1
|
Stephenson EH, Higgins JMG. Pharmacological approaches to understanding protein kinase signaling networks. Front Pharmacol 2023; 14:1310135. [PMID: 38164473 PMCID: PMC10757940 DOI: 10.3389/fphar.2023.1310135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Accepted: 11/27/2023] [Indexed: 01/03/2024] Open
Abstract
Protein kinases play vital roles in controlling cell behavior, and an array of kinase inhibitors are used successfully for treatment of disease. Typical drug development pipelines involve biological studies to validate a protein kinase target, followed by the identification of small molecules that effectively inhibit this target in cells, animal models, and patients. However, it is clear that protein kinases operate within complex signaling networks. These networks increase the resilience of signaling pathways, which can render cells relatively insensitive to inhibition of a single kinase, and provide the potential for pathway rewiring, which can result in resistance to therapy. It is therefore vital to understand the properties of kinase signaling networks in health and disease so that we can design effective multi-targeted drugs or combinations of drugs. Here, we outline how pharmacological and chemo-genetic approaches can contribute to such knowledge, despite the known low selectivity of many kinase inhibitors. We discuss how detailed profiling of target engagement by kinase inhibitors can underpin these studies; how chemical probes can be used to uncover kinase-substrate relationships, and how these tools can be used to gain insight into the configuration and function of kinase signaling networks.
Collapse
Affiliation(s)
| | - Jonathan M. G. Higgins
- Faculty of Medical Sciences, Biosciences Institute, Newcastle University, Newcastle uponTyne, United Kingdom
| |
Collapse
|
2
|
Berlow NE. Probabilistic Boolean Modeling of Pre-clinical Tumor Models for Biomarker Identification in Cancer Drug Development. Curr Protoc 2021; 1:e269. [PMID: 34661991 DOI: 10.1002/cpz1.269] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
As high-throughput sequencing experiments become more widely used in pre-clinical and clinical settings, pharmacogenetic and pharmacogenomic biomarker development plays an increasingly important role in oncology drug development pipelines and programs. Consequently, computer-based learning approaches have entered into use at multiple stages in pre-clinical and clinical pipelines. However, few approaches are available to identify interpretable and implementable biomarkers of response early in the drug development process when only small pre-clinical data packages are available. To address the need for early-stage biomarker development using pre-clinical tumor models, we have adapted the previously published Probabilistic Target Inhibitor Map (PTIM) platform to the challenge of biomarker hypothesis development, and denoted this approach the Probabilistic Target Map-Biomarker (PTM-Biomarker). In this article, we detail the history and design philosophy of PTM-Biomarker, and present two case studies using the biomarker discovery tool to illustrate its utility in guiding cancer drug development. © 2021 Wiley Periodicals LLC.
Collapse
|
3
|
He B, Hou F, Ren C, Bing P, Xiao X. A Review of Current In Silico Methods for Repositioning Drugs and Chemical Compounds. Front Oncol 2021; 11:711225. [PMID: 34367996 PMCID: PMC8340770 DOI: 10.3389/fonc.2021.711225] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 07/07/2021] [Indexed: 12/23/2022] Open
Abstract
Drug repositioning is a new way of applying the existing therapeutics to new disease indications. Due to the exorbitant cost and high failure rate in developing new drugs, the continued use of existing drugs for treatment, especially anti-tumor drugs, has become a widespread practice. With the assistance of high-throughput sequencing techniques, many efficient methods have been proposed and applied in drug repositioning and individualized tumor treatment. Current computational methods for repositioning drugs and chemical compounds can be divided into four categories: (i) feature-based methods, (ii) matrix decomposition-based methods, (iii) network-based methods, and (iv) reverse transcriptome-based methods. In this article, we comprehensively review the widely used methods in the above four categories. Finally, we summarize the advantages and disadvantages of these methods and indicate future directions for more sensitive computational drug repositioning methods and individualized tumor treatment, which are critical for further experimental validation.
Collapse
Affiliation(s)
- Binsheng He
- Academician Workstation, Changsha Medical University, Changsha, China
| | - Fangxing Hou
- Queen Mary School, Nanchang University, Jiangxi, China
| | - Changjing Ren
- School of Science, Dalian Maritime University, Dalian, China.,Genies Beijing Co., Ltd., Beijing, China
| | - Pingping Bing
- Academician Workstation, Changsha Medical University, Changsha, China
| | - Xiangzuo Xiao
- Department of Radiology, The First Affiliated Hospital of Nanchang University, Jiangxi, China
| |
Collapse
|
4
|
Pulkkinen OI, Gautam P, Mustonen V, Aittokallio T. Multiobjective optimization identifies cancer-selective combination therapies. PLoS Comput Biol 2020; 16:e1008538. [PMID: 33370253 PMCID: PMC7793282 DOI: 10.1371/journal.pcbi.1008538] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 01/08/2021] [Accepted: 11/13/2020] [Indexed: 12/11/2022] Open
Abstract
Combinatorial therapies are required to treat patients with advanced cancers that have become resistant to monotherapies through rewiring of redundant pathways. Due to a massive number of potential drug combinations, there is a need for systematic approaches to identify safe and effective combinations for each patient, using cost-effective methods. Here, we developed an exact multiobjective optimization method for identifying pairwise or higher-order combinations that show maximal cancer-selectivity. The prioritization of patient-specific combinations is based on Pareto-optimization in the search space spanned by the therapeutic and nonselective effects of combinations. We demonstrate the performance of the method in the context of BRAF-V600E melanoma treatment, where the optimal solutions predicted a number of co-inhibition partners for vemurafenib, a selective BRAF-V600E inhibitor, approved for advanced melanoma. We experimentally validated many of the predictions in BRAF-V600E melanoma cell line, and the results suggest that one can improve selective inhibition of BRAF-V600E melanoma cells by combinatorial targeting of MAPK/ERK and other compensatory pathways using pairwise and third-order drug combinations. Our mechanism-agnostic optimization method is widely applicable to various cancer types, and it takes as input only measurements of a subset of pairwise drug combinations, without requiring target information or genomic profiles. Such data-driven approaches may become useful for functional precision oncology applications that go beyond the cancer genetic dependency paradigm to optimize cancer-selective combinatorial treatments.
Collapse
Affiliation(s)
- Otto I Pulkkinen
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland.,Helsinki Institute for Information Technology (HIIT), Department of Computer Science, University of Helsinki, Helsinki, Finland.,Computational Physics Laboratory, Tampere University, Tampere, Finland.,Department of Mathematics and Statistics, University of Turku, Turku, Finland
| | - Prson Gautam
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
| | - Ville Mustonen
- Helsinki Institute for Information Technology (HIIT), Department of Computer Science, University of Helsinki, Helsinki, Finland.,Organismal and Evolutionary Biology Research Programme, Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Tero Aittokallio
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland.,Department of Mathematics and Statistics, University of Turku, Turku, Finland.,Institute for Cancer Research, Department of Cancer Genetics, Oslo University Hospital, Oslo, Norway.,Oslo Centre for Biostatistics and Epidemiology (OCBE), Faculty of Medicine, University of Oslo, Oslo, Norway
| |
Collapse
|
5
|
Rahman R, Dhruba SR, Matlock K, De-Niz C, Ghosh S, Pal R. Evaluating the consistency of large-scale pharmacogenomic studies. Brief Bioinform 2020; 20:1734-1753. [PMID: 31846027 DOI: 10.1093/bib/bby046] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Revised: 05/04/2018] [Indexed: 12/21/2022] Open
Abstract
Recent years have seen an increase in the availability of pharmacogenomic databases such as Genomics of Drug Sensitivity in Cancer (GDSC) and Cancer Cell Line Encyclopedia (CCLE) that provide genomic and functional characterization information for multiple cell lines. Studies have alluded to the fact that specific characterizations may be inconsistent between different databases. Analysis of the potential discrepancies in the different databases is highly significant, as these sources are frequently used to analyze and validate methodologies for personalized cancer therapies. In this article, we review the recent developments in investigating the correspondence between different pharmacogenomics databases and discuss the potential factors that require attention when incorporating these sources in any modeling analysis. Furthermore, we explored the consistency among these databases using copulas that can capture nonlinear dependencies between two sets of data.
Collapse
Affiliation(s)
- Raziur Rahman
- Department of Electrical and Computer Engineering, Texas Tech University, Lubbock, TX 79409, USA
| | - Saugato Rahman Dhruba
- Department of Electrical and Computer Engineering, Texas Tech University, Lubbock, TX 79409, USA
| | - Kevin Matlock
- Department of Electrical and Computer Engineering, Texas Tech University, Lubbock, TX 79409, USA
| | - Carlos De-Niz
- Department of Electrical and Computer Engineering, Texas Tech University, Lubbock, TX 79409, USA
| | - Souparno Ghosh
- Department of Mathematics and Statistics, Texas Tech University, Lubbock, TX 79409, USA
| | - Ranadip Pal
- Department of Electrical and Computer Engineering, Texas Tech University, Lubbock, TX 79409, USA.,Department of Mathematics and Statistics, Texas Tech University, Lubbock, TX 79409, USA
| |
Collapse
|
6
|
Berlow NE, Grasso CS, Quist MJ, Cheng M, Gandour-Edwards R, Hernandez BS, Michalek JE, Ryan C, Spellman P, Pal R, Million LS, Renneker M, Keller C. Deep Functional and Molecular Characterization of a High-Risk Undifferentiated Pleomorphic Sarcoma. Sarcoma 2020; 2020:6312480. [PMID: 32565715 PMCID: PMC7285280 DOI: 10.1155/2020/6312480] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Revised: 02/07/2020] [Accepted: 02/10/2020] [Indexed: 11/29/2022] Open
Abstract
Nonrhabdomyosarcoma soft-tissue sarcomas (STSs) are a class of 50+ cancers arising in muscle and soft tissues of children, adolescents, and adults. Rarity of each subtype often precludes subtype-specific preclinical research, leaving many STS patients with limited treatment options should frontline therapy be insufficient. When clinical options are exhausted, personalized therapy assignment approaches may help direct patient care. Here, we report the results of an adult female STS patient with relapsed undifferentiated pleomorphic sarcoma (UPS) who self-drove exploration of a wide array of personalized Clinical Laboratory Improvement Amendments (CLIAs) level and research-level diagnostics, including state of the art genomic, proteomic, ex vivo live cell chemosensitivity testing, a patient-derived xenograft model, and immunoscoring. Her therapeutic choices were also diverse, including neoadjuvant chemotherapy, radiation therapy, and surgeries. Adjuvant and recurrence strategies included off-label and natural medicines, several immunotherapies, and N-of-1 approaches. Identified treatment options, especially those validated during the in vivo study, were not introduced into the course of clinical treatment but did provide plausible treatment regimens based on FDA-approved clinical agents.
Collapse
Affiliation(s)
- Noah E. Berlow
- Children's Cancer Therapy Development Institute, Beaverton, OR 97005, USA
- Electrical and Computer Engineering, Texas Tech University, Lubbock, TX 79409, USA
- Division of Hematology-Oncology, University of California, Los Angeles, CA 90095, USA
| | - Catherine S. Grasso
- Division of Hematology-Oncology, University of California, Los Angeles, CA 90095, USA
| | - Michael J. Quist
- Division of Hematology-Oncology, University of California, Los Angeles, CA 90095, USA
| | | | - Regina Gandour-Edwards
- Department of Pathology & Laboratory Medicine, UC Davis Health System, Sacramento, CA 95817, USA
| | - Brian S. Hernandez
- Department of Epidemiology and Biostatistics, University of Texas Health Science Center San Antonio, San Antonio, TX 78229, USA
| | - Joel E. Michalek
- Department of Epidemiology and Biostatistics, University of Texas Health Science Center San Antonio, San Antonio, TX 78229, USA
| | - Christopher Ryan
- School of Medicine, Oregon Health and Science University, Portland, OR 97239, USA
| | - Paul Spellman
- School of Medicine, Oregon Health and Science University, Portland, OR 97239, USA
| | - Ranadip Pal
- Electrical and Computer Engineering, Texas Tech University, Lubbock, TX 79409, USA
| | - Lynn S. Million
- Department of Radiation Oncology, Stanford University, Stanford, CA 94305, USA
| | - Mark Renneker
- Patient-Directed Consultations, San Francisco, CA 94116, USA
- Department of Family Medicine, University of California San Francisco, San Francisco, CA 94143, USA
| | - Charles Keller
- Children's Cancer Therapy Development Institute, Beaverton, OR 97005, USA
- Electrical and Computer Engineering, Texas Tech University, Lubbock, TX 79409, USA
| |
Collapse
|
7
|
Receptor Tyrosine Kinases in Osteosarcoma: 2019 Update. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1258:141-155. [PMID: 32767239 DOI: 10.1007/978-3-030-43085-6_9] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The primary conclusions of our 2014 contribution to this series were as follows: Multiple receptor tyrosine kinases (RTKs) likely contribute to aggressive phenotypes in osteosarcoma and, therefore, inhibition of multiple RTKs is likely necessary for successful clinical outcomes. Inhibition of multiple RTKs may also be useful to overcome resistance to inhibitors of individual RTKs as well as resistance to conventional chemotherapies. Different combinations of RTKs are likely important in individual patients. AXL, EPHB2, FGFR2, IGF1R, and RET were identified as promising therapeutic targets by our in vitro phosphoproteomic/siRNA screen of 42 RTKs in the highly metastatic LM7 and 143B human osteosarcoma cell lines. This chapter is intended to provide an update on these topics as well as the large number of osteosarcoma clinical studies of inhibitors of multiple tyrosine kinases (multi-TKIs) that were recently published.
Collapse
|
8
|
Naveja JJ, Stumpfe D, Medina-Franco JL, Bajorath J. Exploration of Target Synergy in Cancer Treatment by Cell-Based Screening Assay and Network Propagation Analysis. J Chem Inf Model 2019; 59:3072-3079. [PMID: 31013082 DOI: 10.1021/acs.jcim.9b00036] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Computational approaches have previously been introduced to predict compounds with activity against multiple targets or compound combinations with synergistic functional effects. By contrast, there are no computational studies available that explore combinations of targets that might act synergistically upon small molecule treatment. Herein, we introduce an approach designed to identify synergistic target pairs on the basis of cell-based screening data and compounds with known target annotations. The targets involved in forming synergistic pairs were analyzed through a novel network propagation algorithm for rationalizing possible common synergy mechanisms. This algorithm enabled further analysis of each synergistic target pair and the identification of "interactors", i.e., proteins with higher propagation scores than would be expected by adding the individual contributions of each target in the synergistic pair. We detected 137 synergistic target pairs including 51 unique targets. A global network analysis of these 51 targets made it possible to derive a subnetwork of proteins with significant synergy. Furthermore, interactors were identified for 87 synergistic target pairs upon individual analysis of the network propagation of each pair. These interactors were associated with pathways related to cancer and apoptosis, membrane transport, and steroid metabolism and provided possible explanations of synergistic effects.
Collapse
Affiliation(s)
- J Jesús Naveja
- Department of Life Science Informatics, B-IT, LIMES Program Unit Chemical Biology and Medicinal Chemistry , Rheinische Friedrich-Wilhelms-Universität , Endenicher Allee 19c , D-53115 Bonn , Germany.,PECEM, Faculty of Medicine , Universidad Nacional Autónoma de México , Mexico City , 04510 , Mexico.,Department of Pharmacy, School of Chemistry , Universidad Nacional Autónoma de México , Mexico City , 04510 , Mexico
| | - Dagmar Stumpfe
- Department of Life Science Informatics, B-IT, LIMES Program Unit Chemical Biology and Medicinal Chemistry , Rheinische Friedrich-Wilhelms-Universität , Endenicher Allee 19c , D-53115 Bonn , Germany
| | - José L Medina-Franco
- Department of Pharmacy, School of Chemistry , Universidad Nacional Autónoma de México , Mexico City , 04510 , Mexico
| | - Jürgen Bajorath
- Department of Life Science Informatics, B-IT, LIMES Program Unit Chemical Biology and Medicinal Chemistry , Rheinische Friedrich-Wilhelms-Universität , Endenicher Allee 19c , D-53115 Bonn , Germany
| |
Collapse
|
9
|
Berlow NE, Rikhi R, Geltzeiler M, Abraham J, Svalina MN, Davis LE, Wise E, Mancini M, Noujaim J, Mansoor A, Quist MJ, Matlock KL, Goros MW, Hernandez BS, Doung YC, Thway K, Tsukahara T, Nishio J, Huang ET, Airhart S, Bult CJ, Gandour-Edwards R, Maki RG, Jones RL, Michalek JE, Milovancev M, Ghosh S, Pal R, Keller C. Probabilistic modeling of personalized drug combinations from integrated chemical screen and molecular data in sarcoma. BMC Cancer 2019; 19:593. [PMID: 31208434 PMCID: PMC6580486 DOI: 10.1186/s12885-019-5681-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Accepted: 05/07/2019] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Cancer patients with advanced disease routinely exhaust available clinical regimens and lack actionable genomic medicine results, leaving a large patient population without effective treatments options when their disease inevitably progresses. To address the unmet clinical need for evidence-based therapy assignment when standard clinical approaches have failed, we have developed a probabilistic computational modeling approach which integrates molecular sequencing data with functional assay data to develop patient-specific combination cancer treatments. METHODS Tissue taken from a murine model of alveolar rhabdomyosarcoma was used to perform single agent drug screening and DNA/RNA sequencing experiments; results integrated via our computational modeling approach identified a synergistic personalized two-drug combination. Cells derived from the primary murine tumor were allografted into mouse models and used to validate the personalized two-drug combination. Computational modeling of single agent drug screening and RNA sequencing of multiple heterogenous sites from a single patient's epithelioid sarcoma identified a personalized two-drug combination effective across all tumor regions. The heterogeneity-consensus combination was validated in a xenograft model derived from the patient's primary tumor. Cell cultures derived from human and canine undifferentiated pleomorphic sarcoma were assayed by drug screen; computational modeling identified a resistance-abrogating two-drug combination common to both cell cultures. This combination was validated in vitro via a cell regrowth assay. RESULTS Our computational modeling approach addresses three major challenges in personalized cancer therapy: synergistic drug combination predictions (validated in vitro and in vivo in a genetically engineered murine cancer model), identification of unifying therapeutic targets to overcome intra-tumor heterogeneity (validated in vivo in a human cancer xenograft), and mitigation of cancer cell resistance and rewiring mechanisms (validated in vitro in a human and canine cancer model). CONCLUSIONS These proof-of-concept studies support the use of an integrative functional approach to personalized combination therapy prediction for the population of high-risk cancer patients lacking viable clinical options and without actionable DNA sequencing-based therapy.
Collapse
Affiliation(s)
- Noah E. Berlow
- Children’s Cancer Therapy Development Institute, 12655 SW Beaverdam Road-West, Beaverton, OR 97005 USA
- Electrical and Computer Engineering, Texas Tech University, Lubbock, TX 79409 USA
| | - Rishi Rikhi
- Children’s Cancer Therapy Development Institute, 12655 SW Beaverdam Road-West, Beaverton, OR 97005 USA
| | - Mathew Geltzeiler
- Department of Pediatrics, Oregon Health & Science University, Portland, OR 97239 USA
- Department of Otolaryngology – Head and Neck Surgery, Oregon Health & Science University, Portland, OR 97239 USA
| | - Jinu Abraham
- Department of Pediatrics, Oregon Health & Science University, Portland, OR 97239 USA
| | - Matthew N. Svalina
- Children’s Cancer Therapy Development Institute, 12655 SW Beaverdam Road-West, Beaverton, OR 97005 USA
| | - Lara E. Davis
- Department of Pediatrics, Oregon Health & Science University, Portland, OR 97239 USA
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239 USA
| | - Erin Wise
- Champions Oncology, Baltimore, MD 21205 USA
| | | | - Jonathan Noujaim
- Royal Marsden Hospital and Institute of Cancer Research, London, SW3 6JJ UK
- Hôpital Maisonneuve-Rosemont, Montreal, H1T 2M4 Canada
| | - Atiya Mansoor
- Department of Pathology, Oregon Health & Science University, Portland, OR 97239 USA
| | - Michael J. Quist
- Center for Spatial Systems Biomedicine Department of Molecular and Medical Genetics, Oregon Health & Science University, Portland, OR 97239 USA
| | - Kevin L. Matlock
- Electrical and Computer Engineering, Texas Tech University, Lubbock, TX 79409 USA
- Omics Data Automation, 12655 SW Beaverdam Road, Beaverton, OR 97005 USA
| | - Martin W. Goros
- Department of Epidemiology and Biostatistics, University of Texas Health Science Center San Antonio, San Antonio, TX 78229 USA
| | - Brian S. Hernandez
- Department of Epidemiology and Biostatistics, University of Texas Health Science Center San Antonio, San Antonio, TX 78229 USA
| | - Yee C. Doung
- Department of Orthopedic Surgery, Oregon Health & Science University, Portland, OR 97239 USA
| | - Khin Thway
- Royal Marsden Hospital and Institute of Cancer Research, London, SW3 6JJ UK
| | - Tomohide Tsukahara
- Department of Pathology, Sapporo Medical University School of Medicine, Sapporo, 060-8556 Japan
| | - Jun Nishio
- Department of Orthopaedic Surgery, Faculty of Medicine, Fukuoka University, Fukuoka, 814-0180 Japan
| | - Elaine T. Huang
- Department of Pediatrics, Oregon Health & Science University, Portland, OR 97239 USA
| | | | | | - Regina Gandour-Edwards
- Department of Pathology & Laboratory Medicine, UC Davis Health System, Sacramento, CA 95817 USA
| | - Robert G. Maki
- Sarcoma Program, Zucker School of Medicine at Hofstra/Northwell & Cold Spring Harbor Laboratory, Long Island, NY 10142 USA
| | - Robin L. Jones
- Royal Marsden Hospital and Institute of Cancer Research, London, SW3 6JJ UK
| | - Joel E. Michalek
- Department of Epidemiology and Biostatistics, University of Texas Health Science Center San Antonio, San Antonio, TX 78229 USA
| | - Milan Milovancev
- Carlson College of Veterinary Medicine, Oregon State University, Corvallis, OR 97331 USA
| | - Souparno Ghosh
- Department of Mathematics and Statistics, Texas Tech University, Lubbock, TX 79409 USA
| | - Ranadip Pal
- Electrical and Computer Engineering, Texas Tech University, Lubbock, TX 79409 USA
| | - Charles Keller
- Children’s Cancer Therapy Development Institute, 12655 SW Beaverdam Road-West, Beaverton, OR 97005 USA
| |
Collapse
|
10
|
Matlock K, Berlow N, Keller C, Pal R. Combination therapy design for maximizing sensitivity and minimizing toxicity. BMC Bioinformatics 2017; 18:116. [PMID: 28361667 PMCID: PMC5374708 DOI: 10.1186/s12859-017-1523-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Background Design of personalized targeted therapies involve modeling of patient sensitivity to various drugs and drug combinations. Majority of studies evaluate the sensitivity of tumor cells to targeted drugs without modeling the effect of the drugs on normal cells. In this article, we consider the individual modeling of drug responses to tumor and normal cells and utilize them to design targeted combination therapies that maximize sensitivity over tumor cells and minimize toxicity over normal cells. Results The problem is formulated as maximizing sensitivity over tumor cell models while maintaining sensitivity below a threshold over normal cell models. We utilize the constrained structure of tumor proliferation models to design an accelerated lexicographic search algorithm for generating the optimal solution. For comparison purposes, we also designed two suboptimal search algorithms based on evolutionary algorithms and hill-climbing based techniques. Results over synthetic models and models generated from Genomics of Drug Sensitivity in Cancer database shows the ability of the proposed algorithms to arrive at optimal or close to optimal solutions in significantly lower number of steps as compared to exhaustive search. We also present the theoretical analysis of the expected number of comparisons required for the proposed Lexicographic search that compare favorably with the observed number of computations. Conclusions The proposed algorithms provide a framework for design of combination therapy that tackles tumor heterogeneity while satisfying toxicity constraints.
Collapse
Affiliation(s)
- Kevin Matlock
- Department of Electrical and Computer Engineering, Texas Tech University, 1012 Boston Ave, Lubbock, 79409, TX, USA
| | - Noah Berlow
- Children's Cancer Therapy Development Institute, Portland, 97005, OR, USA
| | - Charles Keller
- Children's Cancer Therapy Development Institute, Portland, 97005, OR, USA
| | - Ranadip Pal
- Department of Electrical and Computer Engineering, Texas Tech University, 1012 Boston Ave, Lubbock, 79409, TX, USA.
| |
Collapse
|
11
|
Tan AC, Vyse S, Huang PH. Exploiting receptor tyrosine kinase co-activation for cancer therapy. Drug Discov Today 2017; 22:72-84. [PMID: 27452454 PMCID: PMC5346155 DOI: 10.1016/j.drudis.2016.07.010] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2016] [Revised: 06/15/2016] [Accepted: 07/15/2016] [Indexed: 01/04/2023]
Abstract
Studies over the past decade have shown that many cancers have evolved receptor tyrosine kinase (RTK) co-activation as a mechanism to drive tumour progression and limit the lethal effects of therapy. This review summarises the general principles of RTK co-activation and discusses approaches to exploit this phenomenon in cancer therapy and drug discovery. Computational strategies to predict kinase co-dependencies by integrating drug screening data and kinase inhibitor selectivity profiles will also be described. We offer a perspective on the implications of RTK co-activation on tumour heterogeneity and cancer evolution and conclude by surveying emerging computational and experimental approaches that will provide insights into RTK co-activation biology and deliver new developments in effective cancer therapies.
Collapse
Affiliation(s)
- Aik-Choon Tan
- Translational Bioinformatics and Cancer Systems Biology Laboratory, Division of Medical Oncology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
| | - Simon Vyse
- Division of Cancer Biology, The Institute of Cancer Research, London SW3 6JB, UK
| | - Paul H Huang
- Division of Cancer Biology, The Institute of Cancer Research, London SW3 6JB, UK.
| |
Collapse
|
12
|
|
13
|
Integrating Domain Specific Knowledge and Network Analysis to Predict Drug Sensitivity of Cancer Cell Lines. PLoS One 2016; 11:e0162173. [PMID: 27607242 PMCID: PMC5015856 DOI: 10.1371/journal.pone.0162173] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2015] [Accepted: 08/18/2016] [Indexed: 12/20/2022] Open
Abstract
One of fundamental challenges in cancer studies is that varying molecular characteristics of different tumor types may lead to resistance to certain drugs. As a result, the same drug can lead to significantly different results in different types of cancer thus emphasizing the need for individualized medicine. Individual prediction of drug response has great potential to aid in improving the clinical outcome and reduce the financial costs associated with prescribing chemotherapy drugs to which the patient's tumor might be resistant. In this paper we develop a network based classifier (NBC) method for predicting sensitivity of cell lines to anticancer drugs from transcriptome data. In the literature, this strategy has been used for predicting cancer types. Here, we extend it to estimate sensitivity of cells from different tumor types to various anticancer drugs. Furthermore, we incorporate domain specific knowledge such as the use of apoptotic gene list and clinical dose information in our method to impart biological significance to the prediction. Our experimental results suggest that our network based classifier (NBC) method outperforms existing classifiers in estimating sensitivity of cell lines for different drugs.
Collapse
|
14
|
Prioritization of anticancer drugs against a cancer using genomic features of cancer cells: A step towards personalized medicine. Sci Rep 2016; 6:23857. [PMID: 27030518 PMCID: PMC4814902 DOI: 10.1038/srep23857] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Accepted: 03/15/2016] [Indexed: 12/12/2022] Open
Abstract
In this study, we investigated drug profile of 24 anticancer drugs tested against a large number of cell lines in order to understand the relation between drug resistance and altered genomic features of a cancer cell line. We detected frequent mutations, high expression and high copy number variations of certain genes in both drug resistant cell lines and sensitive cell lines. It was observed that a few drugs, like Panobinostat, are effective against almost all types of cell lines, whereas certain drugs are effective against only a limited type of cell lines. Tissue-specific preference of drugs was also seen where a drug is more effective against cell lines belonging to a specific tissue. Genomic features based models have been developed for each anticancer drug and achieved average correlation between predicted and actual growth inhibition of cell lines in the range of 0.43 to 0.78. We hope, our study will throw light in the field of personalized medicine, particularly in designing patient-specific anticancer drugs. In order to serve the scientific community, a webserver, CancerDP, has been developed for predicting priority/potency of an anticancer drug against a cancer cell line using its genomic features (http://crdd.osdd.net/raghava/cancerdp/).
Collapse
|
15
|
Ryall KA, Kim J, Klauck PJ, Shin J, Yoo M, Ionkina A, Pitts TM, Tentler JJ, Diamond JR, Eckhardt SG, Heasley LE, Kang J, Tan AC. An integrated bioinformatics analysis to dissect kinase dependency in triple negative breast cancer. BMC Genomics 2015; 16 Suppl 12:S2. [PMID: 26681397 PMCID: PMC4682411 DOI: 10.1186/1471-2164-16-s12-s2] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Triple-Negative Breast Cancer (TNBC) is an aggressive disease with a poor prognosis. Clinically, TNBC patients have limited treatment options besides chemotherapy. The goal of this study was to determine the kinase dependency in TNBC cell lines and to predict compounds that could inhibit these kinases using integrative bioinformatics analysis. RESULTS We integrated publicly available gene expression data, high-throughput pharmacological profiling data, and quantitative in vitro kinase binding data to determine the kinase dependency in 12 TNBC cell lines. We employed Kinase Addiction Ranker (KAR), a novel bioinformatics approach, which integrated these data sources to dissect kinase dependency in TNBC cell lines. We then used the kinase dependency predicted by KAR for each TNBC cell line to query K-Map for compounds targeting these kinases. We validated our predictions using published and new experimental data. CONCLUSIONS In summary, we implemented an integrative bioinformatics analysis that determines kinase dependency in TNBC. Our analysis revealed candidate kinases as potential targets in TNBC for further pharmacological and biological studies.
Collapse
Affiliation(s)
- Karen A Ryall
- Division of Medical Oncology, Department of Medicine, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora CO 80045 USA
| | - Jihye Kim
- Division of Medical Oncology, Department of Medicine, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora CO 80045 USA
| | - Peter J Klauck
- Division of Medical Oncology, Department of Medicine, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora CO 80045 USA
| | - Jimin Shin
- Division of Medical Oncology, Department of Medicine, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora CO 80045 USA
| | - Minjae Yoo
- Division of Medical Oncology, Department of Medicine, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora CO 80045 USA
| | - Anastasia Ionkina
- Division of Medical Oncology, Department of Medicine, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora CO 80045 USA
| | - Todd M Pitts
- Division of Medical Oncology, Department of Medicine, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora CO 80045 USA
| | - John J Tentler
- Division of Medical Oncology, Department of Medicine, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora CO 80045 USA
| | - Jennifer R Diamond
- Division of Medical Oncology, Department of Medicine, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora CO 80045 USA
| | - S Gail Eckhardt
- Division of Medical Oncology, Department of Medicine, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora CO 80045 USA
| | - Lynn E Heasley
- Department of Craniofacial Biology, School of Dental Medicine, University of Colorado Anschutz Medical Campus, Aurora CO 80045 USA
| | - Jaewoo Kang
- Department of Computer Science, Korea University, Seoul, Korea
| | - Aik Choon Tan
- Division of Medical Oncology, Department of Medicine, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora CO 80045 USA
- Department of Computer Science, Korea University, Seoul, Korea
- Department of Biostatistics and Informatics, Colorado School of Public Health, University of Colorado Anschutz Medical Campus, Aurora CO 80045 USA
| |
Collapse
|
16
|
Ryall KA, Shin J, Yoo M, Hinz TK, Kim J, Kang J, Heasley LE, Tan AC. Identifying kinase dependency in cancer cells by integrating high-throughput drug screening and kinase inhibition data. Bioinformatics 2015. [PMID: 26206305 DOI: 10.1093/bioinformatics/btv427] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
MOTIVATION Targeted kinase inhibitors have dramatically improved cancer treatment, but kinase dependency for an individual patient or cancer cell can be challenging to predict. Kinase dependency does not always correspond with gene expression and mutation status. High-throughput drug screens are powerful tools for determining kinase dependency, but drug polypharmacology can make results difficult to interpret. RESULTS We developed Kinase Addiction Ranker (KAR), an algorithm that integrates high-throughput drug screening data, comprehensive kinase inhibition data and gene expression profiles to identify kinase dependency in cancer cells. We applied KAR to predict kinase dependency of 21 lung cancer cell lines and 151 leukemia patient samples using published datasets. We experimentally validated KAR predictions of FGFR and MTOR dependence in lung cancer cell line H1581, showing synergistic reduction in proliferation after combining ponatinib and AZD8055. AVAILABILITY AND IMPLEMENTATION KAR can be downloaded as a Python function or a MATLAB script along with example inputs and outputs at: http://tanlab.ucdenver.edu/KAR/. CONTACT aikchoon.tan@ucdenver.edu. SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Karen A Ryall
- Translational Bioinformatics and Cancer Systems Biology Laboratory, Division of Medical Oncology, Department of Medicine
| | - Jimin Shin
- Translational Bioinformatics and Cancer Systems Biology Laboratory, Division of Medical Oncology, Department of Medicine
| | - Minjae Yoo
- Translational Bioinformatics and Cancer Systems Biology Laboratory, Division of Medical Oncology, Department of Medicine
| | - Trista K Hinz
- Department of Craniofacial Biology, School of Dental Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Jihye Kim
- Translational Bioinformatics and Cancer Systems Biology Laboratory, Division of Medical Oncology, Department of Medicine
| | - Jaewoo Kang
- Department of Computer Science and Engineering, Korea University, Seoul, Korea and
| | - Lynn E Heasley
- Translational Bioinformatics and Cancer Systems Biology Laboratory, Division of Medical Oncology, Department of Medicine
| | - Aik Choon Tan
- Translational Bioinformatics and Cancer Systems Biology Laboratory, Division of Medical Oncology, Department of Medicine, Department of Computer Science and Engineering, Korea University, Seoul, Korea and Department of Biostatistics and Informatics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| |
Collapse
|
17
|
Grasso CS, Tang Y, Truffaux N, Berlow NE, Liu L, Debily MA, Quist MJ, Davis LE, Huang EC, Woo PJ, Ponnuswami A, Chen S, Johung TB, Sun W, Kogiso M, Du Y, Qi L, Huang Y, Hütt-Cabezas M, Warren KE, Le Dret L, Meltzer PS, Mao H, Quezado M, van Vuurden DG, Abraham J, Fouladi M, Svalina MN, Wang N, Hawkins C, Nazarian J, Alonso MM, Raabe EH, Hulleman E, Spellman PT, Li XN, Keller C, Pal R, Grill J, Monje M. Functionally defined therapeutic targets in diffuse intrinsic pontine glioma. Nat Med 2015; 21:555-9. [PMID: 25939062 PMCID: PMC4862411 DOI: 10.1038/nm.3855] [Citation(s) in RCA: 393] [Impact Index Per Article: 43.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2014] [Accepted: 04/10/2015] [Indexed: 12/16/2022]
Abstract
Diffuse Intrinsic Pontine Glioma (DIPG) is a fatal childhood cancer. We performed a chemical screen in patient-derived DIPG cultures along with RNAseq analyses and integrated computational modeling to identify potentially effective therapeutic strategies. The multi-histone deacetylase inhibitor panobinostat demonstrated efficacy in vitro and in DIPG orthotopic xenograft models. Combination testing of panobinostat with histone demethylase inhibitor GSKJ4 revealed synergy. Together, these data suggest a promising therapeutic strategy for DIPG.
Collapse
Affiliation(s)
- Catherine S Grasso
- Center for Spatial Systems Biomedicine, Department of Molecular and Medical Genetics, Oregon Health &Science University (OHSU), Portland, Oregon, USA
| | - Yujie Tang
- 1] Department of Neurology, Stanford University, Stanford, California, USA. [2] Department of Neurosurgery, Stanford University, Stanford, California, USA. [3] Department of Pediatrics, Stanford University, Stanford, California, USA. [4] Department of Pathology, Stanford University, Stanford, California, USA. [5] Present addresses: Key Laboratory of Cell Differentiation and Apoptosis of National Ministry of Education, Department of Pathophysiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China (Y.T.) and Department of Neurosurgery, The First Affiliated Hospital of Suzhou University, Suzhou, China (Y.H.)
| | | | - Noah E Berlow
- Electrical and Computer Engineering, Texas Tech University, Lubbock, Texas, USA
| | - Lining Liu
- 1] Department of Neurology, Stanford University, Stanford, California, USA. [2] Department of Neurosurgery, Stanford University, Stanford, California, USA. [3] Department of Pediatrics, Stanford University, Stanford, California, USA. [4] Department of Pathology, Stanford University, Stanford, California, USA
| | - Marie-Anne Debily
- 1] CNRS, UMR 8203, Gustave Roussy, Université Paris-Sud, Villejuif, France. [2] Département de biologie, Université d'Evry-Val d'Essone, Evry, France
| | - Michael J Quist
- Center for Spatial Systems Biomedicine, Department of Molecular and Medical Genetics, Oregon Health &Science University (OHSU), Portland, Oregon, USA
| | - Lara E Davis
- Pediatric Cancer Biology Program, Papé Family Pediatric Research Institute, Department of Pediatrics, OHSU, Portland, Oregon, USA
| | - Elaine C Huang
- Pediatric Cancer Biology Program, Papé Family Pediatric Research Institute, Department of Pediatrics, OHSU, Portland, Oregon, USA
| | - Pamelyn J Woo
- 1] Department of Neurology, Stanford University, Stanford, California, USA. [2] Department of Neurosurgery, Stanford University, Stanford, California, USA. [3] Department of Pediatrics, Stanford University, Stanford, California, USA. [4] Department of Pathology, Stanford University, Stanford, California, USA
| | - Anitha Ponnuswami
- 1] Department of Neurology, Stanford University, Stanford, California, USA. [2] Department of Neurosurgery, Stanford University, Stanford, California, USA. [3] Department of Pediatrics, Stanford University, Stanford, California, USA. [4] Department of Pathology, Stanford University, Stanford, California, USA
| | - Spenser Chen
- 1] Department of Neurology, Stanford University, Stanford, California, USA. [2] Department of Neurosurgery, Stanford University, Stanford, California, USA. [3] Department of Pediatrics, Stanford University, Stanford, California, USA. [4] Department of Pathology, Stanford University, Stanford, California, USA
| | - Tessa B Johung
- 1] Department of Neurology, Stanford University, Stanford, California, USA. [2] Department of Neurosurgery, Stanford University, Stanford, California, USA. [3] Department of Pediatrics, Stanford University, Stanford, California, USA. [4] Department of Pathology, Stanford University, Stanford, California, USA
| | - Wenchao Sun
- Department of Neurology, Stanford University, Stanford, California, USA
| | - Mari Kogiso
- Laboratory of Molecular Neurooncology, Texas Children's Cancer Center, Baylor College of Medicine, Houston, Texas, USA
| | - Yuchen Du
- Laboratory of Molecular Neurooncology, Texas Children's Cancer Center, Baylor College of Medicine, Houston, Texas, USA
| | - Lin Qi
- Laboratory of Molecular Neurooncology, Texas Children's Cancer Center, Baylor College of Medicine, Houston, Texas, USA
| | - Yulun Huang
- 1] Laboratory of Molecular Neurooncology, Texas Children's Cancer Center, Baylor College of Medicine, Houston, Texas, USA. [2] Present addresses: Key Laboratory of Cell Differentiation and Apoptosis of National Ministry of Education, Department of Pathophysiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China (Y.T.) and Department of Neurosurgery, The First Affiliated Hospital of Suzhou University, Suzhou, China (Y.H.)
| | - Marianne Hütt-Cabezas
- 1] Department of Oncology, Johns Hopkins University, Baltimore, Maryland, USA. [2] Department of Pathology, Johns Hopkins University, Baltimore, Maryland, USA
| | | | - Ludivine Le Dret
- CNRS, UMR 8203, Gustave Roussy, Université Paris-Sud, Villejuif, France
| | | | - Hua Mao
- Laboratory of Molecular Neurooncology, Texas Children's Cancer Center, Baylor College of Medicine, Houston, Texas, USA
| | | | - Dannis G van Vuurden
- 1] Department of Pediatric Oncology and Hematology, VU University Medical Center, Amsterdam, The Netherlands. [2] Neuro-Oncology Research Group Cancer Center Amsterdam, VU University Medical Center, Amsterdam, The Netherlands
| | - Jinu Abraham
- Pediatric Cancer Biology Program, Papé Family Pediatric Research Institute, Department of Pediatrics, OHSU, Portland, Oregon, USA
| | - Maryam Fouladi
- Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Matthew N Svalina
- 1] Center for Spatial Systems Biomedicine, Department of Molecular and Medical Genetics, Oregon Health &Science University (OHSU), Portland, Oregon, USA. [2] Children's Cancer Therapy Development Institute, Fort Collins, Colorado, USA
| | - Nicholas Wang
- Center for Spatial Systems Biomedicine, Department of Molecular and Medical Genetics, Oregon Health &Science University (OHSU), Portland, Oregon, USA
| | - Cynthia Hawkins
- 1] Department of Pediatric Laboratory Medicine, University of Toronto, Toronto, Ontario, Canada. [2] Labatt Brain Tumor Research Centre, Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | - Javad Nazarian
- Center for Research Institute, Children's National Health Systems, Washington, DC, USA
| | - Marta M Alonso
- Department of Oncology, University Hospital of Navarra, Pamplona, Spain
| | - Eric H Raabe
- 1] Department of Oncology, Johns Hopkins University, Baltimore, Maryland, USA. [2] Department of Pathology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Esther Hulleman
- 1] Department of Pediatric Oncology and Hematology, VU University Medical Center, Amsterdam, The Netherlands. [2] Neuro-Oncology Research Group Cancer Center Amsterdam, VU University Medical Center, Amsterdam, The Netherlands
| | - Paul T Spellman
- Center for Spatial Systems Biomedicine, Department of Molecular and Medical Genetics, Oregon Health &Science University (OHSU), Portland, Oregon, USA
| | - Xiao-Nan Li
- Laboratory of Molecular Neurooncology, Texas Children's Cancer Center, Baylor College of Medicine, Houston, Texas, USA
| | - Charles Keller
- 1] Pediatric Cancer Biology Program, Papé Family Pediatric Research Institute, Department of Pediatrics, OHSU, Portland, Oregon, USA. [2] Children's Cancer Therapy Development Institute, Fort Collins, Colorado, USA
| | - Ranadip Pal
- Electrical and Computer Engineering, Texas Tech University, Lubbock, Texas, USA
| | - Jacques Grill
- 1] CNRS, UMR 8203, Gustave Roussy, Université Paris-Sud, Villejuif, France. [2] Departement de Cancerologie de l'Enfant et de l'Adolescent, Institut Gustave Roussy, Université Paris-Sud, Villejuif, France
| | - Michelle Monje
- 1] Department of Neurology, Stanford University, Stanford, California, USA. [2] Department of Neurosurgery, Stanford University, Stanford, California, USA. [3] Department of Pediatrics, Stanford University, Stanford, California, USA. [4] Department of Pathology, Stanford University, Stanford, California, USA
| |
Collapse
|
18
|
Ryall KA, Tan AC. Systems biology approaches for advancing the discovery of effective drug combinations. J Cheminform 2015; 7:7. [PMID: 25741385 PMCID: PMC4348553 DOI: 10.1186/s13321-015-0055-9] [Citation(s) in RCA: 95] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2014] [Accepted: 02/02/2015] [Indexed: 01/23/2023] Open
Abstract
Complex diseases like cancer are regulated by large, interconnected networks with many pathways affecting cell proliferation, invasion, and drug resistance. However, current cancer therapy predominantly relies on the reductionist approach of one gene-one disease. Combinations of drugs may overcome drug resistance by limiting mutations and induction of escape pathways, but given the enormous number of possible drug combinations, strategies to reduce the search space and prioritize experiments are needed. In this review, we focus on the use of computational modeling, bioinformatics and high-throughput experimental methods for discovery of drug combinations. We highlight cutting-edge systems approaches, including large-scale modeling of cell signaling networks, network motif analysis, statistical association-based models, identifying correlations in gene signatures, functional genomics, and high-throughput combination screens. We also present a list of publicly available data and resources to aid in discovery of drug combinations. Integration of these systems approaches will enable faster discovery and translation of clinically relevant drug combinations. Spectrum of Systems Biology Approaches for Drug Combinations. ![]()
Collapse
Affiliation(s)
- Karen A Ryall
- Translational Bioinformatics and Cancer Systems Biology Laboratory, Division of Medical Oncology, Department of Medicine, School of Medicine, University of Colorado Anschutz Medical Campus, 12801 E.17th Ave., L18-8116, Aurora, CO 80045 USA
| | - Aik Choon Tan
- Translational Bioinformatics and Cancer Systems Biology Laboratory, Division of Medical Oncology, Department of Medicine, School of Medicine, University of Colorado Anschutz Medical Campus, 12801 E.17th Ave., L18-8116, Aurora, CO 80045 USA ; Department of Biostatistics and Informatics, Colorado School of Public Health, University of Colorado Anschutz Medical Campus, Aurora, CO USA ; Department of Computer Science and Engineering, Korea University, Seoul, South Korea
| |
Collapse
|
19
|
Hejase HA, Chan C. Improving Drug Sensitivity Prediction Using Different Types of Data. CPT-PHARMACOMETRICS & SYSTEMS PHARMACOLOGY 2015. [PMID: 26225231 PMCID: PMC4360670 DOI: 10.1002/psp4.2] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The algorithms and models used to address the two subchallenges that are part of the NCI-DREAM (Dialogue for Reverse Engineering Assessments and Methods) Drug Sensitivity Prediction Challenge (2012) are presented. In subchallenge 1, a bidirectional search algorithm is introduced and optimized using an ensemble scheme and a nonlinear support vector machine (SVM) is then applied to predict the effects of the drug compounds on breast cancer cell lines. In subchallenge 2, a weighted Euclidean distance method is introduced to predict and rank the drug combinations from the most to the least effective in reducing the viability of a diffuse large B-cell lymphoma (DLBCL) cell line.
Collapse
Affiliation(s)
- H A Hejase
- Department of Computer Science and Engineering, Michigan State University East Lansing, Michigan, USA
| | - C Chan
- Department of Computer Science and Engineering, Michigan State University East Lansing, Michigan, USA ; Department of Chemical Engineering and Materials Science, Michigan State University East Lansing, Michigan, USA ; Department of Biochemistry and Molecular Biology, Michigan State University East Lansing, Michigan, USA
| |
Collapse
|
20
|
Berlow N, Haider S, Wan Q, Geltzeiler M, Davis LE, Keller C, Pal R. An Integrated Approach to Anti-Cancer Drug Sensitivity Prediction. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2014; 11:995-1008. [PMID: 26357038 DOI: 10.1109/tcbb.2014.2321138] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
A framework for design of personalized cancer therapy requires the ability to predict the sensitivity of a tumor to anticancer drugs. The predictive modeling of tumor sensitivity to anti-cancer drugs has primarily focused on generating functions that map gene expressions and genetic mutation profiles to drug sensitivity. In this paper, we present a new approach for drug sensitivity prediction and combination therapy design based on integrated functional and genomic characterizations. The modeling approach when applied to data from the Cancer Cell Line Encyclopedia shows a significant gain in prediction accuracy as compared to elastic net and random forest techniques based on genomic characterizations. Utilizing a Mouse Embryonal Rhabdomyosarcoma cell culture and a drug screen of 60 targeted drugs, we show that predictive modeling based on functional data alone can also produce high accuracy predictions. The framework also allows us to generate personalized tumor proliferation circuits to gain further insights on the individualized biological pathway.
Collapse
|
21
|
Berlow N, Davis L, Keller C, Pal R. Inference of dynamic biological networks based on responses to drug perturbations. EURASIP JOURNAL ON BIOINFORMATICS & SYSTEMS BIOLOGY 2014; 2014:14. [PMID: 28194164 PMCID: PMC5270455 DOI: 10.1186/s13637-014-0014-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/31/2014] [Accepted: 07/21/2014] [Indexed: 12/23/2022]
Abstract
Drugs that target specific proteins are a major paradigm in cancer research. In this article, we extend a modeling framework for drug sensitivity prediction and combination therapy design based on drug perturbation experiments. The recently proposed target inhibition map approach can infer stationary pathway models from drug perturbation experiments, but the method is limited to a steady-state snapshot of the underlying dynamical model. We consider the inverse problem of possible dynamic models that can generate the static target inhibition map model. From a deterministic viewpoint, we analyze the inference of Boolean networks that can generate the observed binarized sensitivities under different target inhibition scenarios. From a stochastic perspective, we investigate the generation of Markov chain models that satisfy the observed target inhibition sensitivities.
Collapse
Affiliation(s)
- Noah Berlow
- Department of Electrical and Computer Engineering, Texas Tech University, Lubbock, 79409 TX USA
| | - Lara Davis
- Department of Pediatrics, Oregon Health & Science University, Portland, 97239 OR USA
| | - Charles Keller
- Department of Pediatrics, Oregon Health & Science University, Portland, 97239 OR USA
| | - Ranadip Pal
- Department of Electrical and Computer Engineering, Texas Tech University, Lubbock, 79409 TX USA
| |
Collapse
|
22
|
A diverse stochastic search algorithm for combination therapeutics. BIOMED RESEARCH INTERNATIONAL 2014; 2014:873436. [PMID: 24738075 PMCID: PMC3971504 DOI: 10.1155/2014/873436] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/17/2013] [Revised: 01/20/2014] [Accepted: 02/06/2014] [Indexed: 11/26/2022]
Abstract
Background. Design of drug combination cocktails to maximize sensitivity for individual patients presents a challenge in terms of minimizing the number of experiments to attain the desired objective. The enormous number of possible drug combinations constrains exhaustive experimentation approaches, and personal variations in genetic diseases restrict the use of prior knowledge in optimization. Results. We present a stochastic search algorithm that consisted of a parallel experimentation phase followed by a combination of focused and diversified sequential search. We evaluated our approach on seven synthetic examples; four of them were evaluated twice with different parameters, and two biological examples of bacterial and lung cancer cell inhibition response to combination drugs. The performance of our approach as compared to recently proposed adaptive reference update approach was superior for all the examples considered, achieving an average of 45% reduction in the number of experimental iterations. Conclusions. As the results illustrate, the proposed diverse stochastic search algorithm can produce optimized combinations in relatively smaller number of iterative steps. This approach can be combined with available knowledge on the genetic makeup of the patient to design optimal selection of drug cocktails.
Collapse
|