unfavorable clinical response. Collectively these results suggest complementary roles for biomarker genes and biomarker pathways when predicting clinical responses from preclinical data.
Collapse
Affiliation(s)
- David G. Covell
- Information Technology Branch, Developmental Therapeutics Program, National Cancer Institute, Frederick, MD, United States of America
| |
Collapse
|
5
|
Li R, Kim D, Ritchie MD. Methods to analyze big data in pharmacogenomics research. Pharmacogenomics 2017; 18:807-820. [PMID: 28612644 DOI: 10.2217/pgs-2016-0152] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
The scale and scope of pharmacogenomics research continues to expand as the cost and efficiency of molecular data generation techniques advance. These new technologies give rise to enormous opportunity for the identification of important genetic and genomic factors important for drug treatment response. With this opportunity come significant challenges. Most of these can be categorized as 'big data' issues, facing not only pharmacogenomics, but other fields in the life sciences as well. In this review, we describe some of the analysis techniques and tools being implemented for genetic/genomic discovery in pharmacogenomics.
Collapse
Affiliation(s)
- Ruowang Li
- Bioinformatics & Genomics Graduate Program, The Pennsylvania State University, University Park, PA 16802, USA
| | - Dokyoon Kim
- Biomedical & Translational Informatics Institute, Geisinger Health System, Danville, PA 17821, USA
| | - Marylyn D Ritchie
- Bioinformatics & Genomics Graduate Program, The Pennsylvania State University, University Park, PA 16802, USA.,Biomedical & Translational Informatics Institute, Geisinger Health System, Danville, PA 17821, USA
| |
Collapse
|
6
|
Hanson C, Cairns J, Wang L, Sinha S. Computational discovery of transcription factors associated with drug response. THE PHARMACOGENOMICS JOURNAL 2016; 16:573-582. [PMID: 26503816 PMCID: PMC4848185 DOI: 10.1038/tpj.2015.74] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/23/2015] [Revised: 08/04/2015] [Accepted: 08/07/2015] [Indexed: 02/01/2023]
Abstract
This study integrates gene expression, genotype and drug response data in lymphoblastoid cell lines with transcription factor (TF)-binding sites from ENCODE (Encyclopedia of Genomic Elements) in a novel methodology that elucidates regulatory contexts associated with cytotoxicity. The method, GENMi (Gene Expression iN the Middle), postulates that single-nucleotide polymorphisms within TF-binding sites putatively modulate its regulatory activity, and the resulting variation in gene expression leads to variation in drug response. Analysis of 161 TFs and 24 treatments revealed 334 significantly associated TF-treatment pairs. Investigation of 20 selected pairs yielded literature support for 13 of these associations, often from studies where perturbation of the TF expression changes drug response. Experimental validation of significant GENMi associations in taxanes and anthracyclines across two triple-negative breast cancer cell lines corroborates our findings. The method is shown to be more sensitive than an alternative, genome-wide association study-based approach that does not use gene expression. These results demonstrate the utility of GENMi in identifying TFs that influence drug response and provide a number of candidates for further testing.
Collapse
Affiliation(s)
- C Hanson
- Department of Computer Science, University of Illinois at Urbana–Champaign, Urbana, IL, USA
| | - J Cairns
- Division of Clinical Pharmacology, Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
| | - L Wang
- Division of Clinical Pharmacology, Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
| | - S Sinha
- Department of Computer Science and Institute of Genomic Biology, University of Illinois at Urbana–Champaign, Urbana, IL, USA
| |
Collapse
|
7
|
Brnjic S, Mazurkiewicz M, Fryknäs M, Sun C, Zhang X, Larsson R, D'Arcy P, Linder S. Induction of tumor cell apoptosis by a proteasome deubiquitinase inhibitor is associated with oxidative stress. Antioxid Redox Signal 2014; 21:2271-85. [PMID: 24011031 PMCID: PMC4241954 DOI: 10.1089/ars.2013.5322] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
AIMS b-AP15 is a recently described inhibitor of the USP14/UCHL5 deubiquitinases (DUBs) of the 19S proteasome. Exposure to b-AP15 results in blocking of proteasome function and accumulation of polyubiquitinated protein substrates in cells. This novel mechanism of proteasome inhibition may potentially be exploited for cancer therapy, in particular for treatment of malignancies resistant to currently used proteasome inhibitors. The aim of the present study was to characterize the cellular response to b-AP15-mediated proteasome DUB inhibition. RESULTS We report that b-AP15 elicits a similar, but yet distinct, cellular response as the clinically used proteasome inhibitor bortezomib. b-AP15 induces a rapid apoptotic response, associated with enhanced induction of oxidative stress and rapid activation of Jun-N-terminal kinase 1/2 (JNK)/activating protein-1 signaling. Scavenging of reactive oxygen species and pharmacological inhibition of JNK reduced b-AP15-induced apoptosis. We further report that endoplasmic reticulum (ER) stress is induced by b-AP15 and is involved in apoptosis induction. In contrast to bortezomib, ER stress is associated with induction of α-subunit of eukaryotic initiation factor 2 phosphorylation. INNOVATION The findings establish that different modes of proteasome inhibition result in distinct cellular responses, a finding of potential therapeutic importance. CONCLUSION Our data show that enhanced oxidative stress and ER stress are major determinants of the strong apoptotic response elicited by the 19S DUB inhibitor b-AP15.
Collapse
Affiliation(s)
- Slavica Brnjic
- 1 Department of Oncology and Pathology, Cancer Center Karolinska, Karolinska Institute , Stockholm, Sweden
| | | | | | | | | | | | | | | |
Collapse
|
8
|
Systematic analysis of time-series gene expression data on tumor cell-selective apoptotic responses to HDAC inhibitors. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2014; 2014:867289. [PMID: 25371703 PMCID: PMC4211306 DOI: 10.1155/2014/867289] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/13/2014] [Accepted: 08/07/2014] [Indexed: 01/20/2023]
Abstract
SAHA (suberoylanilide hydroxamic acid or vorinostat) is the first nonselective histone deacetylase (HDAC) inhibitor approved by the US Food and Drug Administration (FDA). SAHA affects histone acetylation in chromatin and a variety of nonhistone substrates, thus influencing many cellular processes. In particularly, SAHA induces selective apoptosis of tumor cells, although the mechanism is not well understood. A series of microarray experiments was recently conducted to investigate tumor cell-selective proapoptotic transcriptional responses induced by SAHA. Based on that gene expression time series, we propose a novel framework for detailed analysis of the mechanism of tumor cell apoptosis selectively induced by SAHA. Our analyses indicated that SAHA selectively disrupted the DNA damage response, cell cycle, p53 expression, and mitochondrial integrity of tumor samples to induce selective tumor cell apoptosis. Our results suggest a possible regulation network. Our research extends the existing research.
Collapse
|
9
|
Integrating systems biology sources illuminates drug action. Clin Pharmacol Ther 2014; 95:663-9. [PMID: 24577151 PMCID: PMC4029855 DOI: 10.1038/clpt.2014.51] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2013] [Accepted: 02/18/2014] [Indexed: 12/26/2022]
Abstract
There are significant gaps in our understanding of the pathways by which drugs act. This incomplete knowledge limits our ability to use mechanistic molecular information rationally to repurpose drugs, understand their side effects, and predict their interactions with other drugs. Here we present DrugRouter: a novel method for generating drug-specific pathways of action by linking target genes, disease genes and pharmacogenes using gene interaction networks. We construct pathways for over a hundred drugs, and show that the genes included in our pathways (1) co-occur with the query drug in the literature, (2) significantly overlap or are adjacent to known drug-response pathways, and (3) are adjacent to genes that are hits in genome wide association studies assessing drug response. Finally, these computed pathways suggest novel drug repositioning opportunities (e.g., statins for follicular thyroid cancer), gene-side effect associations, and gene-drug interactions. Thus, DrugRouter generates hypotheses about drug actions using systems biology data.
Collapse
|
10
|
Pan Y, Cheng T, Wang Y, Bryant SH. Pathway analysis for drug repositioning based on public database mining. J Chem Inf Model 2014; 54:407-18. [PMID: 24460210 PMCID: PMC3956470 DOI: 10.1021/ci4005354] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
![]()
Sixteen FDA-approved
drugs were investigated to elucidate their
mechanisms of action (MOAs) and clinical functions by pathway analysis
based on retrieved drug targets interacting with or affected by the
investigated drugs. Protein and gene targets and associated pathways
were obtained by data-mining of public databases including the MMDB,
PubChem BioAssay, GEO DataSets, and the BioSystems databases. Entrez
E-Utilities were applied, and in-house Ruby scripts were developed
for data retrieval and pathway analysis to identify and evaluate relevant
pathways common to the retrieved drug targets. Pathways pertinent
to clinical uses or MOAs were obtained for most drugs. Interestingly,
some drugs identified pathways responsible for other diseases than
their current therapeutic uses, and these pathways were verified retrospectively
by in vitro tests, in vivo tests, or clinical trials. The pathway
enrichment analysis based on drug target information from public databases
could provide a novel approach for elucidating drug MOAs and repositioning,
therefore benefiting the discovery of new therapeutic treatments for
diseases.
Collapse
Affiliation(s)
- Yongmei Pan
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health , 8600 Rockville Pike, Bethesda, Maryland 20894, United States
| | | | | | | |
Collapse
|
11
|
Matheny CJ, Wei MC, Bassik MC, Donnelly AJ, Kampmann M, Iwasaki M, Piloto O, Solow-Cordero DE, Bouley DM, Rau R, Brown P, McManus MT, Weissman JS, Cleary ML. Next-generation NAMPT inhibitors identified by sequential high-throughput phenotypic chemical and functional genomic screens. ACTA ACUST UNITED AC 2013; 20:1352-63. [PMID: 24183972 DOI: 10.1016/j.chembiol.2013.09.014] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2013] [Revised: 09/16/2013] [Accepted: 09/24/2013] [Indexed: 01/09/2023]
Abstract
Phenotypic high-throughput chemical screens allow for discovery of small molecules that modulate complex phenotypes and provide lead compounds for novel therapies; however, identification of the mechanistically relevant targets remains a major experimental challenge. We report the application of sequential unbiased high-throughput chemical and ultracomplex small hairpin RNA (shRNA) screens to identify a distinctive class of inhibitors that target nicotinamide phosphoribosyl transferase (NAMPT), a rate-limiting enzyme in the biosynthesis of nicotinamide adenine dinucleotide, a crucial cofactor in many biochemical processes. The lead compound STF-118804 is a highly specific NAMPT inhibitor, improves survival in an orthotopic xenotransplant model of high-risk acute lymphoblastic leukemia, and targets leukemia stem cells. Tandem high-throughput screening using chemical and ultracomplex shRNA libraries, therefore, provides a rapid chemical genetics approach for seamless progression from small-molecule lead identification to target discovery and validation.
Collapse
Affiliation(s)
- Christina J Matheny
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Pediatrics, Stanford University School of Medicine, Stanford, CA 94305, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Fernald GH, Altman RB. Using molecular features of xenobiotics to predict hepatic gene expression response. J Chem Inf Model 2013; 53:2765-73. [PMID: 24010729 PMCID: PMC3810861 DOI: 10.1021/ci3005868] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Despite recent advances in molecular medicine and rational drug design, many drugs still fail because toxic effects arise at the cellular and tissue level. In order to better understand these effects, cellular assays can generate high-throughput measurements of gene expression changes induced by small molecules. However, our understanding of how the chemical features of small molecules influence gene expression is very limited. Therefore, we investigated the extent to which chemical features of small molecules can reliably be associated with significant changes in gene expression. Specifically, we analyzed the gene expression response of rat liver cells to 170 different drugs and searched for genes whose expression could be related to chemical features alone. Surprisingly, we can predict the up-regulation of 87 genes (increased expression of at least 1.5 times compared to controls). We show an average cross-validation predictive area under the receiver operating characteristic curve (AUROC) of 0.7 or greater for each of these 87 genes. We applied our method to an external data set of rat liver gene expression response to a novel drug and achieved an AUROC of 0.7. We also validated our approach by predicting up-regulation of Cytochrome P450 1A2 (CYP1A2) in three drugs known to induce CYP1A2 that were not in our data set. Finally, a detailed analysis of the CYP1A2 predictor allowed us to identify which fragments made significant contributions to the predictive scores.
Collapse
Affiliation(s)
- Guy Haskin Fernald
- Biomedical Informatics Training Program, Stanford University School of Medicine and ‡Departments of Bioengineering and Genetics, Stanford University , Stanford, California 94305, United States
| | | |
Collapse
|
13
|
Mezencev R, Wang L, McDonald JF. Identification of inhibitors of ovarian cancer stem-like cells by high-throughput screening. J Ovarian Res 2012; 5:30. [PMID: 23078816 PMCID: PMC3484114 DOI: 10.1186/1757-2215-5-30] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2012] [Accepted: 10/13/2012] [Indexed: 12/14/2022] Open
Abstract
Background Ovarian cancer stem cells are characterized by self-renewal capacity, ability to differentiate into distinct lineages, as well as higher invasiveness and resistance to many anticancer agents. Since they may be responsible for the recurrence of ovarian cancer after initial response to chemotherapy, development of new therapies targeting this special cellular subpopulation embedded within bulk ovarian cancers is warranted. Methods A high-throughput screening (HTS) campaign was performed with 825 compounds from the Mechanistic Set chemical library [Developmental Therapeutics Program (DTP)/National Cancer Institute (NCI)] against ovarian cancer stem-like cells (CSC) using a resazurin-based cell cytotoxicity assay. Identified sets of active compounds were projected onto self-organizing maps to identify their putative cellular response groups. Results From 793 screening compounds with evaluable data, 158 were found to have significant inhibitory effects on ovarian CSC. Computational analysis indicates that the majority of these compounds are associated with mitotic cellular responses. Conclusions Our HTS has uncovered a number of candidate compounds that may, after further testing, prove effective in targeting both ovarian CSC and their more differentiated progeny.
Collapse
Affiliation(s)
- Roman Mezencev
- School of Biology and Integrated Cancer Research Center, Georgia Institute of Technology, 310 Ferst Dr, Atlanta, GA, 30332, USA.
| | | | | |
Collapse
|
14
|
Covell DG. Integrating constitutive gene expression and chemoactivity: mining the NCI60 anticancer screen. PLoS One 2012; 7:e44631. [PMID: 23056181 PMCID: PMC3462800 DOI: 10.1371/journal.pone.0044631] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2012] [Accepted: 08/06/2012] [Indexed: 01/10/2023] Open
Abstract
Studies into the genetic origins of tumor cell chemoactivity pose significant challenges to bioinformatic mining efforts. Connections between measures of gene expression and chemoactivity have the potential to identify clinical biomarkers of compound response, cellular pathways important to efficacy and potential toxicities; all vital to anticancer drug development. An investigation has been conducted that jointly explores tumor-cell constitutive NCI60 gene expression profiles and small-molecule NCI60 growth inhibition chemoactivity profiles, viewed from novel applications of self-organizing maps (SOMs) and pathway-centric analyses of gene expressions, to identify subsets of over- and under-expressed pathway genes that discriminate chemo-sensitive and chemo-insensitive tumor cell types. Linear Discriminant Analysis (LDA) is used to quantify the accuracy of discriminating genes to predict tumor cell chemoactivity. LDA results find 15% higher prediction accuracies, using ∼30% fewer genes, for pathway-derived discriminating genes when compared to genes derived using conventional gene expression-chemoactivity correlations. The proposed pathway-centric data mining procedure was used to derive discriminating genes for ten well-known compounds. Discriminating genes were further evaluated using gene set enrichment analysis (GSEA) to reveal a cellular genetic landscape, comprised of small numbers of key over and under expressed on- and off-target pathway genes, as important for a compound’s tumor cell chemoactivity. Literature-based validations are provided as support for chemo-important pathways derived from this procedure. Qualitatively similar results are found when using gene expression measurements derived from different microarray platforms. The data used in this analysis is available at http://pubchem.ncbi.nlm.nih.gov/andhttp://www.ncbi.nlm.nih.gov/projects/geo (GPL96, GSE32474).
Collapse
Affiliation(s)
- David G Covell
- Developmental Therapeutics Program, Frederick National Laboratory, National Institutes of Health, Frederick, Maryland, United States of America.
| |
Collapse
|
15
|
Silberberg Y, Gottlieb A, Kupiec M, Ruppin E, Sharan R. Large-scale elucidation of drug response pathways in humans. J Comput Biol 2012; 19:163-74. [PMID: 22300318 DOI: 10.1089/cmb.2011.0264] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Elucidating signaling pathways is a fundamental step in understanding cellular processes and developing new therapeutic strategies. Here we introduce a method for the large-scale elucidation of signaling pathways involved in cellular response to drugs. Combining drug targets, drug response expression profiles, and the human physical interaction network, we infer 99 human drug response pathways and study their properties. Based on the newly inferred pathways, we develop a pathway-based drug-drug similarity measure and compare it to two common, gold standard drug-drug similarity measures. Remarkably, our measure provides better correspondence to these gold standards than similarity measures that are based on associations between drugs and known pathways, or on drug-specific gene expression profiles. It further improves the prediction of drug side effects and indications, elucidating specific response pathways that may be associated with these drug properties. Supplementary Material for this article is available at www.liebertonline.com/cmb.
Collapse
Affiliation(s)
- Yael Silberberg
- Department of Molecular Microbiology and Biotechnology, Tel Aviv University, Tel Aviv, Israel
| | | | | | | | | |
Collapse
|
16
|
Wan P, Li Q, Larsen JEP, Eklund AC, Parlesak A, Rigina O, Nielsen SJ, Björkling F, Jónsdóttir SÓ. Prediction of drug efficacy for cancer treatment based on comparative analysis of chemosensitivity and gene expression data. Bioorg Med Chem 2011; 20:167-76. [PMID: 22154557 DOI: 10.1016/j.bmc.2011.11.019] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2011] [Revised: 11/06/2011] [Accepted: 11/11/2011] [Indexed: 01/24/2023]
Abstract
The NCI60 database is the largest available collection of compounds with measured anti-cancer activity. The strengths and limitations for using the NCI60 database as a source of new anti-cancer agents are explored and discussed in relation to previous studies. We selected a sub-set of 2333 compounds with reliable experimental half maximum growth inhibitions (GI(50)) values for 30 cell lines from the NCI60 data set and evaluated their growth inhibitory effect (chemosensitivity) with respect to tissue of origin. This was done by identifying natural clusters in the chemosensitivity data set and in a data set of expression profiles of 1901 genes for the corresponding tumor cell lines. Five clusters were identified based on the gene expression data using self-organizing maps (SOM), comprising leukemia, melanoma, ovarian and prostate, basal breast, and luminal breast cancer cells, respectively. The strong difference in gene expression between basal and luminal breast cancer cells was reflected clearly in the chemosensitivity data. Although most compounds in the data set were of low potency, high efficacy compounds that showed specificity with respect to tissue of origin could be found. Furthermore, eight potential topoisomerase II inhibitors were identified using a structural similarity search. Finally, a set of genes with expression profiles that were significantly correlated with anti-cancer drug activity was identified. Our study demonstrates that the combined data sets, which provide comprehensive information on drug activity and gene expression profiles of tumor cell lines studied, are useful for identifying potential new active compounds.
Collapse
Affiliation(s)
- Peng Wan
- Center for Biological Sequence Analysis, Department of Systems Biology, Technical University of Denmark, Bldg. 208, DK-2800 Kgs. Lyngby, Denmark.
| | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Abstract
The field of pharmacogenomics is focused on the characterization of genetic factors contributing to the response of patients to pharmacological interventions. Drug response and toxicity are complex traits; therefore the effects are likely influenced by multiple genes. The investigation of the genetic basis of drug response has evolved from a focus on single genes to relevant pathways to the entire genome. Preclinical (cell-based models) and clinical genome-wide association studies (GWAS) in oncology provide an unprecedented opportunity for a comprehensive and unbiased assessment of the heritable factors associated with drug response. The primary challenge with attempting to identify pharmacogenomic markers from clinical studies is that they require a homogeneous population of patients treated with the same dosage regimen and minimal confounding variables. Therefore, the development of cell-based models for pharmacogenomic marker identification has utility for the field since performing these types of studies in humans is difficult and costly. This review intends to provide a current report on the status of genomic studies in oncology, the methods for discovery, and implications for patient care. We present a perspective and summary of the challenges and opportunities in translating heritable genomic discoveries to patients.
Collapse
Affiliation(s)
- Federico Innocenti
- Department of Medicine, Comprehensive Cancer Center, The University of Chicago, Chicago, IL 60637, USA
| | | | | |
Collapse
|
18
|
Roschke AV, Kirsch IR. Targeting karyotypic complexity and chromosomal instability of cancer cells. Curr Drug Targets 2011; 11:1341-50. [PMID: 20840077 DOI: 10.2174/1389450111007011341] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2010] [Accepted: 03/12/2010] [Indexed: 11/22/2022]
Abstract
Multiple karyotypic abnormalities and chromosomal instability are characteristic features of many cancers that are relatively resistant to chemotherapeutic agents currently used in the clinic. These same features represent potentially targetable "states" that are essentially tumor specific. The assessment of the chromosomal state of a cancer cell population may provide a guide for the selection or development of drugs active against aggressive and intractable cancers.
Collapse
Affiliation(s)
- Anna V Roschke
- Genetics Branch, Center for Cancer Research, National Cancer Institute, Building NNMC8, Room 5101, Bethesda, MD 20889-5105, USA.
| | | |
Collapse
|
19
|
Greshock J, Bachman KE, Degenhardt YY, Jing J, Wen YH, Eastman S, McNeil E, Moy C, Wegrzyn R, Auger K, Hardwicke MA, Wooster R. Molecular target class is predictive of in vitro response profile. Cancer Res 2010; 70:3677-86. [PMID: 20406975 DOI: 10.1158/0008-5472.can-09-3788] [Citation(s) in RCA: 97] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Preclinical cellular response profiling of tumor models has become a cornerstone in the development of novel cancer therapeutics. As efforts to predict clinical efficacy using cohorts of in vitro tumor models have been successful, expansive panels of tumor-derived cell lines can recapitulate an "all comers" efficacy trial, thereby identifying which tumors are most likely to benefit from treatment. The response profile of a therapy is most often studied in isolation; however, drug treatment effect patterns in tumor models across a diverse panel of compounds can help determine the value of unique molecular target classes in specific tumor cohorts. To this end, a panel of 19 compounds was evaluated against a diverse group of cancer cell lines (n = 311). The primary oncogenic targets were a key determinant of concentration-dependent proliferation response, as a total of five of six, four of four, and five of five phosphatidylinositol 3-kinase (PI3K)/AKT/mammalian target of rapamycin (mTOR) pathway, insulin-like growth factor-I receptor (IGF-IR), and mitotic inhibitors, respectively, clustered with others of that common target class. In addition, molecular target class was correlated with increased responsiveness in certain histologies. A cohort of PI3K/AKT/mTOR inhibitors was more efficacious in breast cancers compared with other tumor types, whereas IGF-IR inhibitors more selectively inhibited growth in colon cancer lines. Finally, specific phenotypes play an important role in cellular response profiles. For example, luminal breast cancer cells (nine of nine; 100%) segregated from basal cells (six of seven; 86%). The convergence of a common cellular response profile for different molecules targeting the same oncogenic pathway substantiates a rational clinical path for patient populations most likely to benefit from treatment. Cancer Res; 70(9); 3677-86. (c)2010 AACR.
Collapse
Affiliation(s)
- Joel Greshock
- Cancer Metabolism Drug Discovery, GlaxoSmithKline, Collegeville, Pennsylvania 19426, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Welsh M, Mangravite L, Medina MW, Tantisira K, Zhang W, Huang RS, McLeod H, Dolan ME. Pharmacogenomic discovery using cell-based models. Pharmacol Rev 2009; 61:413-29. [PMID: 20038569 PMCID: PMC2802425 DOI: 10.1124/pr.109.001461] [Citation(s) in RCA: 99] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Quantitative variation in response to drugs in human populations is multifactorial; genetic factors probably contribute to a significant extent. Identification of the genetic contribution to drug response typically comes from clinical observations and use of classic genetic tools. These clinical studies are limited by our inability to control environmental factors in vivo and the difficulty of manipulating the in vivo system to evaluate biological changes. Recent progress in dissecting genetic contribution to natural variation in drug response through the use of cell lines has been made and is the focus of this review. A general overview of current cell-based models used in pharmacogenomic discovery and validation is included. Discussion includes the current approach to translate findings generated from these cell-based models into the clinical arena and the use of cell lines for functional studies. Specific emphasis is given to recent advances emerging from cell line panels, including the International HapMap Project and the NCI60 cell panel. These panels provide a key resource of publicly available genotypic, expression, and phenotypic data while allowing researchers to generate their own data related to drug treatment to identify genetic variation of interest. Interindividual and interpopulation differences can be evaluated because human lymphoblastoid cell lines are available from major world populations of European, African, Chinese, and Japanese ancestry. The primary focus is recent progress in the pharmacogenomic discovery area through ex vivo models.
Collapse
Affiliation(s)
- Marleen Welsh
- Department of Medicine, University of Chicago, Chicago, Illinois 60637, USA
| | | | | | | | | | | | | | | |
Collapse
|
21
|
Portugal J. Evaluation of molecular descriptors for antitumor drugs with respect to noncovalent binding to DNA and antiproliferative activity. BMC Pharmacol 2009; 9:11. [PMID: 19758437 PMCID: PMC2758867 DOI: 10.1186/1471-2210-9-11] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2009] [Accepted: 09/16/2009] [Indexed: 11/29/2022] Open
Abstract
Background Small molecules that bind reversibly to DNA are among the antitumor drugs currently used in chemotherapy. In the pursuit of a more rational approach to cancer chemotherapy based upon these molecules, it is necessary to exploit the interdependency between DNA-binding affinity, sequence selectivity and cytotoxicity. For drugs binding noncovalently to DNA, it is worth exploring whether molecular descriptors, such as their molecular weight or the number of potential hydrogen acceptors/donors, can account for their DNA-binding affinity and cytotoxicity. Results Fifteen antitumor agents, which are in clinical use or being evaluated as part of the National Cancer Institute's drug screening effort, were analyzed in silico to assess the contribution of various molecular descriptors to their DNA-binding affinity, and the capacity of the descriptors and DNA-binding constants for predicting cell cytotoxicity. Equations to predict drug-DNA binding constants and growth-inhibitory concentrations were obtained by multiple regression following rigorous statistical procedures. Conclusion For drugs binding reversibly to DNA, both their strength of binding and their cytoxicity are fairly predicted from molecular descriptors by using multiple regression methods. The equations derived may be useful for rational drug design. The results obtained agree with that compounds more active across the National Cancer Institute's 60-cell line data set tend to have common structural features.
Collapse
Affiliation(s)
- José Portugal
- Instituto de Biología Molecular de Barcelona, CSIC, Parc Cientific de Barcelona, Baldiri Reixac, 10, E-08028 Barcelona, Spain.
| |
Collapse
|
22
|
Shedden K, Yang Y, Rosania G. Gene expression associations with the growth inhibitory effects of small molecules on live cells: specificity of effects and uniformity of mechanisms. Stat Anal Data Min 2009; 2:175-185. [PMID: 20657799 DOI: 10.1002/sam.10049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
The NCI60 human tumor cell line screen is a public resource for studying selective and non-selective growth inhibition of small molecules against cancer cells. By coupling growth inhibition screening data with biological characterizations of the different cell lines, it becomes possible to infer mechanisms of action underlying some of the observable patterns of selective activity. Using these data, mechanistic relationships have been identified including specific associations between single genes and small families of closely related compounds, and less specific relationships between biological processes involving several cooperating genes and broader families of compounds. Here we aim to characterize the degree to which such specific and general relationships are present in these data. A related question is whether genes tend to act with a uniform mechanism for all associated compounds, or whether multiple mechanisms are commonly involved. We address these two issues in a statistical framework placing special emphasis on the effects of measurement error in the gene expression and chemical screening data. We find that as measurement accuracy increases, the pattern of apparent associations shifts from one dominated by isolated gene/compound pairs, to one in which families consisting of an average of 25 compounds are associated to the same gene. At the same time, the number of genes that appear to play a role in influencing compound activities decreases. For less than half of the genes, the presence of both positive and negative correlations indicates pleiotropic associations with molecules via different mechanisms of action.
Collapse
Affiliation(s)
- Kerby Shedden
- Department of Statistics, University of Michigan, Ann Arbor MI USA
| | | | | |
Collapse
|
23
|
Berndtsson M, Hernlund E, Shoshan MC, Linder S. Conditional drug screening shows that mitotic inhibitors induce AKT/PKB-insensitive apoptosis. J Chem Biol 2009; 2:81-7. [PMID: 19568785 PMCID: PMC2701489 DOI: 10.1007/s12154-009-0017-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2008] [Revised: 02/09/2008] [Accepted: 02/15/2008] [Indexed: 12/27/2022] Open
Abstract
The phosphatidylinositol 3-kinase (PI3K)/AKT pathway is frequently upregulated in human cancer. Activation of this pathway has been reported to be associated with resistance to various chemotherapeutical agents. We here used a chemical biology/chemical informatic approach to identify apoptotic mechanisms that are insensitive to activation of the PI3K/AKT pathway. The National Cancer Institute (NCI) Mechanistic Set drug library was screened for agents that induce apoptosis in colon carcinoma cells expressing a constitutively active form of AKT1. The cytotoxicity screening data available as self-organized maps at the Developmental Therapeutics Program (DTP) of the NCI was then used to classify the identified compounds according to mechanism of action. The results showed that drugs that interfere with the mitotic process induce apoptosis which is comparatively insensitive to constitutive AKT1 activity. The conditional screening approach described here is expected to be useful for identifying relationships between the state of activation of signaling pathways and sensitivity to anticancer agents.
Collapse
Affiliation(s)
- Maria Berndtsson
- Cancer Center Karolinska, Department of Oncology and Pathology, Karolinska Institute and Hospital, Cancer Center Karolinska, R8:00, S-171 76, Stockholm, Sweden
| | | | | | | |
Collapse
|
24
|
Braun R, Cope L, Parmigiani G. Identifying differential correlation in gene/pathway combinations. BMC Bioinformatics 2008; 9:488. [PMID: 19017408 PMCID: PMC2613418 DOI: 10.1186/1471-2105-9-488] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2008] [Accepted: 11/18/2008] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND An important emerging trend in the analysis of microarray data is to incorporate known pathway information a priori. Expression level "summaries" for pathways, obtained from the expression data for the genes constituting the pathway, permit the inclusion of pathway information, reduce the high dimensionality of microarray data, and have the power to elucidate gene-interaction dependencies which are not already accounted for through known pathway identification. RESULTS We present a novel method for the analysis of microarray data that identifies joint differential expression in gene-pathway pairs. This method takes advantage of known gene pathway memberships to compute a summary expression level for each pathway as a whole. Correlations between the pathway expression summary and the expression levels of genes not already known to be associated with the pathway provide clues to gene interaction dependencies that are not already accounted for through known pathway identification, and statistically significant differences between gene-pathway correlations in phenotypically different cells (e.g., where the expression level of a single gene and a given pathway summary correlate strongly in normal cells but weakly in tumor cells) may indicate biologically relevant gene-pathway interactions. Here, we detail the methodology and present the results of this method applied to two gene-expression datasets, identifying gene-pathway pairs which exhibit differential joint expression by phenotype. CONCLUSION The method described herein provides a means by which interactions between large numbers of genes may be identified by incorporating known pathway information to reduce the dimensionality of gene interactions. The method is efficient and easily applied to data sets of ~102 arrays. Application of this method to two publicly-available cancer data sets yields suggestive and promising results. This method has the potential to complement gene-at-a-time analysis techniques for microarray analysis by indicating relationships between pathways and genes that have not previously been identified and which may play a role in disease.
Collapse
Affiliation(s)
- Rosemary Braun
- National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA.
| | | | | |
Collapse
|
25
|
Kuhn M, von Mering C, Campillos M, Jensen LJ, Bork P. STITCH: interaction networks of chemicals and proteins. Nucleic Acids Res 2007; 36:D684-8. [PMID: 18084021 PMCID: PMC2238848 DOI: 10.1093/nar/gkm795] [Citation(s) in RCA: 611] [Impact Index Per Article: 33.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
The knowledge about interactions between proteins and small molecules is essential for the understanding of molecular and cellular functions. However, information on such interactions is widely dispersed across numerous databases and the literature. To facilitate access to this data, STITCH (‘search tool for interactions of chemicals’) integrates information about interactions from metabolic pathways, crystal structures, binding experiments and drug–target relationships. Inferred information from phenotypic effects, text mining and chemical structure similarity is used to predict relations between chemicals. STITCH further allows exploring the network of chemical relations, also in the context of associated binding proteins. Each proposed interaction can be traced back to the original data sources. Our database contains interaction information for over 68 000 different chemicals, including 2200 drugs, and connects them to 1.5 million genes across 373 genomes and their interactions contained in the STRING database. STITCH is available at http://stitch.embl.de/
Collapse
Affiliation(s)
- Michael Kuhn
- European Molecular Biology Laboratory, Meyerhofstrasse 1, 69117 Heidelberg, Germany
| | | | | | | | | |
Collapse
|
26
|
Connecting chemosensitivity, gene expression and disease. Trends Pharmacol Sci 2007; 29:1-5. [PMID: 18055024 DOI: 10.1016/j.tips.2007.10.015] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2007] [Revised: 10/03/2007] [Accepted: 10/05/2007] [Indexed: 01/21/2023]
Abstract
Omics-based investigations offer potentially powerful readouts that might be useful for probing the underlying biology of normal and diseased states, identifying novel therapeutic targets and proposing relevant markers for designing treatment strategies. A vital component of these investigations involves a systematic analysis of gene expression and chemosensitivity data in the context of disease states and small molecule probes into the function of targets responsible for a disease phenotype. Systematic analysis of chemical and pharmacogenetics data offers a possible means to identify novel, small-molecule, potentially therapeutic, agents that affect the phenotype of a particular target. Elegantly simple in concept, the covariation of genetic and chemosensitivity readouts provide a hypothetical link for relating compounds through genomic expression profiles to underlying biology.
Collapse
|
27
|
Covell DG, Huang R, Wallqvist A. Anticancer medicines in development: assessment of bioactivity profiles within the National Cancer Institute anticancer screening data. Mol Cancer Ther 2007; 6:2261-70. [PMID: 17699723 DOI: 10.1158/1535-7163.mct-06-0787] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
We present an analysis of current anticancer compounds that are in phase I, II, or III clinical trials and their structural analogues that have been screened in the National Cancer Institute (NCI) anticancer screening program. Bioactivity profiles, measured across the NCI 60 cell lines, were examined for a correspondence between the type of cancer proposed for clinical testing and selective sensitivity to appropriately matched tumor subpanels in the NCI screen. These results find strongest support for using the NCI anticancer screen to select analogue compounds with selective sensitivity to the leukemia, colon, central nervous system, melanoma, and ovarian panels, but not for renal, prostate, and breast panels. These results are extended to applications of two-dimensional structural features to further refine compound selections based on tumor panel sensitivity obtained from tumor screening results.
Collapse
Affiliation(s)
- David G Covell
- National Cancer Institute-Frederick, Developmental Therapeutics Program, Screening Technologies Branch, Laboratory of Computational Technologies, Frederick, MD 21702, USA.
| | | | | |
Collapse
|
28
|
Wallqvist A, Huang R, Covell DG. Chemoinformatic analysis of NCI preclinical tumor data: evaluating compound efficacy from mouse xenograft data, NCI-60 screening data, and compound descriptors. J Chem Inf Model 2007; 47:1414-27. [PMID: 17555311 DOI: 10.1021/ci700132u] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
We provide a chemoinformatic examination of the NCI public human tumor xenograft data to explore relationships between small molecules, treatment modality, efficacy, and toxicity. Efficacy endpoints of tumor weight reduction (TW) and survival time increase (ST) compared to tumor bearing control mice were augmented by a toxicity measure, defined as the survival advantage of treated versus control animals (TX). These endpoints were used to define two independent therapeutic indices (TIs) as the ratio of efficacy (TW or ST) to toxicity (TX). Linear models predictive of xenograft endpoints were successfully constructed (0.67 < r(2) < or = 0.74)(observed_versus_predicted) using a model comprised of variables in treatment modality, chemoinformatic descriptors, and in vitro cell growth inhibition in the NCI 60-cell assay. Cross-validation analysis based on randomly chosen training subsets found these predictive correlations to be robust. Model-based sensitivity analysis found chemistry and growth inhibition to provide the best, and treatment modality the worst, indicators of xenograft endpoint. The poor predictive power derived from treatment alone appears to be of less importance to xenograft outcome for compounds having strongly similar chemical and biological features. ROC-based model validation found a 70% positive predictive value for distinguishing FDA approved oncology agents from available xenograft tested compounds. Additional chemoinformatic applications are provided that relate xenograft outcome to biological pathways and putative mechanism of compound action. These results find a strong relationship between xenograft efficacy and pathways comprised of genes having highly correlated mRNA expressions. Our analysis demonstrates that chemoinformatic studies utilizing a combination of xenograft data and in vitro preclinical testing offer an effective means to identify compound classes with superior efficacy and reduced toxicity.
Collapse
Affiliation(s)
- Anders Wallqvist
- Laboratory of Computational Technologies, SAIC-Frederick, Inc., NCI-Frederick, Frederick, Maryland 21702, USA.
| | | | | |
Collapse
|
29
|
Rosania GR, Crippen G, Woolf P, States D, Shedden K. A Cheminformatic Toolkit for Mining Biomedical Knowledge. Pharm Res 2007; 24:1791-802. [PMID: 17385012 DOI: 10.1007/s11095-007-9285-5] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2007] [Accepted: 02/27/2007] [Indexed: 01/31/2023]
Abstract
PURPOSE Cheminformatics can be broadly defined to encompass any activity related to the application of information technology to the study of properties, effects and uses of chemical agents. One of the most important current challenges in cheminformatics is to allow researchers to search databases of biomedical knowledge, using chemical structures as input. MATERIALS AND METHODS An important step towards this goal was the establishment of PubChem, an open, centralized database of small molecules accessible through the World Wide Web. While PubChem is primarily intended to serve as a repository for high throughput screening data from federally-funded screening centers and academic research laboratories, the major impact of PubChem could also reside in its ability to serve as a chemical gateway to biomedical databases such as PubMed. CONCLUSION This article will review cheminformatic tools that can be applied to facilitate annotation of PubChem through links to the scientific literature; to integrate PubChem with transcriptomic, proteomic, and metabolomic datasets; to incorporate results of numerical simulations of physiological systems into PubChem annotation; and ultimately, to translate data of chemical genomics screening efforts into information that will benefit biomedical researchers and physician scientists across all therapeutic areas.
Collapse
Affiliation(s)
- Gus R Rosania
- Department of Pharmaceutical Sciences, University of Michigan College of Pharmacy, 428 Church Street, Ann Arbor, MI 48109, USA.
| | | | | | | | | |
Collapse
|
30
|
Huang R, Wallqvist A, Covell DG. Targeting changes in cancer: assessing pathway stability by comparing pathway gene expression coherence levels in tumor and normal tissues. Mol Cancer Ther 2006; 5:2417-27. [PMID: 16985076 DOI: 10.1158/1535-7163.mct-06-0239] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The purpose of this study is to examine gene expression changes occurring in cancer from a pathway perspective by analyzing the level of pathway coherence in tumor tissues in comparison with their normal counterparts. Instability in pathway regulation patterns can be considered either as a result of or as a contributing factor to genetic instability and possibly cancer. Our analysis has identified pathways that show a significant change in their coherence level in tumor tissues, some of which are tumor type specific, indicating novel targets for cancer type-specific therapies. Pathways are found to have a general tendency to lose their gene expression coherence in tumor tissues when compared with normal tissues, especially for signaling pathways. The selective growth advantage of cancer cells over normal cells seems to originate from their preserved control over vital pathways to ensure survival and altered signaling, allowing excessive proliferation. We have additionally investigated the tissue-related instability of pathways, providing valuable clues to the cellular processes underlying the tumorigenesis and/or growth of specific cancer types. Pathways that contain known cancer genes (i.e., "cancer pathways") show significantly greater instability and are more likely to become incoherent in tumor tissues. Finally, we have proposed strategies to target instability (i.e., pathways that are prone to changes) by identifying compound groups that show selective activity against pathways with a detectable coherence change in cancer. These results can serve as guidelines for selecting novel agents that have the potential to specifically target a particular pathway that has relevance in cancer.
Collapse
Affiliation(s)
- Ruili Huang
- Developmental Therapeutics Program, Screening Technologies Branch, Laboratory of Computational Technologies, National Cancer Institute-Frederick, Frederick, MD 21702, USA
| | | | | |
Collapse
|