101
|
Catara G, Spano D. Combinatorial Strategies to Target Molecular and Signaling Pathways to Disarm Cancer Stem Cells. Front Oncol 2021; 11:689131. [PMID: 34381714 PMCID: PMC8352560 DOI: 10.3389/fonc.2021.689131] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 07/01/2021] [Indexed: 12/14/2022] Open
Abstract
Cancer is an urgent public health issue with a very huge number of cases all over the world expected to increase by 2040. Despite improved diagnosis and therapeutic protocols, it remains the main leading cause of death in the world. Cancer stem cells (CSCs) constitute a tumor subpopulation defined by ability to self-renewal and to generate the heterogeneous and differentiated cell lineages that form the tumor bulk. These cells represent a major concern in cancer treatment due to resistance to conventional protocols of radiotherapy, chemotherapy and molecular targeted therapy. In fact, although partial or complete tumor regression can be achieved in patients, these responses are often followed by cancer relapse due to the expansion of CSCs population. The aberrant activation of developmental and oncogenic signaling pathways plays a relevant role in promoting CSCs therapy resistance. Although several targeted approaches relying on monotherapy have been developed to affect these pathways, they have shown limited efficacy. Therefore, an urgent need to design alternative combinatorial strategies to replace conventional regimens exists. This review summarizes the preclinical studies which provide a proof of concept of therapeutic efficacy of combinatorial approaches targeting the CSCs.
Collapse
Affiliation(s)
- Giuliana Catara
- Institute of Biochemistry and Cell Biology, National Research Council, Naples, Italy
| | - Daniela Spano
- Institute of Biochemistry and Cell Biology, National Research Council, Naples, Italy
| |
Collapse
|
102
|
Mohanty S, Rashid MHA, Mohanty C, Swayamsiddha S. Modern computational intelligence based drug repurposing for diabetes epidemic. Diabetes Metab Syndr 2021; 15:102180. [PMID: 34186343 DOI: 10.1016/j.dsx.2021.06.017] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 06/12/2021] [Accepted: 06/14/2021] [Indexed: 12/18/2022]
Abstract
BACKGROUND AND AIM Objectives are to explore recent advances in discovery of new antidiabetic agents using repurposing strategies and to discuss modern technologies used for drug repurposing highlighting diabetic specific web portal. METHODS Recent literature were studied and analyzed from various sources such as Scopus, PubMed, and IEEE Xplore databases. RESULTS Drugs like Niclosamideethanolamine, Methazolamide, Diacerein, Berberine, Clobetasol, etc. with possibility of repurposing to curb diabetes can be potential late-stage clinical candidates, providing access to information on pharmacology, formulation, and probable toxicity if any. CONCLUSIONS With collaboration of artificial intelligence (AI) with pharmacology, the efficiency of drug repurposing can improve significantly.
Collapse
Affiliation(s)
- Sweta Mohanty
- School of Applied Science, KIIT University, Bhubaneswar, Odisha, India
| | | | - Chandana Mohanty
- School of Applied Science, KIIT University, Bhubaneswar, Odisha, India.
| | - Swati Swayamsiddha
- School of Electronics Engineering, KIIT University, Bhubaneswar, Odisha, India.
| |
Collapse
|
103
|
Park Y, Heider D, Hauschild AC. Integrative Analysis of Next-Generation Sequencing for Next-Generation Cancer Research toward Artificial Intelligence. Cancers (Basel) 2021; 13:3148. [PMID: 34202427 PMCID: PMC8269018 DOI: 10.3390/cancers13133148] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 06/16/2021] [Accepted: 06/21/2021] [Indexed: 12/18/2022] Open
Abstract
The rapid improvement of next-generation sequencing (NGS) technologies and their application in large-scale cohorts in cancer research led to common challenges of big data. It opened a new research area incorporating systems biology and machine learning. As large-scale NGS data accumulated, sophisticated data analysis methods became indispensable. In addition, NGS data have been integrated with systems biology to build better predictive models to determine the characteristics of tumors and tumor subtypes. Therefore, various machine learning algorithms were introduced to identify underlying biological mechanisms. In this work, we review novel technologies developed for NGS data analysis, and we describe how these computational methodologies integrate systems biology and omics data. Subsequently, we discuss how deep neural networks outperform other approaches, the potential of graph neural networks (GNN) in systems biology, and the limitations in NGS biomedical research. To reflect on the various challenges and corresponding computational solutions, we will discuss the following three topics: (i) molecular characteristics, (ii) tumor heterogeneity, and (iii) drug discovery. We conclude that machine learning and network-based approaches can add valuable insights and build highly accurate models. However, a well-informed choice of learning algorithm and biological network information is crucial for the success of each specific research question.
Collapse
Affiliation(s)
- Youngjun Park
- Department of Mathematics and Computer Science, Philipps-University of Marburg, 35032 Marburg, Germany; (Y.P.); (D.H.)
| | - Dominik Heider
- Department of Mathematics and Computer Science, Philipps-University of Marburg, 35032 Marburg, Germany; (Y.P.); (D.H.)
| | - Anne-Christin Hauschild
- Department of Mathematics and Computer Science, Philipps-University of Marburg, 35032 Marburg, Germany; (Y.P.); (D.H.)
- Department of Medical Informatics, University Medical Center Göttingen, 37075 Göttingen, Germany
| |
Collapse
|
104
|
Falvo P, Orecchioni S, Roma S, Raveane A, Bertolini F. Drug Repurposing in Oncology, an Attractive Opportunity for Novel Combinatorial Regimens. Curr Med Chem 2021; 28:2114-2136. [PMID: 33109033 DOI: 10.2174/0929867327999200817104912] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 05/21/2020] [Accepted: 05/26/2020] [Indexed: 11/22/2022]
Abstract
The costs of developing, validating and buying new drugs are dramatically increasing. On the other hand, sobering economies have difficulties in sustaining their healthcare systems, particularly in countries with an elderly population requiring increasing welfare. This conundrum requires immediate action, and a possible option is to study the large, already present arsenal of drugs approved and to use them for innovative therapies. This possibility is particularly interesting in oncology, where the complexity of the cancer genome dictates in most patients a multistep therapeutic approach. In this review, we discuss a) Computational approaches; b) preclinical models; c) currently ongoing or already published clinical trials in the drug repurposing field in oncology; and d) drug repurposing to overcome resistance to previous therapies.
Collapse
Affiliation(s)
- Paolo Falvo
- Laboratory of Hematology-Oncology, European Institute of Oncology IRCCS, 20141 Milan, Italy
| | - Stefania Orecchioni
- Laboratory of Hematology-Oncology, European Institute of Oncology IRCCS, 20141 Milan, Italy
| | - Stefania Roma
- Laboratory of Hematology-Oncology, European Institute of Oncology IRCCS, 20141 Milan, Italy
| | - Alessandro Raveane
- Laboratory of Hematology-Oncology, European Institute of Oncology IRCCS, 20141 Milan, Italy
| | - Francesco Bertolini
- Laboratory of Hematology-Oncology, European Institute of Oncology IRCCS, 20141 Milan, Italy
| |
Collapse
|
105
|
Samart K, Tuyishime P, Krishnan A, Ravi J. Reconciling multiple connectivity scores for drug repurposing. Brief Bioinform 2021; 22:6278144. [PMID: 34013329 PMCID: PMC8597919 DOI: 10.1093/bib/bbab161] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 04/02/2021] [Accepted: 04/07/2021] [Indexed: 12/16/2022] Open
Abstract
The basis of several recent methods for drug repurposing is the key principle that an
efficacious drug will reverse the disease molecular ‘signature’ with minimal side effects.
This principle was defined and popularized by the influential ‘connectivity map’ study in
2006 regarding reversal relationships between disease- and drug-induced gene expression
profiles, quantified by a disease-drug ‘connectivity score.’ Over the past 15 years,
several studies have proposed variations in calculating connectivity scores toward
improving accuracy and robustness in light of massive growth in reference drug profiles.
However, these variations have been formulated inconsistently using various notations and
terminologies even though they are based on a common set of conceptual and statistical
ideas. Therefore, we present a systematic reconciliation of multiple disease-drug
similarity metrics (\documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{upgreek}
\usepackage{mathrsfs}
\setlength{\oddsidemargin}{-69pt}
\begin{document}
}{}$ES$\end{document}, \documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{upgreek}
\usepackage{mathrsfs}
\setlength{\oddsidemargin}{-69pt}
\begin{document}
}{}$css$\end{document},
\documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{upgreek}
\usepackage{mathrsfs}
\setlength{\oddsidemargin}{-69pt}
\begin{document}
}{}$Sum$\end{document}, \documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{upgreek}
\usepackage{mathrsfs}
\setlength{\oddsidemargin}{-69pt}
\begin{document}
}{}$Cosine$\end{document},
\documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{upgreek}
\usepackage{mathrsfs}
\setlength{\oddsidemargin}{-69pt}
\begin{document}
}{}$XSum$\end{document}, \documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{upgreek}
\usepackage{mathrsfs}
\setlength{\oddsidemargin}{-69pt}
\begin{document}
}{}$XCor$\end{document},
\documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{upgreek}
\usepackage{mathrsfs}
\setlength{\oddsidemargin}{-69pt}
\begin{document}
}{}$XSpe$\end{document}, \documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{upgreek}
\usepackage{mathrsfs}
\setlength{\oddsidemargin}{-69pt}
\begin{document}
}{}$XCos$\end{document},
\documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{upgreek}
\usepackage{mathrsfs}
\setlength{\oddsidemargin}{-69pt}
\begin{document}
}{}$EWCos$\end{document}) and connectivity scores
(\documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{upgreek}
\usepackage{mathrsfs}
\setlength{\oddsidemargin}{-69pt}
\begin{document}
}{}$CS$\end{document}, \documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{upgreek}
\usepackage{mathrsfs}
\setlength{\oddsidemargin}{-69pt}
\begin{document}
}{}$RGES$\end{document},
\documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{upgreek}
\usepackage{mathrsfs}
\setlength{\oddsidemargin}{-69pt}
\begin{document}
}{}$NCS$\end{document}, \documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{upgreek}
\usepackage{mathrsfs}
\setlength{\oddsidemargin}{-69pt}
\begin{document}
}{}$WCS$\end{document},
\documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{upgreek}
\usepackage{mathrsfs}
\setlength{\oddsidemargin}{-69pt}
\begin{document}
}{}$Tau$\end{document}, \documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{upgreek}
\usepackage{mathrsfs}
\setlength{\oddsidemargin}{-69pt}
\begin{document}
}{}$CSS$\end{document},
\documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{upgreek}
\usepackage{mathrsfs}
\setlength{\oddsidemargin}{-69pt}
\begin{document}
}{}$EMUDRA$\end{document}) by defining them using consistent
notation and terminology. In addition to providing clarity and deeper insights, this
coherent definition of connectivity scores and their relationships provides a unified
scheme that newer methods can adopt, enabling the computational drug-development community
to compare and investigate different approaches easily. To facilitate the continuous and
transparent integration of newer methods, this article will be available as a live
document (https://jravilab.github.io/connectivity_scores) coupled with a GitHub
repository (https://github.com/jravilab/connectivity_scores) that any researcher can
build on and push changes to.
Collapse
Affiliation(s)
- Kewalin Samart
- Computational Mathematics, and Computational Math, Science & Engineering at Michigan State University, East Lansing, MI, USA
| | - Phoebe Tuyishime
- College of Agriculture and Natural Resources at Michigan State University, East Lansing, MI, USA
| | - Arjun Krishnan
- Departments of Computational Math, Science & Engineering, and Biochemistry & Molecular Biology at Michigan State University, East Lansing, MI, USA
| | - Janani Ravi
- Pathobiology and Diagnostic Investigation at Michigan State University, East Lansing, MI, USA
| |
Collapse
|
106
|
Yin J, Li X, Li F, Lu Y, Zeng S, Zhu F. Identification of the key target profiles underlying the drugs of narrow therapeutic index for treating cancer and cardiovascular disease. Comput Struct Biotechnol J 2021; 19:2318-2328. [PMID: 33995923 PMCID: PMC8105181 DOI: 10.1016/j.csbj.2021.04.035] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 04/09/2021] [Accepted: 04/15/2021] [Indexed: 12/14/2022] Open
Abstract
An appropriate therapeutic index is crucial for drug discovery and development since narrow therapeutic index (NTI) drugs with slight dosage variation may induce severe adverse drug reactions or potential treatment failure. To date, the shared characteristics underlying the targets of NTI drugs have been explored by several studies, which have been applied to identify potential drug targets. However, the association between the drug therapeutic index and the related disease has not been dissected, which is important for revealing the NTI drug mechanism and optimizing drug design. Therefore, in this study, two classes of disease (cancers and cardiovascular disorders) with the largest number of NTI drugs were selected, and the target property of the corresponding NTI drugs was analyzed. By calculating the biological system profiles and human protein–protein interaction (PPI) network properties of drug targets and adopting an AI-based algorithm, differentiated features between two diseases were discovered to reveal the distinct underlying mechanisms of NTI drugs in different diseases. Consequently, ten shared features and four unique features were identified for both diseases to distinguish NTI from NNTI drug targets. These computational discoveries, as well as the newly found features, suggest that in the clinical study of avoiding narrow therapeutic index in those diseases, the ability of target to be a hub and the efficiency of target signaling in the human PPI network should be considered, and it could thus provide novel guidance in the drug discovery and clinical research process and help to estimate the drug safety of cancer and cardiovascular disease.
Collapse
Affiliation(s)
- Jiayi Yin
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Xiaoxu Li
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Fengcheng Li
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Yinjing Lu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Su Zeng
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China.,Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Hangzhou 310018, China
| | - Feng Zhu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China.,Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Hangzhou 310018, China.,Alibaba-Zhejiang University Joint Research Center of Future Digital Healthcare, Hangzhou 330110, China
| |
Collapse
|
107
|
Nie X, Takalkar MA, Duan M, Zhang H, Xu M. GEME: Dual-stream multi-task GEnder-based micro-expression recognition. Neurocomputing 2021. [DOI: 10.1016/j.neucom.2020.10.082] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
108
|
Hernández-Lemus E, Martínez-García M. Pathway-Based Drug-Repurposing Schemes in Cancer: The Role of Translational Bioinformatics. Front Oncol 2021; 10:605680. [PMID: 33520715 PMCID: PMC7841291 DOI: 10.3389/fonc.2020.605680] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2020] [Accepted: 11/24/2020] [Indexed: 12/11/2022] Open
Abstract
Cancer is a set of complex pathologies that has been recognized as a major public health problem worldwide for decades. A myriad of therapeutic strategies is indeed available. However, the wide variability in tumor physiology, response to therapy, added to multi-drug resistance poses enormous challenges in clinical oncology. The last years have witnessed a fast-paced development of novel experimental and translational approaches to therapeutics, that supplemented with computational and theoretical advances are opening promising avenues to cope with cancer defiances. At the core of these advances, there is a strong conceptual shift from gene-centric emphasis on driver mutations in specific oncogenes and tumor suppressors-let us call that the silver bullet approach to cancer therapeutics-to a systemic, semi-mechanistic approach based on pathway perturbations and global molecular and physiological regulatory patterns-we will call this the shrapnel approach. The silver bullet approach is still the best one to follow when clonal mutations in driver genes are present in the patient, and when there are targeted therapies to tackle those. Unfortunately, due to the heterogeneous nature of tumors this is not the common case. The wide molecular variability in the mutational level often is reduced to a much smaller set of pathway-based dysfunctions as evidenced by the well-known hallmarks of cancer. In such cases "shrapnel gunshots" may become more effective than "silver bullets". Here, we will briefly present both approaches and will abound on the discussion on the state of the art of pathway-based therapeutic designs from a translational bioinformatics and computational oncology perspective. Further development of these approaches depends on building collaborative, multidisciplinary teams to resort to the expertise of clinical oncologists, oncological surgeons, and molecular oncologists, but also of cancer cell biologists and pharmacologists, as well as bioinformaticians, computational biologists and data scientists. These teams will be capable of engaging on a cycle of analyzing high-throughput experiments, mining databases, researching on clinical data, validating the findings, and improving clinical outcomes for the benefits of the oncological patients.
Collapse
Affiliation(s)
- Enrique Hernández-Lemus
- Computational Genomics Division, National Institute of Genomic Medicine, Mexico City, Mexico
- Centro de Ciencias de la Complejidad, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Mireya Martínez-García
- Sociomedical Research Unit, National Institute of Cardiology “Ignacio Chávez”, Mexico City, Mexico
| |
Collapse
|
109
|
|
110
|
|
111
|
Kumar R, Dhanda SK. Bird Eye View of Protein Subcellular Localization Prediction. Life (Basel) 2020; 10:E347. [PMID: 33327400 PMCID: PMC7764902 DOI: 10.3390/life10120347] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 12/11/2020] [Accepted: 12/11/2020] [Indexed: 12/12/2022] Open
Abstract
Proteins are made up of long chain of amino acids that perform a variety of functions in different organisms. The activity of the proteins is determined by the nucleotide sequence of their genes and by its 3D structure. In addition, it is essential for proteins to be destined to their specific locations or compartments to perform their structure and functions. The challenge of computational prediction of subcellular localization of proteins is addressed in various in silico methods. In this review, we reviewed the progress in this field and offered a bird eye view consisting of a comprehensive listing of tools, types of input features explored, machine learning approaches employed, and evaluation matrices applied. We hope the review will be useful for the researchers working in the field of protein localization predictions.
Collapse
Affiliation(s)
- Ravindra Kumar
- Biometric Research Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, NIH, 9609 Medical Center Drive, Rockville, MD 20850, USA
| | - Sandeep Kumar Dhanda
- Department of Oncology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| |
Collapse
|
112
|
Mokou M, Lygirou V, Angelioudaki I, Paschalidis N, Stroggilos R, Frantzi M, Latosinska A, Bamias A, Hoffmann MJ, Mischak H, Vlahou A. A Novel Pipeline for Drug Repurposing for Bladder Cancer Based on Patients' Omics Signatures. Cancers (Basel) 2020; 12:E3519. [PMID: 33255925 PMCID: PMC7759896 DOI: 10.3390/cancers12123519] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 11/19/2020] [Accepted: 11/20/2020] [Indexed: 12/24/2022] Open
Abstract
Multi-omics signatures of patients with bladder cancer (BC) can guide the identification of known de-risked therapeutic compounds through drug repurposing, an approach not extensively explored yet. In this study, we target drug repurposing in the context of BC, driven by tissue omics signatures. To identify compounds that can reverse aggressive high-risk Non-Muscle Invasive BC (NMIBC) to less aggressive low-risk molecular subtypes, the next generation Connectivity Map (CMap) was employed using as input previously published proteomics and transcriptomics respective signatures. Among the identified compounds, the ATP-competitive inhibitor of mTOR, WYE-354, showed a consistently very high score for reversing the aggressive BC molecular signatures. WYE-354 impact was assessed in a panel of eight multi-origin BC cell lines and included impaired colony growth and proliferation rate without any impact on apoptosis. Overall, with this study we introduce a promising pipeline for the repurposing of drugs for BC treatment, based on patients' omics signatures.
Collapse
Affiliation(s)
- Marika Mokou
- Biotechnology Division, Biomedical Research Foundation of the Academy of Athens, 11527 Athens, Greece; (M.M.); (V.L.); (I.A.); (R.S.)
- Mosaiques Diagnostics GmbH, 30659 Hannover, Germany; (M.F.); (A.L.); (H.M.)
| | - Vasiliki Lygirou
- Biotechnology Division, Biomedical Research Foundation of the Academy of Athens, 11527 Athens, Greece; (M.M.); (V.L.); (I.A.); (R.S.)
| | - Ioanna Angelioudaki
- Biotechnology Division, Biomedical Research Foundation of the Academy of Athens, 11527 Athens, Greece; (M.M.); (V.L.); (I.A.); (R.S.)
| | - Nikolaos Paschalidis
- Cellular Immunology Laboratory, Center for Basic Research, Biomedical Research Foundation of the Academy of Athens, 11527 Athens, Greece;
| | - Rafael Stroggilos
- Biotechnology Division, Biomedical Research Foundation of the Academy of Athens, 11527 Athens, Greece; (M.M.); (V.L.); (I.A.); (R.S.)
| | - Maria Frantzi
- Mosaiques Diagnostics GmbH, 30659 Hannover, Germany; (M.F.); (A.L.); (H.M.)
| | | | - Aristotelis Bamias
- Haematology-Oncology Unit, Department of Clinical Therapeutics, Alexandra Hospital, National and Kapodistrian University of Athens, 11528 Athens, Greece;
| | - Michèle J. Hoffmann
- Department of Urology, Medical Faculty, Heinrich-Heine-University Düsseldorf, 40225 Düsseldorf, Germany;
| | - Harald Mischak
- Mosaiques Diagnostics GmbH, 30659 Hannover, Germany; (M.F.); (A.L.); (H.M.)
- British Heart Foundation Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow G12 8QQ, UK
| | - Antonia Vlahou
- Biotechnology Division, Biomedical Research Foundation of the Academy of Athens, 11527 Athens, Greece; (M.M.); (V.L.); (I.A.); (R.S.)
| |
Collapse
|
113
|
Cabrera-Andrade A, López-Cortés A, Jaramillo-Koupermann G, González-Díaz H, Pazos A, Munteanu CR, Pérez-Castillo Y, Tejera E. A Multi-Objective Approach for Anti-Osteosarcoma Cancer Agents Discovery through Drug Repurposing. Pharmaceuticals (Basel) 2020; 13:ph13110409. [PMID: 33266378 PMCID: PMC7700154 DOI: 10.3390/ph13110409] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 11/11/2020] [Accepted: 11/12/2020] [Indexed: 02/08/2023] Open
Abstract
Osteosarcoma is the most common type of primary malignant bone tumor. Although nowadays 5-year survival rates can reach up to 60–70%, acute complications and late effects of osteosarcoma therapy are two of the limiting factors in treatments. We developed a multi-objective algorithm for the repurposing of new anti-osteosarcoma drugs, based on the modeling of molecules with described activity for HOS, MG63, SAOS2, and U2OS cell lines in the ChEMBL database. Several predictive models were obtained for each cell line and those with accuracy greater than 0.8 were integrated into a desirability function for the final multi-objective model. An exhaustive exploration of model combinations was carried out to obtain the best multi-objective model in virtual screening. For the top 1% of the screened list, the final model showed a BEDROC = 0.562, EF = 27.6, and AUC = 0.653. The repositioning was performed on 2218 molecules described in DrugBank. Within the top-ranked drugs, we found: temsirolimus, paclitaxel, sirolimus, everolimus, and cabazitaxel, which are antineoplastic drugs described in clinical trials for cancer in general. Interestingly, we found several broad-spectrum antibiotics and antiretroviral agents. This powerful model predicts several drugs that should be studied in depth to find new chemotherapy regimens and to propose new strategies for osteosarcoma treatment.
Collapse
Affiliation(s)
- Alejandro Cabrera-Andrade
- Grupo de Bio-Quimioinformática, Universidad de Las Américas, Quito 170125, Ecuador;
- Carrera de Enfermería, Facultad de Ciencias de la Salud, Universidad de Las Américas, Quito 170125, Ecuador
- Department of Computer Science and Information Technologies, Faculty of Computer Science, University of A Coruña, CITIC, Campus Elviña s/n, 15071 A Coruña, Spain; (A.L.-C.); (A.P.); (C.R.M.)
- Correspondence: (A.C.-A.); (E.T.)
| | - Andrés López-Cortés
- Department of Computer Science and Information Technologies, Faculty of Computer Science, University of A Coruña, CITIC, Campus Elviña s/n, 15071 A Coruña, Spain; (A.L.-C.); (A.P.); (C.R.M.)
- Centro de Investigación Genética y Genómica, Facultad de Ciencias de la Salud Eugenio Espejo, Universidad UTE, Quito 170129, Ecuador
- Latin American Network for Implementation and Validation of Clinical Pharmacogenomics Guidelines (RELIVAF-CYTED), 28029 Madrid, Spain
| | - Gabriela Jaramillo-Koupermann
- Laboratorio de Biología Molecular, Subproceso de Anatomía Patológica, Hospital de Especialidades Eugenio Espejo, Quito 170403, Ecuador;
| | - Humberto González-Díaz
- Department of Organic and Inorganic Chemistry, and Basque Center for Biophysics CSIC-UPV/EHU, University of the Basque Country UPV/EHU, 48940 Leioa, Spain;
- IKERBASQUE, Basque Foundation for Science, 48011 Bilbao, Spain
| | - Alejandro Pazos
- Department of Computer Science and Information Technologies, Faculty of Computer Science, University of A Coruña, CITIC, Campus Elviña s/n, 15071 A Coruña, Spain; (A.L.-C.); (A.P.); (C.R.M.)
- Biomedical Research Institute of A Coruña (INIBIC), University Hospital Complex of A Coruña (CHUAC), 15006 A Coruña, Spain
| | - Cristian R. Munteanu
- Department of Computer Science and Information Technologies, Faculty of Computer Science, University of A Coruña, CITIC, Campus Elviña s/n, 15071 A Coruña, Spain; (A.L.-C.); (A.P.); (C.R.M.)
- Biomedical Research Institute of A Coruña (INIBIC), University Hospital Complex of A Coruña (CHUAC), 15006 A Coruña, Spain
| | - Yunierkis Pérez-Castillo
- Grupo de Bio-Quimioinformática, Universidad de Las Américas, Quito 170125, Ecuador;
- Escuela de Ciencias Físicas y Matemáticas, Universidad de Las Américas, Quito 170125, Ecuador
| | - Eduardo Tejera
- Grupo de Bio-Quimioinformática, Universidad de Las Américas, Quito 170125, Ecuador;
- Facultad de Ingeniería y Ciencias Agropecuarias, Universidad de Las Américas, Quito 170125, Ecuador
- Correspondence: (A.C.-A.); (E.T.)
| |
Collapse
|
114
|
Gan X, Luo Y, Dai G, Lin J, Liu X, Zhang X, Li A. Identification of Gene Signatures for Diagnosis and Prognosis of Hepatocellular Carcinomas Patients at Early Stage. Front Genet 2020; 11:857. [PMID: 32849835 PMCID: PMC7406719 DOI: 10.3389/fgene.2020.00857] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Accepted: 07/14/2020] [Indexed: 12/12/2022] Open
Abstract
The onset of liver cancer is insidious. Currently, there is no effective method for the early detection of hepatocellular carcinoma (HCC). Transcriptomic profiles of 826 tissue samples from the Gene Expression Omnibus (GEO), The Cancer Genome Atlas (TCGA), Genotype tissue expression (GTEx), and International Cancer Genome Consortium (ICGC) databases were utilized to establish models for early detection and surveillance of HCC. The overlapping differentially expressed genes (DEGs) were screened by elastic net and robust rank aggregation (RRA) analyses to construct the diagnostic prediction model for early HCC (DP.eHCC). Prognostic prediction genes were screened by univariate cox regression and lasso cox regression analyses to construct the survival risk prediction model for early HCC (SP.eHCC). The relationship between the variation of transcriptome profile and the oncogenic risk-score of early HCC was analyzed by combining Weighted Correlation Network Analysis (WGCNA), Gene Set Enrichment Analysis (GSEA), and genome networks (GeNets). The results showed that the AUC of DP.eHCC model for the diagnosis of early HCC was 0.956 (95% CI: 0.941–0.972; p < 0.001) with a sensitivity of 90.91%, a specificity of 92.97%. The SP.eHCC model performed well for predicting the overall survival risk of HCC patients (HR = 10.79; 95% CI: 6.16–18.89; p < 0.001). The oncogenesis of early HCC was revealed mainly involving in pathways associated with cell proliferation and tumor microenvironment. And the transcription factors including EZH2, EGR1, and SOX17 were screened in the genome networks as the promising targets used for precise treatment in patients with HCC. Our findings provide robust models for the early diagnosis and prognosis of HCC, and are crucial for the development of novel targets applied in the precision therapy of HCC.
Collapse
Affiliation(s)
- Xiaoning Gan
- Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China.,Cancer Center, Southern Medical University, Guangzhou, China.,Department of Physiology, Michigan State University, East Lansing, MI, United States
| | - Yue Luo
- Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China.,Cancer Center, Southern Medical University, Guangzhou, China.,Department of Physiology, Michigan State University, East Lansing, MI, United States
| | - Guanqi Dai
- Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China.,Cancer Center, Southern Medical University, Guangzhou, China
| | - Junhao Lin
- Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China.,Cancer Center, Southern Medical University, Guangzhou, China
| | - Xinhui Liu
- Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China.,Cancer Center, Southern Medical University, Guangzhou, China.,Department of Physiology, Michigan State University, East Lansing, MI, United States
| | - Xiangqun Zhang
- Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Aimin Li
- Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China.,Cancer Center, Southern Medical University, Guangzhou, China.,Department of Physiology, Michigan State University, East Lansing, MI, United States
| |
Collapse
|
115
|
Abstract
The current global pandemic COVID-19 caused by the SARS-CoV-2 virus has already inflicted insurmountable damage both to the human lives and global economy. There is an immediate need for identification of effective drugs to contain the disastrous virus outbreak. Global efforts are already underway at a war footing to identify the best drug combination to address the disease. In this review, an attempt has been made to understand the SARS-CoV-2 life cycle, and based on this information potential druggable targets against SARS-CoV-2 are summarized. Also, the strategies for ongoing and future drug discovery against the SARS-CoV-2 virus are outlined. Given the urgency to find a definitive cure, ongoing drug repurposing efforts being carried out by various organizations are also described. The unprecedented crisis requires extraordinary efforts from the scientific community to effectively address the issue and prevent further loss of human lives and health.
Collapse
Affiliation(s)
- Ambrish Saxena
- Indian Institute of Technology Tirupati, Tirupati, India
| |
Collapse
|
116
|
Zhang Z, Zhou L, Xie N, Nice EC, Zhang T, Cui Y, Huang C. Overcoming cancer therapeutic bottleneck by drug repurposing. Signal Transduct Target Ther 2020; 5:113. [PMID: 32616710 PMCID: PMC7331117 DOI: 10.1038/s41392-020-00213-8] [Citation(s) in RCA: 290] [Impact Index Per Article: 58.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 06/03/2020] [Accepted: 06/04/2020] [Indexed: 02/06/2023] Open
Abstract
Ever present hurdles for the discovery of new drugs for cancer therapy have necessitated the development of the alternative strategy of drug repurposing, the development of old drugs for new therapeutic purposes. This strategy with a cost-effective way offers a rare opportunity for the treatment of human neoplastic disease, facilitating rapid clinical translation. With an increased understanding of the hallmarks of cancer and the development of various data-driven approaches, drug repurposing further promotes the holistic productivity of drug discovery and reasonably focuses on target-defined antineoplastic compounds. The "treasure trove" of non-oncology drugs should not be ignored since they could target not only known but also hitherto unknown vulnerabilities of cancer. Indeed, different from targeted drugs, these old generic drugs, usually used in a multi-target strategy may bring benefit to patients. In this review, aiming to demonstrate the full potential of drug repurposing, we present various promising repurposed non-oncology drugs for clinical cancer management and classify these candidates into their proposed administration for either mono- or drug combination therapy. We also summarize approaches used for drug repurposing and discuss the main barriers to its uptake.
Collapse
Affiliation(s)
- Zhe Zhang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, 610041, Chengdu, China
| | - Li Zhou
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, 610041, Chengdu, China
| | - Na Xie
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, 610041, Chengdu, China
| | - Edouard C Nice
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC, Australia
| | - Tao Zhang
- The School of Biological Science and Technology, Chengdu Medical College, 610083, Chengdu, China.
- Department of Oncology, The Second Affiliated Hospital of Chengdu Medical College, China National Nuclear Corporation 416 Hospital, Chengdu, 610051, Sichuan, China.
| | - Yongping Cui
- Cancer Institute, Peking University Shenzhen Hospital, Shenzhen Peking University-the Hong Kong University of Science and Technology (PKU-HKUST) Medical Center, and Cancer Institute, Shenzhen Bay Laboratory Shenzhen, 518035, Shenzhen, China.
- Department of Pathology & Shanxi Key Laboratory of Carcinogenesis and Translational Research on Esophageal Cancer, Shanxi Medical University, Taiyuan, 030001, Shanxi, China.
| | - Canhua Huang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, 610041, Chengdu, China.
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, Sichuan, China.
| |
Collapse
|
117
|
Coulouarn C. Artificial intelligence and omics in cancer. Artif Intell Cancer 2020; 1:1-7. [DOI: 10.35713/aic.v1.i1.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 06/09/2020] [Accepted: 06/12/2020] [Indexed: 02/06/2023] Open
Abstract
Cancer is a major public health problem worldwide. Current predictions suggest that 13 million people will die each year from cancer by 2030. Thus, new ideas are urgently needed to change paradigms in the global fight against cancer. Over the last decades, artificial intelligence (AI) emerged in the field of cancer research as a new and promising discipline. Although emerging, a great potential is appreciated in AI to improve cancer diagnosis and prognosis, as well as to identify relevant therapeutics in the current era of personalized medicine. Developing pipelines connecting patient-generated health data easily translatable into clinical practice to assist clinicians in decision making represents a challenging but fascinating task. AI algorithms are mainly fueled by multi omics data which, in the case of cancer research, have been largely derived from international cancer programs, including The Cancer Genome Atlas (TCGA). Here, I briefly review some examples of supervised and unsupervised big data derived from TCGA programs and comment on how AI algorithms have been applied to improve the management of patients with cancer. In this context, Artificial Intelligence in Cancer journal was specifically launched to promote the development of this discipline, by serving as a forum to publish high-quality basic and clinical research articles in various fields of AI in oncology.
Collapse
Affiliation(s)
- Cédric Coulouarn
- Institut National de la Sante et de la Recherche Medicale (Inserm), Université de Rennes 1, Rennes F-35000, France
| |
Collapse
|
118
|
Jia Y, Wen X, Gong Y, Wang X. Current scenario of indole derivatives with potential anti-drug-resistant cancer activity. Eur J Med Chem 2020; 200:112359. [PMID: 32531682 DOI: 10.1016/j.ejmech.2020.112359] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 04/09/2020] [Accepted: 04/16/2020] [Indexed: 02/07/2023]
Abstract
Cancer chemotherapy is frequently hampered by drug resistance, so the resistance to anticancer agents represents one of the major obstacles for the effective cancer treatment. Indole derivatives have the potential to act on diverse targets in cancer cells and exhibit promising activity against drug-resistant cancers. Moreover, some indole-containing compounds such as Semaxanib, Sunitinib, Vinorelbine, and Vinblastine have already been applied in clinics for various kinds of cancer even drug-resistant cancer therapy. Thus, indole derivatives are one of significant resources for the development of novel anti-drug-resistant cancer agents. This review focuses on the recent development of indole derivatives with potential therapeutic application for drug-resistant cancers, and the mechanisms of action, the critical aspects of design as well as structure-activity relationships, covering articles published from 2010 to 2020.
Collapse
Affiliation(s)
- Yanshu Jia
- Chongqing Institute of Engineering, Chongqing, 400056, China
| | - Xiaoyue Wen
- The Institute of Infection and Inflammation, China Three Gorges University, Yichang, Hubei, 443000, China
| | - Yufeng Gong
- The Second Affiliated Hospital of Mudanjiang Medical University, Mudanjiang, 157000, China
| | - Xuefeng Wang
- Department of Surgery, Zhuji Affiliated Hospital of Shaoxing University, Zhejiang Province, 311800, China.
| |
Collapse
|