1
|
Muthuramalingam P, Akassh S, Rithiga SB, Prithika S, Gunasekaran R, Shin H, Kumar R, Baskar V, Kim J. Integrated omics profiling and network pharmacology uncovers the prognostic genes and multi-targeted therapeutic bioactives to combat lung cancer. Eur J Pharmacol 2023; 940:175479. [PMID: 36566006 DOI: 10.1016/j.ejphar.2022.175479] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 12/07/2022] [Accepted: 12/20/2022] [Indexed: 12/24/2022]
Abstract
Non-small cell lung cancer (NSCLC) is the frequent subtype of lung cancer and the currently used treatment methods, diagnosis, and chemoresistance are relatively ineffective. Determining the pharmacological targets from active biomolecules of medicinal plants has become a frontiers era for biomedical research to develop novel therapies. In view of these scenarios, this pilot study, network pharmacology, cheminformatics, integrative omics, molecular docking and in vitro anti-cancer analysis were performed to unveil the multi-targeted treatment mechanisms of novel plant bioactives to treat lung cancer. Bioactive molecules from medicinal plants were compiled from PubChem. Network pharmacology approach revealed that 29 compounds efficiently target the 390 human and lung cancer associated genes. In addition, comparative analysis was performed and identified the 7 bioactive molecules significantly targeting 8 lung cancer genes. The integrative omics analysis discovered unique genes between the lung cancer and normal lung tissues. These genes were further validated through protein-protein interaction, gene ontology, gene functional and pathway enrichment, boxplot and overall survival analyses to understand the function of unique genes and their involvement in cancer signaling pathways. Survival heatmap analyses identified the significant prognostic genes. Docking results revealed that, lupeol and p-coumaric acid displayed high binding affinities with MIF, CCNB1, FABP4. Hence, we selected these two bioactives for in vitro analysis. Furthermore, these selected bioactives were showed concentration dependent cytotoxicity against the lung adenocarcinoma cells (A549). This holistic study has opened up novel avenues and unravels the cancer prognostic genes which could serve as druggable target and bioactives with anti-cancerous efficacy. Further functional validations are prerequisites to deciphering these bioactives as commercial drug candidates.
Collapse
Affiliation(s)
- Pandiyan Muthuramalingam
- Division of Horticultural Science, College of Agriculture and Life Sciences, Gyeongsang National University, Jinju, 52725, South Korea; Department of GreenBio Science, Gyeongsang National University, Jinju, 52725, South Korea; Department of Biotechnology, Sri Shakthi Institute of Engineering and Technology, Coimbatore, 641 062, India.
| | - Sakthivel Akassh
- Department of Biotechnology, Sri Shakthi Institute of Engineering and Technology, Coimbatore, 641 062, India
| | | | - Senthilkumar Prithika
- Department of Biotechnology, Sri Shakthi Institute of Engineering and Technology, Coimbatore, 641 062, India
| | - Ravikumar Gunasekaran
- Department of Biotechnology, Sri Shakthi Institute of Engineering and Technology, Coimbatore, 641 062, India
| | - Hyunsuk Shin
- Division of Horticultural Science, College of Agriculture and Life Sciences, Gyeongsang National University, Jinju, 52725, South Korea; Department of GreenBio Science, Gyeongsang National University, Jinju, 52725, South Korea.
| | - Reetesh Kumar
- Department of Agricultural Sciences, Institute of Applied Sciences & Humanities, GLA University, Mathura, 281 406, India
| | - Venkidasamy Baskar
- Department of Oral and Maxillofacial Surgery, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences (SIMATS), Saveetha University, Chennai, 600 077, India
| | - Jinwook Kim
- Division of Horticultural Science, College of Agriculture and Life Sciences, Gyeongsang National University, Jinju, 52725, South Korea; Department of GreenBio Science, Gyeongsang National University, Jinju, 52725, South Korea
| |
Collapse
|
2
|
Munro LJ, Kell DB. Intelligent host engineering for metabolic flux optimisation in biotechnology. Biochem J 2021; 478:3685-3721. [PMID: 34673920 PMCID: PMC8589332 DOI: 10.1042/bcj20210535] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2021] [Revised: 09/22/2021] [Accepted: 09/24/2021] [Indexed: 12/13/2022]
Abstract
Optimising the function of a protein of length N amino acids by directed evolution involves navigating a 'search space' of possible sequences of some 20N. Optimising the expression levels of P proteins that materially affect host performance, each of which might also take 20 (logarithmically spaced) values, implies a similar search space of 20P. In this combinatorial sense, then, the problems of directed protein evolution and of host engineering are broadly equivalent. In practice, however, they have different means for avoiding the inevitable difficulties of implementation. The spare capacity exhibited in metabolic networks implies that host engineering may admit substantial increases in flux to targets of interest. Thus, we rehearse the relevant issues for those wishing to understand and exploit those modern genome-wide host engineering tools and thinking that have been designed and developed to optimise fluxes towards desirable products in biotechnological processes, with a focus on microbial systems. The aim throughput is 'making such biology predictable'. Strategies have been aimed at both transcription and translation, especially for regulatory processes that can affect multiple targets. However, because there is a limit on how much protein a cell can produce, increasing kcat in selected targets may be a better strategy than increasing protein expression levels for optimal host engineering.
Collapse
Affiliation(s)
- Lachlan J. Munro
- Novo Nordisk Foundation Centre for Biosustainability, Technical University of Denmark, Building 220, Kemitorvet, 2800 Kgs. Lyngby, Denmark
| | - Douglas B. Kell
- Novo Nordisk Foundation Centre for Biosustainability, Technical University of Denmark, Building 220, Kemitorvet, 2800 Kgs. Lyngby, Denmark
- Department of Biochemistry and Systems Biology, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Crown St, Liverpool L69 7ZB, U.K
- Mellizyme Biotechnology Ltd, IC1, Liverpool Science Park, 131 Mount Pleasant, Liverpool L3 5TF, U.K
| |
Collapse
|
3
|
Portelinha A, Thompson S, Smith RA, Da Silva Ferreira M, Asgari Z, Knezevic A, Seshan V, de Stanchina E, Gupta S, Denis L, Younes A, Reddy S. ASN007 is a selective ERK1/2 inhibitor with preferential activity against RAS-and RAF-mutant tumors. Cell Rep Med 2021; 2:100350. [PMID: 34337566 PMCID: PMC8324497 DOI: 10.1016/j.xcrm.2021.100350] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 12/21/2020] [Accepted: 06/22/2021] [Indexed: 12/30/2022]
Abstract
Inhibition of the extracellular signal-regulated kinases ERK1 and ERK2 (ERK1/2) offers a promising therapeutic strategy in cancers harboring activated RAS/RAF/MEK/ERK signaling pathways. Here, we describe an orally bioavailable and selective ERK1/2 inhibitor, ASN007, currently in clinical development for the treatment of cancer. In preclinical studies, ASN007 shows strong antiproliferative activity in tumors harboring mutations in BRAF and RAS (KRAS, NRAS, and HRAS). ASN007 demonstrates activity in a BRAFV600E mutant melanoma tumor model that is resistant to BRAF and MEK inhibitors. The PI3K inhibitor copanlisib enhances the antiproliferative activity of ASN007 both in vitro and in vivo due to dual inhibition of RAS/MAPK and PI3K survival pathways. Our data provide a rationale for evaluating ASN007 in RAS/RAF-driven tumors as well as a mechanistic basis for combining ASN007 with PI3K inhibitors.
Collapse
Affiliation(s)
- Ana Portelinha
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | | | | | | - Zahra Asgari
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Andrea Knezevic
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Venkatraman Seshan
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Elisa de Stanchina
- Antitumor Assessment Core, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | | | | - Anas Younes
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Lymphoma Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | |
Collapse
|
4
|
Halder V, McDonnell B, Uthayakumar D, Usher J, Shapiro RS. Genetic interaction analysis in microbial pathogens: unravelling networks of pathogenesis, antimicrobial susceptibility and host interactions. FEMS Microbiol Rev 2021; 45:fuaa055. [PMID: 33145589 DOI: 10.1093/femsre/fuaa055] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Accepted: 10/16/2020] [Indexed: 12/13/2022] Open
Abstract
Genetic interaction (GI) analysis is a powerful genetic strategy that analyzes the fitness and phenotypes of single- and double-gene mutant cells in order to dissect the epistatic interactions between genes, categorize genes into biological pathways, and characterize genes of unknown function. GI analysis has been extensively employed in model organisms for foundational, systems-level assessment of the epistatic interactions between genes. More recently, GI analysis has been applied to microbial pathogens and has been instrumental for the study of clinically important infectious organisms. Here, we review recent advances in systems-level GI analysis of diverse microbial pathogens, including bacterial and fungal species. We focus on important applications of GI analysis across pathogens, including GI analysis as a means to decipher complex genetic networks regulating microbial virulence, antimicrobial drug resistance and host-pathogen dynamics, and GI analysis as an approach to uncover novel targets for combination antimicrobial therapeutics. Together, this review bridges our understanding of GI analysis and complex genetic networks, with applications to diverse microbial pathogens, to further our understanding of virulence, the use of antimicrobial therapeutics and host-pathogen interactions. .
Collapse
Affiliation(s)
- Viola Halder
- Department of Molecular and Cellular Biology, University of Guelph, 50 Stone Road East, Guelph, ON, N1G 2W1, Canada
| | - Brianna McDonnell
- Department of Molecular and Cellular Biology, University of Guelph, 50 Stone Road East, Guelph, ON, N1G 2W1, Canada
| | - Deeva Uthayakumar
- Department of Molecular and Cellular Biology, University of Guelph, 50 Stone Road East, Guelph, ON, N1G 2W1, Canada
| | - Jane Usher
- Medical Research Council Centre for Medical Mycology, University of Exeter, Geoffrey Pope Building, Stocker Road, Exeter EX4 4QD, UK
| | - Rebecca S Shapiro
- Department of Molecular and Cellular Biology, University of Guelph, 50 Stone Road East, Guelph, ON, N1G 2W1, Canada
| |
Collapse
|
5
|
Yilmaz S, Tastan O, Cicek AE. SPADIS: An Algorithm for Selecting Predictive and Diverse SNPs in GWAS. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2021; 18:1208-1216. [PMID: 31443041 DOI: 10.1109/tcbb.2019.2935437] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Phenotypic heritability of complex traits and diseases is seldom explained by individual genetic variants identified in genome-wide association studies (GWAS). Many methods have been developed to select a subset of variant loci, which are associated with or predictive of the phenotype. Selecting connected SNPs on SNP-SNP networks have been proven successful in finding biologically interpretable and predictive SNPs. However, we argue that the connectedness constraint favors selecting redundant features that affect similar biological processes and therefore does not necessarily yield better predictive performance. In this paper, we propose a novel method called SPADIS that favors the selection of remotely located SNPs in order to account for their complementary effects in explaining a phenotype. SPADIS selects a diverse set of loci on a SNP-SNP network. This is achieved by maximizing a submodular set function with a greedy algorithm that ensures a constant factor approximation to the optimal solution. We compare SPADIS to the state-of-the-art method SConES, on a dataset of Arabidopsis Thaliana with continuous flowering time phenotypes. SPADIS has better average phenotype prediction performance in 15 out of 17 phenotypes when the same number of SNPs are selected and provides consistent improvements across multiple networks and settings on average. Moreover, it identifies more candidate genes and runs faster.
Collapse
|
6
|
Paran Y, Liron Y, Batsir S, Mabjeesh N, Geiger B, Kam Z. Multi-parametric characterization of drug effects on cells. F1000Res 2021; 9. [PMID: 33363713 PMCID: PMC7737707 DOI: 10.12688/f1000research.26254.2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/13/2021] [Indexed: 12/28/2022] Open
Abstract
We present here a novel multi-parametric approach for the characterization of multiple cellular features, using images acquired by high-throughput and high-definition light microscopy. We specifically used this approach for deep and unbiased analysis of the effects of a drug library on five cultured cell lines. The presented method enables the acquisition and analysis of millions of images, of treated and control cells, followed by an automated identification of drugs inducing strong responses, evaluating the median effect concentrations and those cellular properties that are most highly affected by the drug. The tools described here provide standardized quantification of multiple attributes for systems level dissection of complex functions in normal and diseased cells, using multiple perturbations. Such analysis of cells, derived from pathological samples, may help in the diagnosis and follow-up of treatment in patients.
Collapse
Affiliation(s)
- Yael Paran
- Department of Molecular Cell Biology, The Weizmann Institute of Science, Rehovot, 76100, Israel.,IDEA Biomedical Ltd., Rehovot, 76705, Israel
| | - Yuvalal Liron
- Department of Molecular Cell Biology, The Weizmann Institute of Science, Rehovot, 76100, Israel
| | - Sarit Batsir
- Department of Molecular Cell Biology, The Weizmann Institute of Science, Rehovot, 76100, Israel
| | - Nicola Mabjeesh
- Department of Urology, Tel Aviv Sourasky Medical Center, Tel Aviv, 64239, Israel
| | - Benjamin Geiger
- Department of Molecular Cell Biology, The Weizmann Institute of Science, Rehovot, 76100, Israel.,Department of Immunology, The Weizmann Institute of Science, Rehovot, 76100, Israel
| | - Zvi Kam
- Department of Molecular Cell Biology, The Weizmann Institute of Science, Rehovot, 76100, Israel
| |
Collapse
|
7
|
Ayaz Z, Zainab B, Khan S, Abbasi AM, Elshikh MS, Munir A, Al-Ghamdi AA, Alajmi AH, Alsubaie QD, Mustafa AEZMA. In silico authentication of amygdalin as a potent anticancer compound in the bitter kernels of family Rosaceae. Saudi J Biol Sci 2020; 27:2444-2451. [PMID: 32884428 PMCID: PMC7451698 DOI: 10.1016/j.sjbs.2020.06.041] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 06/15/2020] [Accepted: 06/23/2020] [Indexed: 02/07/2023] Open
Abstract
Amygdalin a naturally occurring compound, predominantly in the bitter kernels of apricot, almond, apple and other members of Rosaceae family. Though, amygdalin is used as an alternative therapy to treat various types of cancer but its role in cancer pathways has rarely been explored yet. Therefore, present study was intended with the aim to investigate the alleged anti-cancerous effects of amygdalin specifically on PI3K-AKT-mTOR and Ras pathways of cancer in human body. Computational modelling and simulation techniques were used to assess the effect of amygdalin on PI3K-AKT-mTOR and Ras pathways using different level of dosage. It was observed that amygdalin had direct and substantial contribution to regulate PI3K-mTOR activities on threshold levels while the other caner pathways were effected indirectly. Consequently, amygdalin is a down-regulator of a cancer within a specified amount and contribute considerably to reduce various types of cancer in human. Furthermore, in-vitro and in-vivo analyses of amygdalin could be of helpful to authenticate its pharmacological effects.
Collapse
Affiliation(s)
- Zainab Ayaz
- Department of Bioinformatics, Govt. Post Graduate College Mandian Abbottabad, Pakistan.,Department of Environmental Sciences, COMSATS University Islamabad, Abbottabad Campus, 22060, Pakistan
| | - Bibi Zainab
- Department of Bioinformatics, Govt. Post Graduate College Mandian Abbottabad, Pakistan.,Department of Environmental Sciences, COMSATS University Islamabad, Abbottabad Campus, 22060, Pakistan
| | - Sajid Khan
- Department of Bioinformatics, Govt. Post Graduate College Mandian Abbottabad, Pakistan
| | - Arshad Mehmood Abbasi
- Department of Environmental Sciences, COMSATS University Islamabad, Abbottabad Campus, 22060, Pakistan
| | - Mohamed S Elshikh
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Anum Munir
- Department of Bioinformatics, Govt. Post Graduate College Mandian Abbottabad, Pakistan.,Department of Bioinformatics and Biosciences, Capital University of Science and Technology Islamabad, Pakistan
| | - Abdullah Ahmed Al-Ghamdi
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Amal H Alajmi
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Qasi D Alsubaie
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Abd El-Zaher M A Mustafa
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh, Saudi Arabia.,Botany Department, Faculty of Science, Tanta University, Tanta, Egypt
| |
Collapse
|
8
|
Trajčíková E, Kurin E, Slobodníková L, Straka M, Lichváriková A, Dokupilová S, Čičová I, Nagy M, Mučaji P, Bittner Fialová S. Antimicrobial and Antioxidant Properties of Four Lycopus Taxa and an Interaction Study of Their Major Compounds. Molecules 2020; 25:E1422. [PMID: 32245012 PMCID: PMC7144923 DOI: 10.3390/molecules25061422] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 03/13/2020] [Accepted: 03/17/2020] [Indexed: 11/17/2022] Open
Abstract
The compositions of leaf infusions of three genotypes of Lycopus europaeus L. with origins in central Europe, namely L. europaeus A (LeuA), L. europaeus B (LeuB), and L. europaeus C (LeuC), and one genotype of L. exaltatus (Lex), were examined by LC-MS-DAD (Liquid Chromatography Mass Spectrometry and Diode Array Detection) analysis. This revealed the presence of thirteen compounds belonging to the groups of phenolic acids and flavonoids, with a predominance of rosmarinic acid (RA) and luteolin-7-O-glucuronide (LGlr). The antimicrobial activity of leaf infusions was tested on the collection strains of Gram-positive and Gram-negative bacteria, and on the clinical Staphylococcus aureus strains. We detected higher activity against Gram-positive bacteria, of which the most susceptible strains were those of Staphylococcus aureus, including methicillin-resistant and poly-resistant strains. Furthermore, we examined the antioxidant activity of leaf infusions using 2,2-diphenyl-1-picrylhydrazyl (DPPH) and 2,2'-azino-bis(3-ethylbenzothiazoline-6-sulphonic acid) (ABTS) methods, and on NIH/3T3 cell lines using dichlorodihydrofluorescein diacetate (DCFH-DA). We also studied the mutual interactions between selected infusions, namely RA and/or LGlr. In the mixtures of leaf infusion and RA or LGlr, we observed slight synergism and a high dose reduction index in most cases. This leads to the beneficial dose reduction at a given antioxidant effect level in mixtures compared to the doses of the parts used alone. Therefore, our study draws attention to further applications of the Lycopus leaves as a valuable alternative source of natural antioxidants and as a promising topical antibacterial agent for medicinal use.
Collapse
Affiliation(s)
- Eva Trajčíková
- Department of Pharmacognosy and Botany, Faculty of Pharmacy, Comenius University in Bratislava, Odbojárov 10, 832 32 Bratislava, Slovakia; (E.T.); (E.K.); (M.N.); (P.M.)
| | - Elena Kurin
- Department of Pharmacognosy and Botany, Faculty of Pharmacy, Comenius University in Bratislava, Odbojárov 10, 832 32 Bratislava, Slovakia; (E.T.); (E.K.); (M.N.); (P.M.)
| | - Lívia Slobodníková
- Institute of Microbiology, Faculty of Medicine and the University Hospital in Bratislava, Comenius University in Bratislava, Sasinkova 4, 811 08 Bratislava, Slovakia; (L.S.); (M.S.)
| | - Marek Straka
- Institute of Microbiology, Faculty of Medicine and the University Hospital in Bratislava, Comenius University in Bratislava, Sasinkova 4, 811 08 Bratislava, Slovakia; (L.S.); (M.S.)
- Department of Microbiology and Virology, Faculty of Natural Sciences, Comenius University in Bratislava, Mlynská dolina, Ilkovičova 6, 842 15 Bratislava, Slovakia
| | - Aneta Lichváriková
- Department of Molecular Biology, Faculty of Natural Sciences, Comenius University in Bratislava, Mlynská dolina, Ilkovičova 6, 842 15 Bratislava, Slovakia;
| | - Svetlana Dokupilová
- Department of Pharmaceutical Analysis and Nuclear Pharmacy, Faculty of Pharmacy, Comenius University in Bratislava, Odbojárov 10, 832 32 Bratislava, Slovakia;
| | - Iveta Čičová
- National Agricultural and Food Centre, Research Institute of Plant Production, 92168 Piešťany, Slovakia;
| | - Milan Nagy
- Department of Pharmacognosy and Botany, Faculty of Pharmacy, Comenius University in Bratislava, Odbojárov 10, 832 32 Bratislava, Slovakia; (E.T.); (E.K.); (M.N.); (P.M.)
| | - Pavel Mučaji
- Department of Pharmacognosy and Botany, Faculty of Pharmacy, Comenius University in Bratislava, Odbojárov 10, 832 32 Bratislava, Slovakia; (E.T.); (E.K.); (M.N.); (P.M.)
| | - Silvia Bittner Fialová
- Department of Pharmacognosy and Botany, Faculty of Pharmacy, Comenius University in Bratislava, Odbojárov 10, 832 32 Bratislava, Slovakia; (E.T.); (E.K.); (M.N.); (P.M.)
| |
Collapse
|
9
|
Weiss A, Le Roux-Bourdieu M, Zoetemelk M, Ramzy GM, Rausch M, Harry D, Miljkovic-Licina M, Falamaki K, Wehrle-Haller B, Meraldi P, Nowak-Sliwinska P. Identification of a Synergistic Multi-Drug Combination Active in Cancer Cells via the Prevention of Spindle Pole Clustering. Cancers (Basel) 2019; 11:E1612. [PMID: 31652588 PMCID: PMC6826636 DOI: 10.3390/cancers11101612] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 10/14/2019] [Accepted: 10/16/2019] [Indexed: 02/06/2023] Open
Abstract
A major limitation of clinically used cancer drugs is the lack of specificity resulting in toxicity. To address this, we performed a phenotypically-driven screen to identify optimal multidrug combinations acting with high efficacy and selectivity in clear cell renal cell carcinoma (ccRCC). The search was performed using the Therapeutically Guided Multidrug Optimization (TGMO) method in ccRCC cells (786-O) and nonmalignant renal cells and identified a synergistic low-dose four-drug combination (C2) with high efficacy and negligible toxicity. We discovered that C2 inhibits multipolar spindle pole clustering, a survival mechanism employed by cancer cells with spindle abnormalities. This phenotype was also observed in 786-O cells resistant to sunitinib, the first line ccRCC treatment, as well as in melanoma cells with distinct percentages of supernumerary centrosomes. We conclude that C2-treatment shows a high efficacy in cells prone to form multipolar spindles. Our data suggest a highly effective and selective C2 treatment strategy for malignant and drug-resistant cancers.
Collapse
Affiliation(s)
- Andrea Weiss
- Institute of Pharmaceutical Sciences of Western Switzerland, Faculty of Sciences, University of Geneva, 1 Rue Michel-Servet, CMU, 1211 Geneva 4, Switzerland.
- Translational Research Centre in Oncohaematology, 1 Rue Michel-Servet, CMU, 1211 Geneva 4, Switzerland.
| | - Morgan Le Roux-Bourdieu
- Translational Research Centre in Oncohaematology, 1 Rue Michel-Servet, CMU, 1211 Geneva 4, Switzerland.
- Department of Cell Physiology and Metabolism, University of Geneva Medical School, 1 Rue Michel-Servet, CMU, 1211 Geneva 4, Switzerland.
| | - Marloes Zoetemelk
- Institute of Pharmaceutical Sciences of Western Switzerland, Faculty of Sciences, University of Geneva, 1 Rue Michel-Servet, CMU, 1211 Geneva 4, Switzerland.
- Translational Research Centre in Oncohaematology, 1 Rue Michel-Servet, CMU, 1211 Geneva 4, Switzerland.
| | - George M Ramzy
- Institute of Pharmaceutical Sciences of Western Switzerland, Faculty of Sciences, University of Geneva, 1 Rue Michel-Servet, CMU, 1211 Geneva 4, Switzerland.
| | - Magdalena Rausch
- Institute of Pharmaceutical Sciences of Western Switzerland, Faculty of Sciences, University of Geneva, 1 Rue Michel-Servet, CMU, 1211 Geneva 4, Switzerland.
- Translational Research Centre in Oncohaematology, 1 Rue Michel-Servet, CMU, 1211 Geneva 4, Switzerland.
| | - Daniela Harry
- Department of Cell Physiology and Metabolism, University of Geneva Medical School, 1 Rue Michel-Servet, CMU, 1211 Geneva 4, Switzerland.
| | - Marijana Miljkovic-Licina
- Translational Research Centre in Oncohaematology, 1 Rue Michel-Servet, CMU, 1211 Geneva 4, Switzerland.
- Department of Pathology and Immunology, University of Geneva Medical School, 1 Rue Michel-Servet, CMU, 1211 Geneva 4, Switzerland.
| | - Katayoun Falamaki
- Department of Cell Physiology and Metabolism, University of Geneva Medical School, 1 Rue Michel-Servet, CMU, 1211 Geneva 4, Switzerland.
| | - Bernard Wehrle-Haller
- Translational Research Centre in Oncohaematology, 1 Rue Michel-Servet, CMU, 1211 Geneva 4, Switzerland.
- Department of Cell Physiology and Metabolism, University of Geneva Medical School, 1 Rue Michel-Servet, CMU, 1211 Geneva 4, Switzerland.
| | - Patrick Meraldi
- Translational Research Centre in Oncohaematology, 1 Rue Michel-Servet, CMU, 1211 Geneva 4, Switzerland.
- Department of Cell Physiology and Metabolism, University of Geneva Medical School, 1 Rue Michel-Servet, CMU, 1211 Geneva 4, Switzerland.
| | - Patrycja Nowak-Sliwinska
- Institute of Pharmaceutical Sciences of Western Switzerland, Faculty of Sciences, University of Geneva, 1 Rue Michel-Servet, CMU, 1211 Geneva 4, Switzerland.
- Translational Research Centre in Oncohaematology, 1 Rue Michel-Servet, CMU, 1211 Geneva 4, Switzerland.
| |
Collapse
|
10
|
Kim SJ, Ha JW, Kim H, Zhang BT. Bayesian evolutionary hypernetworks for interpretable learning from high-dimensional data. Appl Soft Comput 2019. [DOI: 10.1016/j.asoc.2019.05.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|
11
|
Du D, Chang CH, Wang Y, Tong P, Chan WK, Chiu Y, Peng B, Tan L, Weinstein JN, Lorenzi PL. Response envelope analysis for quantitative evaluation of drug combinations. Bioinformatics 2019; 35:3761-3770. [PMID: 30851108 PMCID: PMC7963081 DOI: 10.1093/bioinformatics/btz091] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Revised: 01/21/2019] [Accepted: 03/07/2019] [Indexed: 02/02/2023] Open
Abstract
MOTIVATION The concept of synergy between two agents, over a century old, is important to the fields of biology, chemistry, pharmacology and medicine. A key step in drug combination analysis is the selection of an additivity model to identify combination effects including synergy, additivity and antagonism. Existing methods for identifying and interpreting those combination effects have limitations. RESULTS We present here a computational framework, termed response envelope analysis (REA), that makes use of 3D response surfaces formed by generalized Loewe Additivity and Bliss Independence models of interaction to evaluate drug combination effects. Because the two models imply two extreme limits of drug interaction (mutually exclusive and mutually non-exclusive), a response envelope defined by them provides a quantitatively stringent additivity model for identifying combination effects without knowing the inhibition mechanism. As a demonstration, we apply REA to representative published data from large screens of anticancer and antibiotic combinations. We show that REA is more accurate than existing methods and provides more consistent results in the context of cross-experiment evaluation. AVAILABILITY AND IMPLEMENTATION The open-source software package associated with REA is available at: https://github.com/4dsoftware/rea. SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Di Du
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Chia-Hua Chang
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Yumeng Wang
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Pan Tong
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Wai Kin Chan
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Yulun Chiu
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Bo Peng
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Lin Tan
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - John N Weinstein
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | |
Collapse
|
12
|
Cava C, Castiglioni I. In silico perturbation of drug targets in pan-cancer analysis combining multiple networks and pathways. Gene 2019; 698:100-106. [PMID: 30840853 DOI: 10.1016/j.gene.2019.02.064] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Revised: 02/13/2019] [Accepted: 02/23/2019] [Indexed: 12/13/2022]
Abstract
The knowledge of cancer cell response to conventional therapies is crucial in order to choose the correct therapy of patients affected by cancer. The major problem is generally attributed to the lack of specific biological processes able to predict the therapy efficacy. Here, we optimized a computational method for the analysis of gene networks able to detect and quantify the effects of a drug in a pan-cancer study. Overall, our method, using several network topological measures has identified a cancer gene network with a key role in biological processes. The gene network, able to classify with a good performance cancer vs normal samples, was modulated in silico to evaluate the effects of new or approved drugs. This computational model could offer an interesting hint to decipher molecular mechanisms contributing to resistance or inefficacy of drugs.
Collapse
Affiliation(s)
- Claudia Cava
- Institute of Molecular Bioimaging and Physiology, National Research Council (IBFM-CNR), Via F.Cervi 93, 20090 Segrate, Milan, Italy.
| | - Isabella Castiglioni
- Institute of Molecular Bioimaging and Physiology, National Research Council (IBFM-CNR), Via F.Cervi 93, 20090 Segrate, Milan, Italy.
| |
Collapse
|
13
|
Cokol-Cakmak M, Bakan F, Cetiner S, Cokol M. Diagonal Method to Measure Synergy Among Any Number of Drugs. J Vis Exp 2018. [PMID: 29985330 PMCID: PMC6101960 DOI: 10.3791/57713] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
A synergistic drug combination has a higher efficacy compared to the effects of individual drugs. Checkerboard assays, where drugs are combined in many doses, allow sensitive measurement of drug interactions. However, these assays are costly and do not scale well for measuring interaction among many drugs. Several recent studies have reported drug interaction measurements using a diagonal sampling of the traditional checkerboard assay. This alternative methodology greatly decreases the cost of drug interaction experiments and allows interaction measurement for combinations with many drugs. Here, we describe a protocol to measure the three pairwise interactions and one three-way interaction among three antibiotics in duplicate, in five days, using only three 96-well microplates and standard laboratory equipment. We present representative results showing that the three-antibiotic combination of Levofloxacin + Nalidixic Acid + Penicillin G is synergistic. Our protocol scales up to measure interactions among many drugs and in other biological contexts, allowing for efficient screens for multi-drug synergies against pathogens and tumors.
Collapse
Affiliation(s)
| | - Feray Bakan
- Nanotechnology Research and Application Center, Sabanci University
| | - Selim Cetiner
- Faculty of Engineering and Natural Sciences, Sabanci University
| | - Murat Cokol
- Faculty of Engineering and Natural Sciences, Sabanci University; Nanotechnology Research and Application Center, Sabanci University; Laboratory of Systems Pharmacology, Harvard Medical School;
| |
Collapse
|
14
|
Liu Q, Yin X, Languino LR, Altieri DC. Evaluation of drug combination effect using a Bliss independence dose-response surface model. Stat Biopharm Res 2018; 10:112-122. [PMID: 30881603 DOI: 10.1080/19466315.2018.1437071] [Citation(s) in RCA: 106] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
To test the anticancer effect of combining two drugs targeting different biological pathways, the popular way to show synergistic effect of drug combination is a heat map or surface plot based on the percent excess the Bliss prediction using the average response measures at each combination dose. Such graphs, however, are inefficient in the drug screening process and it doesn't give a statistical inference on synergistic effect. To make a statistically rigorous and robust conclusion for drug combination effect, we present a two-stage Bliss independence response surface model to estimate an overall interaction index (τ) with 95% confidence interval (CI). By taking into all data points account, the overall τ with 95% CI can be applied to determine if the drug combination effect is synergistic overall. Using some example data, the two-stage model was compared to a couple of classic models following Bliss rule. The data analysis results obtained from our model reflect the pattern shown from other models. The application of overall τ helps investigators to make decision easier and accelerate the preclinical drug screening.
Collapse
Affiliation(s)
- Qin Liu
- Molecular and Cellular Oncogenesis Program, The Wistar Institute, 3601 Spruce Street, Philadelphia, PA 19104
| | - Xiangfan Yin
- Molecular and Cellular Oncogenesis Program, The Wistar Institute, 3601 Spruce Street, Philadelphia, PA 19104
| | - Lucia R Languino
- Department of Cancer Biology, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107
| | - Dario C Altieri
- Immunology, Microenvironment & Metastasis, The Wistar Institute, 3601 Spruce Street, Philadelphia, PA 19104
| |
Collapse
|
15
|
Pathak RK, Baunthiyal M, Pandey N, Pandey D, Kumar A. Modeling of the jasmonate signaling pathway in Arabidopsis thaliana with respect to pathophysiology of Alternaria blight in Brassica. Sci Rep 2017; 7:16790. [PMID: 29196636 PMCID: PMC5711873 DOI: 10.1038/s41598-017-16884-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Accepted: 11/08/2017] [Indexed: 01/01/2023] Open
Abstract
The productivity of Oilseed Brassica, one of the economically important crops of India, is seriously affected by the disease, Alternaria blight. The disease is mainly caused by two major necrotrophic fungi, Alternaria brassicae and Alternaria brassicicola which are responsible for significant yield losses. Till date, no resistant source is available against Alternaria blight, hence plant breeding methods can not be used to develop disease resistant varieties. Jasmonate mediated signalling pathway, which is known to play crucial role during defense response against necrotrophs, could be strengthened in Brassica plants to combat the disease. Since scanty information is available in Brassica-Alternaria pathosystems at molecular level therefore, in the present study efforts have been made to model jasmonic acid pathway in Arabidopsis thaliana to simulate the dynamic behaviour of molecular species in the model. Besides, the developed model was also analyzed topologically for investigation of the hubs node. COI1 is identified as one of the promising candidate genes in response to Alternaria and other linked components of plant defense mechanisms against the pathogens. The findings from present study are therefore informative for understanding the molecular basis of pathophysiology and rational management of Alternaria blight for securing food and nutritional security.
Collapse
Affiliation(s)
- Rajesh Kumar Pathak
- Department of Biotechnology, Govind Ballabh Pant Institute of Engineering & Technology, Pauri Garhwal, 246194, Uttarakhand, India
| | - Mamta Baunthiyal
- Department of Biotechnology, Govind Ballabh Pant Institute of Engineering & Technology, Pauri Garhwal, 246194, Uttarakhand, India.
| | - Neetesh Pandey
- Centre for Agricultural Bioinformatics, ICAR-Indian Agricultural Statistics Research Institute (IASRI), Pusa, 110012, New Delhi, India
| | - Dinesh Pandey
- Department of Molecular Biology & Genetic Engineering, College of Basic Sciences & Humanities, G. B. Pant University of Agriculture & Technology, Pantnagar, 263145, India
| | - Anil Kumar
- Department of Molecular Biology & Genetic Engineering, College of Basic Sciences & Humanities, G. B. Pant University of Agriculture & Technology, Pantnagar, 263145, India.
| |
Collapse
|
16
|
Niche harmony search algorithm for detecting complex disease associated high-order SNP combinations. Sci Rep 2017; 7:11529. [PMID: 28912584 PMCID: PMC5599559 DOI: 10.1038/s41598-017-11064-9] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Accepted: 08/17/2017] [Indexed: 02/01/2023] Open
Abstract
Genome-wide association study is especially challenging in detecting high-order disease-causing models due to model diversity, possible low or even no marginal effect of the model, and extraordinary search and computations. In this paper, we propose a niche harmony search algorithm where joint entropy is utilized as a heuristic factor to guide the search for low or no marginal effect model, and two computationally lightweight scores are selected to evaluate and adapt to diverse of disease models. In order to obtain all possible suspected pathogenic models, niche technique merges with HS, which serves as a taboo region to avoid HS trapping into local search. From the resultant set of candidate SNP-combinations, we use G-test statistic for testing true positives. Experiments were performed on twenty typical simulation datasets in which 12 models are with marginal effect and eight ones are with no marginal effect. Our results indicate that the proposed algorithm has very high detection power for searching suspected disease models in the first stage and it is superior to some typical existing approaches in both detection power and CPU runtime for all these datasets. Application to age-related macular degeneration (AMD) demonstrates our method is promising in detecting high-order disease-causing models.
Collapse
|
17
|
Grixti JM, O'Hagan S, Day PJ, Kell DB. Enhancing Drug Efficacy and Therapeutic Index through Cheminformatics-Based Selection of Small Molecule Binary Weapons That Improve Transporter-Mediated Targeting: A Cytotoxicity System Based on Gemcitabine. Front Pharmacol 2017; 8:155. [PMID: 28396636 PMCID: PMC5366350 DOI: 10.3389/fphar.2017.00155] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Accepted: 03/10/2017] [Indexed: 12/23/2022] Open
Abstract
The transport of drug molecules is mainly determined by the distribution of influx and efflux transporters for which they are substrates. To enable tissue targeting, we sought to develop the idea that we might affect the transporter-mediated disposition of small-molecule drugs via the addition of a second small molecule that of itself had no inhibitory pharmacological effect but that influenced the expression of transporters for the primary drug. We refer to this as a “binary weapon” strategy. The experimental system tested the ability of a molecule that on its own had no cytotoxic effect to increase the toxicity of the nucleoside analog gemcitabine to Panc1 pancreatic cancer cells. An initial phenotypic screen of a 500-member polar drug (fragment) library yielded three “hits.” The structures of 20 of the other 2,000 members of this library suite had a Tanimoto similarity greater than 0.7 to those of the initial hits, and each was itself a hit (the cheminformatics thus providing for a massive enrichment). We chose the top six representatives for further study. They fell into three clusters whose members bore reasonable structural similarities to each other (two were in fact isomers), lending strength to the self-consistency of both our conceptual and experimental strategies. Existing literature had suggested that indole-3-carbinol might play a similar role to that of our fragments, but in our hands it was without effect; nor was it structurally similar to any of our hits. As there was no evidence that the fragments could affect toxicity directly, we looked for effects on transporter transcript levels. In our hands, only the ENT1-3 uptake and ABCC2,3,4,5, and 10 efflux transporters displayed measurable transcripts in Panc1 cultures, along with a ribonucleoside reductase RRM1 known to affect gemcitabine toxicity. Very strikingly, the addition of gemcitabine alone increased the expression of the transcript for ABCC2 (MRP2) by more than 12-fold, and that of RRM1 by more than fourfold, and each of the fragment “hits” served to reverse this. However, an inhibitor of ABCC2 was without significant effect, implying that RRM1 was possibly the more significant player. These effects were somewhat selective for Panc cells. It seems, therefore, that while the effects we measured were here mediated more by efflux than influx transporters, and potentially by other means, the binary weapon idea is hereby fully confirmed: it is indeed possible to find molecules that manipulate the expression of transporters that are involved in the bioactivity of a pharmaceutical drug. This opens up an entirely new area, that of chemical genomics-based drug targeting.
Collapse
Affiliation(s)
- Justine M Grixti
- Faculty of Biology, Medicine and Health, University of ManchesterManchester, UK; Manchester Institute of Biotechnology, University of ManchesterManchester, UK
| | - Steve O'Hagan
- Manchester Institute of Biotechnology, University of ManchesterManchester, UK; School of Chemistry, University of ManchesterManchester, UK; Centre for Synthetic Biology of Fine and Speciality Chemicals, University of ManchesterManchester, UK
| | - Philip J Day
- Faculty of Biology, Medicine and Health, University of ManchesterManchester, UK; Manchester Institute of Biotechnology, University of ManchesterManchester, UK
| | - Douglas B Kell
- Manchester Institute of Biotechnology, University of ManchesterManchester, UK; School of Chemistry, University of ManchesterManchester, UK; Centre for Synthetic Biology of Fine and Speciality Chemicals, University of ManchesterManchester, UK
| |
Collapse
|
18
|
Detecting High-Order Epistasis in Nonlinear Genotype-Phenotype Maps. Genetics 2017; 205:1079-1088. [PMID: 28100592 PMCID: PMC5340324 DOI: 10.1534/genetics.116.195214] [Citation(s) in RCA: 89] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2016] [Accepted: 01/09/2017] [Indexed: 11/18/2022] Open
Abstract
High-order epistasis has been observed in many genotype-phenotype maps. These multi-way interactions between mutations may be useful for dissecting complex traits and could have profound implications for evolution. Alternatively, they could be a statistical artifact. High-order epistasis models assume the effects of mutations should add, when they could in fact multiply or combine in some other nonlinear way. A mismatch in the “scale” of the epistasis model and the scale of the underlying map would lead to spurious epistasis. In this article, we develop an approach to estimate the nonlinear scales of arbitrary genotype-phenotype maps. We can then linearize these maps and extract high-order epistasis. We investigated seven experimental genotype-phenotype maps for which high-order epistasis had been reported previously. We find that five of the seven maps exhibited nonlinear scales. Interestingly, even after accounting for nonlinearity, we found statistically significant high-order epistasis in all seven maps. The contributions of high-order epistasis to the total variation ranged from 2.2 to 31.0%, with an average across maps of 12.7%. Our results provide strong evidence for extensive high-order epistasis, even after nonlinear scale is taken into account. Further, we describe a simple method to estimate and account for nonlinearity in genotype-phenotype maps.
Collapse
|
19
|
Shin SY, Nguyen LK. Dissecting Cell-Fate Determination Through Integrated Mathematical Modeling of the ERK/MAPK Signaling Pathway. Methods Mol Biol 2017; 1487:409-432. [PMID: 27924583 DOI: 10.1007/978-1-4939-6424-6_29] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The past three decades have witnessed an enormous progress in the elucidation of the ERK/MAPK signaling pathway and its involvement in various cellular processes. Because of its importance and complex wiring, the ERK pathway has been an intensive subject for mathematical modeling, which facilitates the unraveling of key dynamic properties and behaviors of the pathway. Recently, however, it became evident that the pathway does not act in isolation but closely interacts with many other pathways to coordinate various cellular outcomes under different pathophysiological contexts. This has led to an increasing number of integrated, large-scale models that link the ERK pathway to other functionally important pathways. In this chapter, we first discuss the essential steps in model development and notable models of the ERK pathway. We then use three examples of integrated, multipathway models to investigate how crosstalk of ERK signaling with other pathways regulates cell-fate decision-making in various physiological and disease contexts. Specifically, we focus on ERK interactions with the phosphoinositide-3 kinase (PI3K), c-Jun N-terminal kinase (JNK), and β-adrenergic receptor (β-AR) signaling pathways. We conclude that integrated modeling in combination with wet-lab experimentation have been and will be instrumental in gaining an in-depth understanding of ERK signaling in multiple biological contexts.
Collapse
Affiliation(s)
- Sung-Young Shin
- Department of Biochemistry and Molecular Biology, School of Biomedical Sciences, Monash University, Clayton, VIC, 3800, Australia.,Biomedicine Discovery Institute, Monash University, Clayton, VIC, 3800, Australia
| | - Lan K Nguyen
- Department of Biochemistry and Molecular Biology, School of Biomedical Sciences, Monash University, Clayton, VIC, 3800, Australia. .,Biomedicine Discovery Institute, Monash University, Clayton, VIC, 3800, Australia.
| |
Collapse
|
20
|
High-Order Drug Combinations Are Required to Effectively Kill Colorectal Cancer Cells. Cancer Res 2016; 76:6950-6963. [DOI: 10.1158/0008-5472.can-15-3425] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Revised: 06/26/2016] [Accepted: 08/02/2016] [Indexed: 11/16/2022]
|
21
|
Prediction of multidimensional drug dose responses based on measurements of drug pairs. Proc Natl Acad Sci U S A 2016; 113:10442-7. [PMID: 27562164 DOI: 10.1073/pnas.1606301113] [Citation(s) in RCA: 115] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Finding potent multidrug combinations against cancer and infections is a pressing therapeutic challenge; however, screening all combinations is difficult because the number of experiments grows exponentially with the number of drugs and doses. To address this, we present a mathematical model that predicts the effects of three or more antibiotics or anticancer drugs at all doses based only on measurements of drug pairs at a few doses, without need for mechanistic information. The model provides accurate predictions on available data for antibiotic combinations, and on experiments presented here on the response matrix of three cancer drugs at eight doses per drug. This approach offers a way to search for effective multidrug combinations using a small number of experiments.
Collapse
|
22
|
Serra-Musach J, Mateo F, Capdevila-Busquets E, de Garibay GR, Zhang X, Guha R, Thomas CJ, Grueso J, Villanueva A, Jaeger S, Heyn H, Vizoso M, Pérez H, Cordero A, Gonzalez-Suarez E, Esteller M, Moreno-Bueno G, Tjärnberg A, Lázaro C, Serra V, Arribas J, Benson M, Gustafsson M, Ferrer M, Aloy P, Pujana MÀ. Cancer network activity associated with therapeutic response and synergism. Genome Med 2016; 8:88. [PMID: 27553366 PMCID: PMC4995628 DOI: 10.1186/s13073-016-0340-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2016] [Accepted: 08/01/2016] [Indexed: 12/14/2022] Open
Abstract
Background Cancer patients often show no or only modest benefit from a given therapy. This major problem in oncology is generally attributed to the lack of specific predictive biomarkers, yet a global measure of cancer cell activity may support a comprehensive mechanistic understanding of therapy efficacy. We reasoned that network analysis of omic data could help to achieve this goal. Methods A measure of “cancer network activity” (CNA) was implemented based on a previously defined network feature of communicability. The network nodes and edges corresponded to human proteins and experimentally identified interactions, respectively. The edges were weighted proportionally to the expression of the genes encoding for the corresponding proteins and relative to the number of direct interactors. The gene expression data corresponded to the basal conditions of 595 human cancer cell lines. Therapeutic responses corresponded to the impairment of cell viability measured by the half maximal inhibitory concentration (IC50) of 130 drugs approved or under clinical development. Gene ontology, signaling pathway, and transcription factor-binding annotations were taken from public repositories. Predicted synergies were assessed by determining the viability of four breast cancer cell lines and by applying two different analytical methods. Results The effects of drug classes were associated with CNAs formed by different cell lines. CNAs also differentiate target families and effector pathways. Proteins that occupy a central position in the network largely contribute to CNA. Known key cancer-associated biological processes, signaling pathways, and master regulators also contribute to CNA. Moreover, the major cancer drivers frequently mediate CNA and therapeutic differences. Cell-based assays centered on these differences and using uncorrelated drug effects reveals novel synergistic combinations for the treatment of breast cancer dependent on PI3K-mTOR signaling. Conclusions Cancer therapeutic responses can be predicted on the basis of a systems-level analysis of molecular interactions and gene expression. Fundamental cancer processes, pathways, and drivers contribute to this feature, which can also be exploited to predict precise synergistic drug combinations. Electronic supplementary material The online version of this article (doi:10.1186/s13073-016-0340-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jordi Serra-Musach
- Breast Cancer and Systems Biology Lab, Program Against Cancer Therapeutic Resistance (ProCURE), Catalan Institute of Oncology (ICO), Bellvitge Institute for Biomedical Research (IDIBELL), Gran via 199, L'Hospitalet del Llobregat, Barcelona, 08908, Catalonia, Spain
| | - Francesca Mateo
- Breast Cancer and Systems Biology Lab, Program Against Cancer Therapeutic Resistance (ProCURE), Catalan Institute of Oncology (ICO), Bellvitge Institute for Biomedical Research (IDIBELL), Gran via 199, L'Hospitalet del Llobregat, Barcelona, 08908, Catalonia, Spain
| | - Eva Capdevila-Busquets
- Joint IRB-BSC-CRG Program in Computational Biology, Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Baldiri Reixac 10, Barcelona, 08028, Catalonia, Spain
| | - Gorka Ruiz de Garibay
- Breast Cancer and Systems Biology Lab, Program Against Cancer Therapeutic Resistance (ProCURE), Catalan Institute of Oncology (ICO), Bellvitge Institute for Biomedical Research (IDIBELL), Gran via 199, L'Hospitalet del Llobregat, Barcelona, 08908, Catalonia, Spain
| | - Xiaohu Zhang
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences (NCATS), National Institutes of Health, 9800 Medical Center Dr. Rockville, Bethesda, MD, 20850, USA
| | - Raj Guha
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences (NCATS), National Institutes of Health, 9800 Medical Center Dr. Rockville, Bethesda, MD, 20850, USA
| | - Craig J Thomas
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences (NCATS), National Institutes of Health, 9800 Medical Center Dr. Rockville, Bethesda, MD, 20850, USA
| | - Judit Grueso
- Experimental Therapeutics Group, Vall d'Hebron Institute of Oncology (VHIO), Cellex Center, Natzaret 115-117, Barcelona, 08035, Catalonia, Spain
| | - Alberto Villanueva
- Breast Cancer and Systems Biology Lab, Program Against Cancer Therapeutic Resistance (ProCURE), Catalan Institute of Oncology (ICO), Bellvitge Institute for Biomedical Research (IDIBELL), Gran via 199, L'Hospitalet del Llobregat, Barcelona, 08908, Catalonia, Spain
| | - Samira Jaeger
- Joint IRB-BSC-CRG Program in Computational Biology, Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Baldiri Reixac 10, Barcelona, 08028, Catalonia, Spain
| | - Holger Heyn
- Cancer Epigenetics and Biology Program (PEBC), IDIBELL, Gran via 199, L'Hospitalet del Llobregat, Barcelona, 08908, Catalonia, Spain
| | - Miguel Vizoso
- Cancer Epigenetics and Biology Program (PEBC), IDIBELL, Gran via 199, L'Hospitalet del Llobregat, Barcelona, 08908, Catalonia, Spain
| | - Hector Pérez
- Cancer Epigenetics and Biology Program (PEBC), IDIBELL, Gran via 199, L'Hospitalet del Llobregat, Barcelona, 08908, Catalonia, Spain
| | - Alex Cordero
- Cancer Epigenetics and Biology Program (PEBC), IDIBELL, Gran via 199, L'Hospitalet del Llobregat, Barcelona, 08908, Catalonia, Spain
| | - Eva Gonzalez-Suarez
- Cancer Epigenetics and Biology Program (PEBC), IDIBELL, Gran via 199, L'Hospitalet del Llobregat, Barcelona, 08908, Catalonia, Spain
| | - Manel Esteller
- Cancer Epigenetics and Biology Program (PEBC), IDIBELL, Gran via 199, L'Hospitalet del Llobregat, Barcelona, 08908, Catalonia, Spain.,Department of Physiological Sciences II, School of Medicine, University of Barcelona, Feixa Llarga s/n, L'Hospitalet del Llobregat, Barcelona, 08908, Catalonia, Spain.,Catalan Institution for Research and Advanced Studies (ICREA), Passeig Lluís Companys 23, Barcelona, 08010, Catalonia, Spain
| | - Gema Moreno-Bueno
- Department of Biochemistry, Autonomous University of Madrid (UAM), Biomedical Research Institute "Alberto Sols" (Spanish National Research Council (CSIC)-UAM), Hospital La Paz Institute for Health Research (IdiPAZ), Arzobispo Morcillo 4, Madrid, 28029, Spain.,MD Anderson International Foundation, Arturo Soria 270, Madrid, 28033, Spain
| | - Andreas Tjärnberg
- The Centre for Individualized Medicine, Department of Clinical and Experimental Medicine, Linköping University, Linköping, 58183, Sweden
| | - Conxi Lázaro
- Hereditary Cancer Program, ICO, IDIBELL, Gran via 199, L'Hospitalet del Llobregat, Barcelona, 08908, Catalonia, Spain
| | - Violeta Serra
- Experimental Therapeutics Group, Vall d'Hebron Institute of Oncology (VHIO), Cellex Center, Natzaret 115-117, Barcelona, 08035, Catalonia, Spain
| | - Joaquín Arribas
- Catalan Institution for Research and Advanced Studies (ICREA), Passeig Lluís Companys 23, Barcelona, 08010, Catalonia, Spain.,Preclinical Research Program, VHIO, Cellex Center, Natzaret 115-117, Barcelona, 08035, Catalonia, Spain.,Department of Biochemistry and Molecular Biology, Medical School Building M, Autonomous University of Barcelona, Bellaterra, 08193, Catalonia, Spain
| | - Mikael Benson
- The Centre for Individualized Medicine, Department of Clinical and Experimental Medicine, Linköping University, Linköping, 58183, Sweden
| | - Mika Gustafsson
- The Centre for Individualized Medicine, Department of Clinical and Experimental Medicine, Linköping University, Linköping, 58183, Sweden
| | - Marc Ferrer
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences (NCATS), National Institutes of Health, 9800 Medical Center Dr. Rockville, Bethesda, MD, 20850, USA.
| | - Patrick Aloy
- Joint IRB-BSC-CRG Program in Computational Biology, Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Baldiri Reixac 10, Barcelona, 08028, Catalonia, Spain. .,Catalan Institution for Research and Advanced Studies (ICREA), Passeig Lluís Companys 23, Barcelona, 08010, Catalonia, Spain.
| | - Miquel Àngel Pujana
- Breast Cancer and Systems Biology Lab, Program Against Cancer Therapeutic Resistance (ProCURE), Catalan Institute of Oncology (ICO), Bellvitge Institute for Biomedical Research (IDIBELL), Gran via 199, L'Hospitalet del Llobregat, Barcelona, 08908, Catalonia, Spain.
| |
Collapse
|
23
|
Chen X, Ren B, Chen M, Wang Q, Zhang L, Yan G. NLLSS: Predicting Synergistic Drug Combinations Based on Semi-supervised Learning. PLoS Comput Biol 2016; 12:e1004975. [PMID: 27415801 PMCID: PMC4945015 DOI: 10.1371/journal.pcbi.1004975] [Citation(s) in RCA: 208] [Impact Index Per Article: 23.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2016] [Accepted: 05/12/2016] [Indexed: 02/05/2023] Open
Abstract
Fungal infection has become one of the leading causes of hospital-acquired infections with high mortality rates. Furthermore, drug resistance is common for fungus-causing diseases. Synergistic drug combinations could provide an effective strategy to overcome drug resistance. Meanwhile, synergistic drug combinations can increase treatment efficacy and decrease drug dosage to avoid toxicity. Therefore, computational prediction of synergistic drug combinations for fungus-causing diseases becomes attractive. In this study, we proposed similar nature of drug combinations: principal drugs which obtain synergistic effect with similar adjuvant drugs are often similar and vice versa. Furthermore, we developed a novel algorithm termed Network-based Laplacian regularized Least Square Synergistic drug combination prediction (NLLSS) to predict potential synergistic drug combinations by integrating different kinds of information such as known synergistic drug combinations, drug-target interactions, and drug chemical structures. We applied NLLSS to predict antifungal synergistic drug combinations and showed that it achieved excellent performance both in terms of cross validation and independent prediction. Finally, we performed biological experiments for fungal pathogen Candida albicans to confirm 7 out of 13 predicted antifungal synergistic drug combinations. NLLSS provides an efficient strategy to identify potential synergistic antifungal combinations. Drug combinations represent a promising strategy for overcoming fungal drug resistance and treating complex diseases. There is an urgent need to establish powerful computational methods for systematic prediction of synergistic drug combination on a large scale. Based on the assumption that principal drugs which obtain synergistic effect with similar adjuvant drugs are often similar and vice versa, NLLSS was developed to predict potential synergistic drug combinations by integrating known synergistic drug combinations, unlabeled drug combinations, drug-target interactions, and drug chemical structures. NLLSS has obtained the reliable performance in the cross validation and experimental validations, which indicated that NLLSS has an excellent performance of identifying potential synergistic drug combinations. Out of 13 predicted antifungal synergistic drug combinations, 7 candidates were experimentally confirmed. It is anticipated that NLLSS would be an important and useful resource by providing a new strategy to identify potential synergistic antifungal combinations, explore new indications of existing drugs, and provide useful insights into the underlying molecular mechanisms of synergistic drug combinations.
Collapse
Affiliation(s)
- Xing Chen
- School of Information and Electrical Engineering, China University of Mining and Technology, Xuzhou, China
| | - Biao Ren
- Chinese Academy of Sciences Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Sichuan, China
| | - Ming Chen
- Chinese Academy of Sciences Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | - Quanxin Wang
- Chinese Academy of Sciences Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Lixin Zhang
- Chinese Academy of Sciences Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
- South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou, China
- * E-mail: (LZ); (GY)
| | - Guiying Yan
- Academy of Mathematics and Systems Science, Chinese Academy of Sciences, Beijing, China
- * E-mail: (LZ); (GY)
| |
Collapse
|
24
|
Garmaroudi FS, Handy DE, Liu YY, Loscalzo J. Systems Pharmacology and Rational Polypharmacy: Nitric Oxide-Cyclic GMP Signaling Pathway as an Illustrative Example and Derivation of the General Case. PLoS Comput Biol 2016; 12:e1004822. [PMID: 26985825 PMCID: PMC4795786 DOI: 10.1371/journal.pcbi.1004822] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2015] [Accepted: 02/19/2016] [Indexed: 11/23/2022] Open
Abstract
Impaired nitric oxide (NO˙)-cyclic guanosine 3', 5'-monophosphate (cGMP) signaling has been observed in many cardiovascular disorders, including heart failure and pulmonary arterial hypertension. There are several enzymatic determinants of cGMP levels in this pathway, including soluble guanylyl cyclase (sGC) itself, the NO˙-activated form of sGC, and phosphodiesterase(s) (PDE). Therapies for some of these disorders with PDE inhibitors have been successful at increasing cGMP levels in both cardiac and vascular tissues. However, at the systems level, it is not clear whether perturbation of PDE alone, under oxidative stress, is the best approach for increasing cGMP levels as compared with perturbation of other potential pathway targets, either alone or in combination. Here, we develop a model-based approach to perturbing this pathway, focusing on single reactions, pairs of reactions, or trios of reactions as targets, then monitoring the theoretical effects of these interventions on cGMP levels. Single perturbations of all reaction steps within this pathway demonstrated that three reaction steps, including the oxidation of sGC, NO˙ dissociation from sGC, and cGMP degradation by PDE, exerted a dominant influence on cGMP accumulation relative to other reaction steps. Furthermore, among all possible single, paired, and triple perturbations of this pathway, the combined perturbations of these three reaction steps had the greatest impact on cGMP accumulation. These computational findings were confirmed in cell-based experiments. We conclude that a combined perturbation of the oxidatively-impaired NO˙-cGMP signaling pathway is a better approach to the restoration of cGMP levels as compared with corresponding individual perturbations. This approach may also yield improved therapeutic responses in other complex pharmacologically amenable pathways. Developing drugs for a well-defined biochemical or molecular pathway has conventionally been approached by optimizing the inhibition (or activation) of a single target by a single pharmacologic agent. On occasion, drug combinations have been used that generally target multiple pathways affecting a common phenotype, again by optimizing the extent of inhibition of individual targets, semi-empirically adjusting their doses to minimize toxicities as they are manifest. Here, we present a computational approach for identifying optimal combinations of agents that can affect (inhibit) a well-defined biochemical pathway, doing so at minimal combined concentrations, thereby potentially minimizing dose-dependent toxicities. This approach is illustrated computationally and experimentally with a well-known pathway, the nitric oxide-cyclic GMP pathway, but is readily generalizable to rational polypharmacy.
Collapse
Affiliation(s)
- Farshid S. Garmaroudi
- Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Diane E. Handy
- Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Yang-Yu Liu
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Joseph Loscalzo
- Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
25
|
Krueger AS, Munck C, Dantas G, Church GM, Galagan J, Lehár J, Sommer MOA. Simulating Serial-Target Antibacterial Drug Synergies Using Flux Balance Analysis. PLoS One 2016; 11:e0147651. [PMID: 26821252 PMCID: PMC4731467 DOI: 10.1371/journal.pone.0147651] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2015] [Accepted: 01/06/2016] [Indexed: 01/09/2023] Open
Abstract
Flux balance analysis (FBA) is an increasingly useful approach for modeling the behavior of metabolic systems. However, standard FBA modeling of genetic knockouts cannot predict drug combination synergies observed between serial metabolic targets, even though such synergies give rise to some of the most widely used antibiotic treatments. Here we extend FBA modeling to simulate responses to chemical inhibitors at varying concentrations, by diverting enzymatic flux to a waste reaction. This flux diversion yields very similar qualitative predictions to prior methods for single target activity. However, we find very different predictions for combinations, where flux diversion, which mimics the kinetics of competitive metabolic inhibitors, can explain serial target synergies between metabolic enzyme inhibitors that we confirmed in Escherichia coli cultures. FBA flux diversion opens the possibility for more accurate genome-scale predictions of drug synergies, which can be used to suggest treatments for infections and other diseases.
Collapse
Affiliation(s)
- Andrew S. Krueger
- Boston University, 44 Cummington St, Boston, MA, United States of America
| | - Christian Munck
- Technical University of Denmark, Novo Nordisk Foundation Center for Biosustainability, Hørsholm, Denmark
| | - Gautam Dantas
- Center for Genome Science & Systems Biology, Washington University School of Medicine, St Louis, Missouri, United States of America
- Department of Pathology & Immunology, Washington University School of Medicine, St Louis, Missouri, United States of America
- Department of Biomedical Engineering, Washington University, St. Louis, Missouri, United States of America
| | - George M. Church
- Department of Genetics, Harvard Medical School, Boston, Massachusetts, United States of America
| | - James Galagan
- Boston University, 44 Cummington St, Boston, MA, United States of America
- Broad Institute, Cambridge Center, Cambridge, Massachusetts, United States of America
| | - Joseph Lehár
- Boston University, 44 Cummington St, Boston, MA, United States of America
- * E-mail: (JL); (MOAS)
| | - Morten O. A. Sommer
- Technical University of Denmark, Novo Nordisk Foundation Center for Biosustainability, Hørsholm, Denmark
- * E-mail: (JL); (MOAS)
| |
Collapse
|
26
|
Chan GKY, Wilson S, Schmidt S, Moffat JG. Unlocking the Potential of High-Throughput Drug Combination Assays Using Acoustic Dispensing. ACTA ACUST UNITED AC 2015; 21:125-32. [PMID: 26160862 DOI: 10.1177/2211068215593759] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Indexed: 11/15/2022]
Abstract
Assessment of synergistic effects of drug combinations in vitro is a critical part of anticancer drug research. However, the complexities of dosing and analyzing two drugs over the appropriate range of doses have generally led to compromises in experimental design that restrict the quality and robustness of the data. In particular, the use of a single dose response of combined drugs, rather than a full two-way matrix of varying doses, has predominated in higher-throughput studies. Acoustic dispensing unlocks the potential of high-throughput dose matrix analysis. We have developed acoustic dispensing protocols that enable compound synergy assays in a 384-well format. This experimental design is considerably more efficient and flexible with respect to time, reagent usage, and labware than is achievable using traditional serial-dilution approaches. Data analysis tools integrated in Genedata Screener were used to efficiently deconvolute the combination compound mapping scheme and calculate compound potency and synergy metrics. We have applied this workflow to evaluate interactions among drugs targeting different nodes of the mitogen-activated protein kinase pathway in a panel of cancer cell lines.
Collapse
Affiliation(s)
- Grace Ka Yan Chan
- Department of Biochemical and Cellular Pharmacology, San Francisco, CA, USA
| | - Stacy Wilson
- Department of Immunology, Tissue Growth and Repair Biomarker Development, Genentech, South San Francisco, CA, USA
| | - Stephen Schmidt
- Department of Biochemical and Cellular Pharmacology, San Francisco, CA, USA
| | - John G Moffat
- Department of Biochemical and Cellular Pharmacology, San Francisco, CA, USA
| |
Collapse
|
27
|
Ding X, Wang J, Zelikovsky A, Guo X, Xie M, Pan Y. Searching High-Order SNP Combinations for Complex Diseases Based on Energy Distribution Difference. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2015; 12:695-704. [PMID: 26357280 DOI: 10.1109/tcbb.2014.2363459] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Single nucleotide polymorphisms, a dominant type of genetic variants, have been used successfully to identify defective genes causing human single gene diseases. However, most common human diseases are complex diseases and caused by gene-gene and gene-environment interactions. Many SNP-SNP interaction analysis methods have been introduced but they are not powerful enough to discover interactions more than three SNPs. The paper proposes a novel method that analyzes all SNPs simultaneously. Different from existing methods, the method regards an individual's genotype data on a list of SNPs as a point with a unit of energy in a multi-dimensional space, and tries to find a new coordinate system where the energy distribution difference between cases and controls reaches the maximum. The method will find different multiple SNPs combinatorial patterns between cases and controls based on the new coordinate system. The experiment on simulated data shows that the method is efficient. The tests on the real data of age-related macular degeneration (AMD) disease show that it can find out more significant multi-SNP combinatorial patterns than existing methods.
Collapse
|
28
|
Tang J, Aittokallio T. Network pharmacology strategies toward multi-target anticancer therapies: from computational models to experimental design principles. Curr Pharm Des 2014; 20:23-36. [PMID: 23530504 PMCID: PMC3894695 DOI: 10.2174/13816128113199990470] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2013] [Accepted: 03/18/2013] [Indexed: 12/12/2022]
Abstract
Polypharmacology has emerged as novel means in drug discovery for improving treatment response in clinical use. However, to really capitalize on the polypharmacological effects of drugs, there is a critical need to better model and understand how the complex interactions between drugs and their cellular targets contribute to drug efficacy and possible side effects. Network graphs provide a convenient modeling framework for dealing with the fact that most drugs act on cellular systems through targeting multiple proteins both through on-target and off-target binding. Network pharmacology models aim at addressing questions such as how and where in the disease network should one target to inhibit disease phenotypes, such as cancer growth, ideally leading to therapies that are less vulnerable to drug resistance and side effects by means of attacking the disease network at the systems level through synergistic and synthetic lethal interactions. Since the exponentially increasing number of potential drug target combinations makes pure experimental approach quickly unfeasible, this review depicts a number of computational models and algorithms that can effectively reduce the search space for determining the most promising combinations for experimental evaluation. Such computational-experimental strategies are geared toward realizing the full potential of multi-target treatments in different disease phenotypes. Our specific focus is on system-level network approaches to polypharmacology designs in anticancer drug discovery, where we give representative examples of how network-centric modeling may offer systematic strategies toward better understanding and even predicting the phenotypic responses to multi-target therapies.
Collapse
|
29
|
Liggi S, Drakakis G, Hendry AE, Hanson KM, Brewerton SC, Wheeler GN, Bodkin MJ, Evans DA, Bender A. Extensions to In Silico Bioactivity Predictions Using Pathway Annotations and Differential Pharmacology Analysis: Application toXenopus laevisPhenotypic Readouts. Mol Inform 2013; 32:1009-24. [DOI: 10.1002/minf.201300102] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2013] [Accepted: 08/06/2013] [Indexed: 12/20/2022]
|
30
|
Sun X, Vilar S, Tatonetti NP. High-Throughput Methods for Combinatorial Drug Discovery. Sci Transl Med 2013; 5:205rv1. [DOI: 10.1126/scitranslmed.3006667] [Citation(s) in RCA: 115] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
|
31
|
Gerlee P, Schmidt L, Monsefi N, Kling T, Jörnsten R, Nelander S. Searching for synergies: matrix algebraic approaches for efficient pair screening. PLoS One 2013; 8:e68598. [PMID: 23935877 PMCID: PMC3723843 DOI: 10.1371/journal.pone.0068598] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2012] [Accepted: 05/31/2013] [Indexed: 11/21/2022] Open
Abstract
Functionally interacting perturbations, such as synergistic drugs pairs or synthetic lethal gene pairs, are of key interest in both pharmacology and functional genomics. However, to find such pairs by traditional screening methods is both time consuming and costly. We present a novel computational-experimental framework for efficient identification of synergistic target pairs, applicable for screening of systems with sizes on the order of current drug, small RNA or SGA (Synthetic Genetic Array) libraries (>1000 targets). This framework exploits the fact that the response of a drug pair in a given system, or a pair of genes' propensity to interact functionally, can be partly predicted by computational means from (i) a small set of experimentally determined target pairs, and (ii) pre-existing data (e.g. gene ontology, PPI) on the similarities between targets. Predictions are obtained by a novel matrix algebraic technique, based on cyclical projections onto convex sets. We demonstrate the efficiency of the proposed method using drug-drug interaction data from seven cancer cell lines and gene-gene interaction data from yeast SGA screens. Our protocol increases the rate of synergism discovery significantly over traditional screening, by up to 7-fold. Our method is easy to implement and could be applied to accelerate pair screening for both animal and microbial systems.
Collapse
Affiliation(s)
- Philip Gerlee
- Sahlgrenska Cancer Center, University of Gothenburg, Gothenburg, Sweden
- Mathematical Sciences, University of Gothenburg and Chalmers University of Technology, Gothenburg, Sweden
| | - Linnéa Schmidt
- Sahlgrenska Cancer Center, University of Gothenburg, Gothenburg, Sweden
- Department of Immunology, Genetics and Pathology and Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Naser Monsefi
- Sahlgrenska Cancer Center, University of Gothenburg, Gothenburg, Sweden
| | - Teresia Kling
- Sahlgrenska Cancer Center, University of Gothenburg, Gothenburg, Sweden
- Department of Immunology, Genetics and Pathology and Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Rebecka Jörnsten
- Mathematical Sciences, University of Gothenburg and Chalmers University of Technology, Gothenburg, Sweden
| | - Sven Nelander
- Sahlgrenska Cancer Center, University of Gothenburg, Gothenburg, Sweden
- Department of Immunology, Genetics and Pathology and Science for Life Laboratory, Uppsala University, Uppsala, Sweden
- * E-mail:
| |
Collapse
|
32
|
Oguz C, Laomettachit T, Chen KC, Watson LT, Baumann WT, Tyson JJ. Optimization and model reduction in the high dimensional parameter space of a budding yeast cell cycle model. BMC SYSTEMS BIOLOGY 2013; 7:53. [PMID: 23809412 PMCID: PMC3702416 DOI: 10.1186/1752-0509-7-53] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/07/2012] [Accepted: 06/19/2013] [Indexed: 01/16/2023]
Abstract
Background Parameter estimation from experimental data is critical for mathematical modeling of protein regulatory networks. For realistic networks with dozens of species and reactions, parameter estimation is an especially challenging task. In this study, we present an approach for parameter estimation that is effective in fitting a model of the budding yeast cell cycle (comprising 26 nonlinear ordinary differential equations containing 126 rate constants) to the experimentally observed phenotypes (viable or inviable) of 119 genetic strains carrying mutations of cell cycle genes. Results Starting from an initial guess of the parameter values, which correctly captures the phenotypes of only 72 genetic strains, our parameter estimation algorithm quickly improves the success rate of the model to 105–111 of the 119 strains. This success rate is comparable to the best values achieved by a skilled modeler manually choosing parameters over many weeks. The algorithm combines two search and optimization strategies. First, we use Latin hypercube sampling to explore a region surrounding the initial guess. From these samples, we choose ∼20 different sets of parameter values that correctly capture wild type viability. These sets form the starting generation of differential evolution that selects new parameter values that perform better in terms of their success rate in capturing phenotypes. In addition to producing highly successful combinations of parameter values, we analyze the results to determine the parameters that are most critical for matching experimental outcomes and the most competitive strains whose correct outcome with a given parameter vector forces numerous other strains to have incorrect outcomes. These “most critical parameters” and “most competitive strains” provide biological insights into the model. Conversely, the “least critical parameters” and “least competitive strains” suggest ways to reduce the computational complexity of the optimization. Conclusions Our approach proves to be a useful tool to help systems biologists fit complex dynamical models to large experimental datasets. In the process of fitting the model to the data, the tool identifies suggestive correlations among aspects of the model and the data.
Collapse
Affiliation(s)
- Cihan Oguz
- Department of Biological Sciences, Virginia Tech, Blacksburg, Virginia 24061, USA
| | | | | | | | | | | |
Collapse
|
33
|
Wang Z, Wang Y. Navigating personalized medicine dependent on modular flexibility. Trends Mol Med 2013; 19:393-5. [PMID: 23711739 DOI: 10.1016/j.molmed.2013.05.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2013] [Accepted: 05/13/2013] [Indexed: 01/07/2023]
Abstract
Deconstructing networks and rewiring alterable modules in a rational way is critical to optimize drug discovery and develop personalized medicine.
Collapse
Affiliation(s)
- Zhong Wang
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, 18 Baixincang, Dongzhimennei, Beijing 100700, China.
| | | |
Collapse
|
34
|
Kell DB. Finding novel pharmaceuticals in the systems biology era using multiple effective drug targets, phenotypic screening and knowledge of transporters: where drug discovery went wrong and how to fix it. FEBS J 2013; 280:5957-80. [PMID: 23552054 DOI: 10.1111/febs.12268] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2013] [Revised: 03/20/2013] [Accepted: 03/26/2013] [Indexed: 12/16/2022]
Abstract
Despite the sequencing of the human genome, the rate of innovative and successful drug discovery in the pharmaceutical industry has continued to decrease. Leaving aside regulatory matters, the fundamental and interlinked intellectual issues proposed to be largely responsible for this are: (a) the move from 'function-first' to 'target-first' methods of screening and drug discovery; (b) the belief that successful drugs should and do interact solely with single, individual targets, despite natural evolution's selection for biochemical networks that are robust to individual parameter changes; (c) an over-reliance on the rule-of-5 to constrain biophysical and chemical properties of drug libraries; (d) the general abandoning of natural products that do not obey the rule-of-5; (e) an incorrect belief that drugs diffuse passively into (and presumably out of) cells across the bilayers portions of membranes, according to their lipophilicity; (f) a widespread failure to recognize the overwhelmingly important role of proteinaceous transporters, as well as their expression profiles, in determining drug distribution in and between different tissues and individual patients; and (g) the general failure to use engineering principles to model biology in parallel with performing 'wet' experiments, such that 'what if?' experiments can be performed in silico to assess the likely success of any strategy. These facts/ideas are illustrated with a reasonably extensive literature review. Success in turning round drug discovery consequently requires: (a) decent systems biology models of human biochemical networks; (b) the use of these (iteratively with experiments) to model how drugs need to interact with multiple targets to have substantive effects on the phenotype; (c) the adoption of polypharmacology and/or cocktails of drugs as a desirable goal in itself; (d) the incorporation of drug transporters into systems biology models, en route to full and multiscale systems biology models that incorporate drug absorption, distribution, metabolism and excretion; (e) a return to 'function-first' or phenotypic screening; and (f) novel methods for inferring modes of action by measuring the properties on system variables at all levels of the 'omes. Such a strategy offers the opportunity of achieving a state where we can hope to predict biological processes and the effect of pharmaceutical agents upon them. Consequently, this should both lower attrition rates and raise the rates of discovery of effective drugs substantially.
Collapse
Affiliation(s)
- Douglas B Kell
- School of Chemistry, The University of Manchester, UK; Manchester Institute of Biotechnology, The University of Manchester, UK
| |
Collapse
|
35
|
An Integrative Platform of TCM Network Pharmacology and Its Application on a Herbal Formula, Qing-Luo-Yin. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2013; 2013:456747. [PMID: 23653662 PMCID: PMC3638581 DOI: 10.1155/2013/456747] [Citation(s) in RCA: 93] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/06/2013] [Accepted: 02/04/2013] [Indexed: 12/20/2022]
Abstract
The scientific understanding of traditional Chinese medicine (TCM) has been hindered by the lack of methods that can explore the complex nature and combinatorial rules of herbal formulae. On the assumption that herbal ingredients mainly target a molecular network to adjust the imbalance of human body, here we present a-self-developed TCM network pharmacology platform for discovering herbal formulae in a systematic manner. This platform integrates a set of network-based methods that we established previously to catch the network regulation mechanism and to identify active ingredients as well as synergistic combinations for a given herbal formula. We then provided a case study on an antirheumatoid arthritis (RA) formula, Qing-Luo-Yin (QLY), to demonstrate the usability of the platform. We revealed the target network of QLY against RA-related key processes including angiogenesis, inflammatory response, and immune response, based on which we not only predicted active and synergistic ingredients from QLY but also interpreted the combinatorial rule of this formula. These findings are either verified by the literature evidence or have the potential to guide further experiments. Therefore, such a network pharmacology strategy and platform is expected to make the systematical study of herbal formulae achievable and to make the TCM drug discovery predictable.
Collapse
|
36
|
Gupta MK, Misra K. Modeling and simulation analysis of propyl-thiouracil (PTU), an anti-thyroid drug on thyroid peroxidase (TPO), thyroid stimulating hormone receptor (TSHR), and sodium iodide (NIS) symporter based on systems biology approach. ACTA ACUST UNITED AC 2013. [DOI: 10.1007/s13721-013-0023-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
37
|
Delporte F, Jacquemin JM, Masson P, Watillon B. Insights into the regenerative property of plant cells and their receptivity to transgenesis: wheat as a research case study. PLANT SIGNALING & BEHAVIOR 2012; 7:1608-20. [PMID: 23072995 PMCID: PMC3578902 DOI: 10.4161/psb.22424] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
From a holistic perspective, the discovery of cellular plasticity, a very interesting property of totipotency, underlies many topical issues in biology with important medical applications, while transgenesis is a core research tool in biology. Partially known, some basic mechanisms involved in the regenerative property of cells and in their receptivity to transgenesis are common to plant and animal cells and highlight the principle of the unity of life. Transgenesis provides an important investigative instrument in plant physiology and is regarded as a valuable tool for crop improvement. The economic, social, cultural and scientific importance of cereals has led to a rich stream of research into their genetics, biology and evolution. Sustained efforts to achieve the results obtained in the fields of genetic engineering and applied biotechnology reflect this deep interest. Difficulties encountered in creating genetically modified cereals, especially wheat, highlighted the central notions of tissue culture regeneration and transformation competencies. From the perspective of combining or encountering these competencies in the same cell lineage, this reputedly recalcitrant species provides a stimulating biological system in which to explore the physiological and genetic complexity of both competencies. The former involves two phases, dedifferentiation and redifferentiation. Cells undergo development switches regulated by extrinsic and intrinsic factors. The re-entry into the cell division cycle progressively culminates in the development of organized structures. This is achieved by global chromatin reorganization associated with the reprogramming of the gene expression pattern. The latter is linked with surveillance mechanisms and DNA repair, aimed at maintaining genome integrity before cells move into mitosis, and with those mechanisms aimed at genome expression control and regulation. In order to clarify the biological basis of these two physiological properties and their interconnectedness, we look at both competencies at the core of defense/adaptive mechanisms and survival, between undifferentiated cell proliferation and organization, constituting a transition phase between two different dynamic regimes, a typical feature of critical dynamic systems. Opting for a candidate-gene strategy, several gene families could be proposed as relevant targets for investigating this hypothesis at the molecular level.
Collapse
Affiliation(s)
- Fabienne Delporte
- Walloon Agricultural Research Centre (CRAw), Department of Life Sciences, Bioengineering Unit, Gembloux, Belgium.
| | | | | | | |
Collapse
|
38
|
Abstract
Comparatively few studies have addressed directly the question of quantifying the benefits to be had from using molecular genetic markers in experimental breeding programmes (e.g. for improved crops and livestock), nor the question of which organisms should be mated with each other to best effect. We argue that this requires in silico modelling, an approach for which there is a large literature in the field of evolutionary computation (EC), but which has not really been applied in this way to experimental breeding programmes. EC seeks to optimise measurable outcomes (phenotypic fitnesses) by optimising in silico the mutation, recombination and selection regimes that are used. We review some of the approaches from EC, and compare experimentally, using a biologically relevant in silico landscape, some algorithms that have knowledge of where they are in the (genotypic) search space (G-algorithms) with some (albeit well-tuned ones) that do not (F-algorithms). For the present kinds of landscapes, F- and G-algorithms were broadly comparable in quality and effectiveness, although we recognise that the G-algorithms were not equipped with any ‘prior knowledge’ of epistatic pathway interactions. This use of algorithms based on machine learning has important implications for the optimisation of experimental breeding programmes in the post-genomic era when we shall potentially have access to the full genome sequence of every organism in a breeding population. The non-proprietary code that we have used is made freely available (via Supplementary information).
Collapse
|
39
|
Sun Z, Sun Y, Zhou Y, Wan Y. Yeast Genomics Technique for High-Throughput Drug Target Discovery. Drug Dev Res 2012. [DOI: 10.1002/ddr.21030] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Zijun Sun
- The Key Laboratory of Developmental Genes and Human Disease; Ministry of Education; Institute of Life Sciences; Southeast University; Nanjing; 210096; China
| | - Yanyan Sun
- The Key Laboratory of Developmental Genes and Human Disease; Ministry of Education; Institute of Life Sciences; Southeast University; Nanjing; 210096; China
| | - Yaxian Zhou
- The Key Laboratory of Developmental Genes and Human Disease; Ministry of Education; Institute of Life Sciences; Southeast University; Nanjing; 210096; China
| | - Yakun Wan
- The Key Laboratory of Developmental Genes and Human Disease; Ministry of Education; Institute of Life Sciences; Southeast University; Nanjing; 210096; China
| |
Collapse
|
40
|
Abstract
Modular protein interaction domains (PIDs) that recognize linear peptide motifs are found in hundreds of proteins within the human genome. Some PIDs such as SH2, 14-3-3, Chromo, and Bromo domains serve to recognize posttranslational modification (PTM) of amino acids (such as phosphorylation, acetylation, methylation, etc.) and translate these into discrete cellular responses. Other modules such as SH3 and PSD-95/Discs-large/ZO-1 (PDZ) domains recognize linear peptide epitopes and serve to organize protein complexes based on localization and regions of elevated concentration. In both cases, the ability to nucleate-specific signaling complexes is in large part dependent on the selectivity of a given protein module for its cognate peptide ligand. High-throughput (HTP) analysis of peptide-binding domains by peptide or protein arrays, phage display, mass spectrometry, or other HTP techniques provides new insight into the potential protein-protein interactions prescribed by individual or even whole families of modules. Systems level analyses have also promoted a deeper understanding of the underlying principles that govern selective protein-protein interactions and how selectivity evolves. Lastly, there is a growing appreciation for the limitations and potential pitfalls associated with HTP analysis of protein-peptide interactomes. This review will examine some of the common approaches utilized for large-scale studies of PIDs and suggest a set of standards for the analysis and validation of datasets from large-scale studies of peptide-binding modules. We will also highlight how data from large-scale studies of modular interaction domain families can provide insight into systems level properties such as the linguistics of selective interactions.
Collapse
Affiliation(s)
- Bernard A Liu
- Samuel Lunenfeld Research Institute, Mount Sinai Hospital, Toronto, ON, Canada
| | | | | |
Collapse
|
41
|
Facchetti G, Zampieri M, Altafini C. Predicting and characterizing selective multiple drug treatments for metabolic diseases and cancer. BMC SYSTEMS BIOLOGY 2012; 6:115. [PMID: 22932283 PMCID: PMC3744170 DOI: 10.1186/1752-0509-6-115] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/18/2012] [Accepted: 08/13/2012] [Indexed: 12/13/2022]
Abstract
BACKGROUND In the field of drug discovery, assessing the potential of multidrug therapies is a difficult task because of the combinatorial complexity (both theoretical and experimental) and because of the requirements on the selectivity of the therapy. To cope with this problem, we have developed a novel method for the systematic in silico investigation of synergistic effects of currently available drugs on genome-scale metabolic networks. RESULTS The algorithm finds the optimal combination of drugs which guarantees the inhibition of an objective function, while minimizing the side effect on the other cellular processes. Two different applications are considered: finding drug synergisms for human metabolic diseases (like diabetes, obesity and hypertension) and finding antitumoral drug combinations with minimal side effect on the normal human cell. The results we obtain are consistent with some of the available therapeutic indications and predict new multiple drug treatments. A cluster analysis on all possible interactions among the currently available drugs indicates a limited variety on the metabolic targets for the approved drugs. CONCLUSION The in silico prediction of drug synergisms can represent an important tool for the repurposing of drugs in a realistic perspective which considers also the selectivity of the therapy. Moreover, for a more profitable exploitation of drug-drug interactions, we have shown that also experimental drugs which have a different mechanism of action can be reconsider as potential ingredients of new multicompound therapeutic indications. Needless to say the clues provided by a computational study like ours need in any case to be thoroughly evaluated experimentally.
Collapse
Affiliation(s)
- Giuseppe Facchetti
- Statistical and Biological Physics Department, SISSA (International School for Advanced Studies), Via Bonomea 265 - 34136, Trieste, Italy
| | - Mattia Zampieri
- Institute of Molecular Systems Biology, ETH (Eidgenoessische Technische Hochschule), Wolfgang Pauli Str. 16 - 8093, Zurich, Switzerland
| | - Claudio Altafini
- Functional Analysis DepartmentSISSA (International School for Advanced Studies), , Via Bonomea 265 - 34136, Trieste, Italy
| |
Collapse
|
42
|
Wang Z, Liu J, Yu Y, Chen Y, Wang Y. Modular pharmacology: the next paradigm in drug discovery. Expert Opin Drug Discov 2012; 7:667-77. [DOI: 10.1517/17460441.2012.692673] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
43
|
Jacobs C, Segrè D. Organization Principles in Genetic Interaction Networks. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2012; 751:53-78. [DOI: 10.1007/978-1-4614-3567-9_3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
44
|
Won JK, Yang HW, Shin SY, Lee JH, Heo WD, Cho KH. The crossregulation between ERK and PI3K signaling pathways determines the tumoricidal efficacy of MEK inhibitor. J Mol Cell Biol 2012; 4:153-63. [PMID: 22561840 DOI: 10.1093/jmcb/mjs021] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
MEK inhibitor has been highlighted as a promising anti-tumor drug but its effect has been reported as varying over a wide range depending on patho-physiological conditions. In this study, we employed a systems approach by combining biochemical experimentation with in silico simulations to investigate the resistance mechanism and functional consequences of MEK inhibitor. To this end, we have developed an extended integrative model of ERK and PI3K signaling pathways by considering the crosstalk between Ras and PI3K, and analyzed the resistance mechanism to the MEK inhibitor under various mutational conditions. We found that the phospho-Akt level under the Raf mutation was remarkably augmented by MEK inhibitor, while the phospho-ERK level was almost completely repressed. These results suggest that bypassing of the ERK signal to the PI3K signal causes the resistance to the MEK inhibitor in a complex oncogenic signaling network. We further investigated the underlying mechanism of the drug resistance and revealed that the MEK inhibitor disrupts the negative feedback loops from ERK to SOS and GAB1, but activates the positive feedback loop composed of GAB1, Ras, and PI3K, which induces the bypass of the ERK signal to the PI3K signal. Based on these core feedback circuits, we suggested promising candidates for combination therapy and examined the improved inhibitory effects.
Collapse
Affiliation(s)
- Jae-Kyung Won
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon 305-701, Korea
| | | | | | | | | | | |
Collapse
|
45
|
High-order SNP combinations associated with complex diseases: efficient discovery, statistical power and functional interactions. PLoS One 2012; 7:e33531. [PMID: 22536319 PMCID: PMC3334940 DOI: 10.1371/journal.pone.0033531] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2011] [Accepted: 02/10/2012] [Indexed: 11/19/2022] Open
Abstract
There has been increased interest in discovering combinations of single-nucleotide polymorphisms (SNPs) that are strongly associated with a phenotype even if each SNP has little individual effect. Efficient approaches have been proposed for searching two-locus combinations from genome-wide datasets. However, for high-order combinations, existing methods either adopt a brute-force search which only handles a small number of SNPs (up to few hundreds), or use heuristic search that may miss informative combinations. In addition, existing approaches lack statistical power because of the use of statistics with high degrees-of-freedom and the huge number of hypotheses tested during combinatorial search. Due to these challenges, functional interactions in high-order combinations have not been systematically explored. We leverage discriminative-pattern-mining algorithms from the data-mining community to search for high-order combinations in case-control datasets. The substantially improved efficiency and scalability demonstrated on synthetic and real datasets with several thousands of SNPs allows the study of several important mathematical and statistical properties of SNP combinations with order as high as eleven. We further explore functional interactions in high-order combinations and reveal a general connection between the increase in discriminative power of a combination over its subsets and the functional coherence among the genes comprising the combination, supported by multiple datasets. Finally, we study several significant high-order combinations discovered from a lung-cancer dataset and a kidney-transplant-rejection dataset in detail to provide novel insights on the complex diseases. Interestingly, many of these associations involve combinations of common variations that occur in small fractions of population. Thus, our approach is an alternative methodology for exploring the genetics of rare diseases for which the current focus is on individually rare variations.
Collapse
|
46
|
Rodríguez-Enríquez S, Pacheco-Velázquez SC, Gallardo-Pérez JC, Marín-Hernández A, Aguilar-Ponce JL, Ruiz-García E, Ruizgodoy-Rivera LM, Meneses-García A, Moreno-Sánchez R. Multi-biomarker pattern for tumor identification and prognosis. J Cell Biochem 2012; 112:2703-15. [PMID: 21678471 DOI: 10.1002/jcb.23224] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
In last decades, the basic, clinical, and translational research efforts have been directed to the identification of standard biomarkers associated with the degree of malignancy. There is an increasingly public health concern for earlier detection of cancer development at stages in which successful treatments can be achieved. To meet this urgent clinical demand, early stage cancer biomarkers supported by reliable and robust experimental data that can be readily applicable in the clinical practice, are required. In the current standard protocols, when one or two of the canonical proliferating index biomarkers are analyzed, contradictory results are frequently reached leading to incorrect cancer diagnostic and unsuccessful therapies. Therefore, the identification of other cellular characteristics or signatures present in the tumor cells either alone or in combination with the well-established proliferation markers emerge as an alternative strategy in the improvement of cancer diagnosis and treatment. Because it is well known that several pathways and processes are altered in tumor cells, the concept of "single marker" in cancer results incorrect. Therefore, this review aims to analyze and discuss the proposal that the molecular profile of different genes or proteins in different altered tumor pathways must be established to provide a better global clinical pattern for cancer detection and prognosis.
Collapse
|
47
|
Rickles RJ, Tam WF, Giordano TP, Pierce LT, Farwell M, McMillin DW, Necheva A, Crowe D, Chen M, Avery W, Kansra V, Nawrocki ST, Carew JS, Giles FJ, Mitsiades CS, Borisy AA, Anderson KC, Lee MS. Adenosine A2A and Beta-2 Adrenergic Receptor Agonists: Novel Selective and Synergistic Multiple Myeloma Targets Discovered through Systematic Combination Screening. Mol Cancer Ther 2012; 11:1432-42. [DOI: 10.1158/1535-7163.mct-11-0925] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
48
|
Gostner JM, Wrulich OA, Jenny M, Fuchs D, Ueberall F. An update on the strategies in multicomponent activity monitoring within the phytopharmaceutical field. Altern Ther Health Med 2012; 12:18. [PMID: 22417247 PMCID: PMC3359261 DOI: 10.1186/1472-6882-12-18] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2011] [Accepted: 03/14/2012] [Indexed: 03/18/2023]
Abstract
Background To-date modern drug research has focused on the discovery and synthesis of single active substances. However, multicomponent preparations are gaining increasing importance in the phytopharmaceutical field by demonstrating beneficial properties with respect to efficacy and toxicity. Discussion In contrast to single drug combinations, a botanical multicomponent therapeutic possesses a complex repertoire of chemicals that belong to a variety of substance classes. This may explain the frequently observed pleiotropic bioactivity spectra of these compounds, which may also suggest that they possess novel therapeutic opportunities. Interestingly, considerable bioactivity properties are exhibited not only by remedies that contain high doses of phytochemicals with prominent pharmaceutical efficacy, but also preparations that lack a sole active principle component. Despite that each individual substance within these multicomponents has a low molar fraction, the therapeutic activity of these substances is established via a potentialization of their effects through combined and simultaneous attacks on multiple molecular targets. Although beneficial properties may emerge from such a broad range of perturbations on cellular machinery, validation and/or prediction of their activity profiles is accompanied with a variety of difficulties in generic risk-benefit assessments. Thus, it is recommended that a comprehensive strategy is implemented to cover the entirety of multicomponent-multitarget effects, so as to address the limitations of conventional approaches. Summary An integration of standard toxicological methods with selected pathway-focused bioassays and unbiased data acquisition strategies (such as gene expression analysis) would be advantageous in building an interaction network model to consider all of the effects, whether they were intended or adverse reactions.
Collapse
|
49
|
Andrusiak K, Piotrowski JS, Boone C. Chemical-genomic profiling: systematic analysis of the cellular targets of bioactive molecules. Bioorg Med Chem 2011; 20:1952-60. [PMID: 22261022 DOI: 10.1016/j.bmc.2011.12.023] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2011] [Revised: 12/05/2011] [Accepted: 12/13/2011] [Indexed: 11/17/2022]
Abstract
Chemical-genomic (CG) profiling of bioactive compounds is a powerful approach for drug target identification and mode of action studies. Within the last decade, research focused largely on the development and application of CG approaches in the model yeast Saccharomyces cerevisiae. The success of these methods has sparked interest in transitioning CG profiling to other biological systems to extend clinical and evolutionary relevance. Additionally, CG profiling has proven to enhance drug-synergy screens for developing combinatorial therapies. Herein, we briefly review CG profiling, focusing on emerging cross-species technologies and novel drug-synergy applications, as well as outlining needs within the field.
Collapse
Affiliation(s)
- Kerry Andrusiak
- Banting and Best Department of Medical Research and Department of Molecular Genetics, Donnelly Centre, University of Toronto, 160 College St., Toronto, ON, Canada M5S 3E1
| | | | | |
Collapse
|
50
|
Severyn B, Liehr RA, Wolicki A, Nguyen KH, Hudak EM, Ferrer M, Caldwell JS, Hermes JD, Li J, Tudor M. Parsimonious discovery of synergistic drug combinations. ACS Chem Biol 2011; 6:1391-8. [PMID: 21974780 DOI: 10.1021/cb2003225] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Combination therapies that enhance efficacy or permit reduced dosages to be administered have seen great success in a variety of therapeutic applications. More fundamentally, the discovery of epistatic pathway interactions not only informs pharmacologic intervention but can be used to better understand the underlying biological system. There is, however, no systematic and efficient method to identify interacting activities as candidates for combination therapy and, in particular, to identify those with synergistic activities. We devised a pooled, self-deconvoluting screening paradigm for the efficient comprehensive interrogation of all pairs of compounds in 1000-compound libraries. We demonstrate the power of the method to recover established synergistic interactions between compounds. We then applied this approach to a cell-based screen for anti-inflammatory activities using an assay for lipopolysaccharide/interferon-induced acute phase response of a monocytic cell line. The described method, which is >20 times as efficient as a naïve approach, was used to test all pairs of 1027 bioactive compounds for interleukin-6 suppression, yielding 11 pairs of compounds that show synergy. These 11 pairs all represent the same two activities: β-adrenergic receptor agonists and phosphodiesterase-4 inhibitors. These activities both act through cyclic AMP elevation and are known to be anti-inflammatory alone and to synergize in combination. Thus we show proof of concept for a robust, efficient technique for the identification of synergistic combinations. Such a tool can enable qualitatively new scales of pharmacological research and chemical genetics.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Jeremy S. Caldwell
- Merck Research Laboratories, West Point, Pennsylvania 19486, United States
| | | | | | | |
Collapse
|