Recent Developments in Cancer Systems Biology

A special issue of Journal of Personalized Medicine (ISSN 2075-4426). This special issue belongs to the section "Mechanisms of Diseases".

Deadline for manuscript submissions: closed (10 December 2020) | Viewed by 51200

Special Issue Editors


E-Mail Website
Guest Editor
Department of Biochemistry and Molecular Biology, Penn State College of Medicine, Hershey, PA, USA
Interests: anti-cancer mechanisms of organo-selenium compounds; cancer biomarkers; chemoprevention; cancer therapy; dietary manipulations; inflammation; systems biology
Special Issues, Collections and Topics in MDPI journals

E-Mail Website
Guest Editor
Department of Bioengineering, Marmara University, 34722 Goztepe, Istanbul, Turkey
Interests: systems biology; bioinformatics; multi-omics data analysis; personalized medicine

Special Issue Information

Dear Colleagues,

Cancer is a complex disease that involves multifaceted mechanisms at transcriptional, translational, and biochemical levels. In addition, several molecules become important players in cancer progression and provide conducive environment for protein–protein interactions, metabolite cross-talk, and cell signaling. Studying such molecules may offer discovery of biomarkers, targetable clues for potentially repurposing drugs, as well as insights for developing predictive models of prognosis and improving cancer-free survival in patients. Systems biology has undoubtedly emerged as an integrative tool to achieve such advances. The Journal of Personalized Medicine is now opening a Special Issue that is fully devoted to ideas and research that build a strong foundation of Systems Biology in cancer research, with a call for papers involving basic, translational, and clinical research on this topic.

Dr. Raghu Sinha
Dr. Kazim Yalcin Arga
Guest Editors

Manuscript Submission Information

Manuscripts should be submitted online at www.mdpi.com by registering and logging in to this website. Once you are registered, click here to go to the submission form. Manuscripts can be submitted until the deadline. All submissions that pass pre-check are peer-reviewed. Accepted papers will be published continuously in the journal (as soon as accepted) and will be listed together on the special issue website. Research articles, review articles as well as short communications are invited. For planned papers, a title and short abstract (about 100 words) can be sent to the Editorial Office for announcement on this website.

Submitted manuscripts should not have been published previously, nor be under consideration for publication elsewhere (except conference proceedings papers). All manuscripts are thoroughly refereed through a single-blind peer-review process. A guide for authors and other relevant information for submission of manuscripts is available on the Instructions for Authors page. Journal of Personalized Medicine is an international peer-reviewed open access monthly journal published by MDPI.

Please visit the Instructions for Authors page before submitting a manuscript. The Article Processing Charge (APC) for publication in this open access journal is 2600 CHF (Swiss Francs). Submitted papers should be well formatted and use good English. Authors may use MDPI's English editing service prior to publication or during author revisions.

Keywords

  • systems biology
  • biomarkers
  • drug repurposing
  • predictive models

Published Papers (13 papers)

Order results
Result details
Select all
Export citation of selected articles as:

Editorial

Jump to: Research, Review

3 pages, 180 KiB  
Editorial
Recent Developments in Cancer Systems Biology: Lessons Learned and Future Directions
by Kazim Y. Arga and Raghu Sinha
J. Pers. Med. 2021, 11(4), 271; https://0-doi-org.brum.beds.ac.uk/10.3390/jpm11040271 - 04 Apr 2021
Cited by 3 | Viewed by 1842
Abstract
Cancer is a complex disease involving multiple mechanisms and critical players, at broad genomic, transcriptional, translational and/or biochemical levels [...] Full article
(This article belongs to the Special Issue Recent Developments in Cancer Systems Biology)

Research

Jump to: Editorial, Review

19 pages, 3581 KiB  
Article
Data-Driven Math Model of FLT3-ITD Acute Myeloid Leukemia Reveals Potential Therapeutic Targets
by David J. Wooten, Melat Gebru, Hong-Gang Wang and Réka Albert
J. Pers. Med. 2021, 11(3), 193; https://0-doi-org.brum.beds.ac.uk/10.3390/jpm11030193 - 11 Mar 2021
Cited by 9 | Viewed by 3276
Abstract
FLT3-mutant acute myeloid leukemia (AML) is an aggressive form of leukemia with poor prognosis. Treatment with FLT3 inhibitors frequently produces a clinical response, but the disease nevertheless often recurs. Recent studies have revealed system-wide gene expression changes in FLT3-mutant AML cell [...] Read more.
FLT3-mutant acute myeloid leukemia (AML) is an aggressive form of leukemia with poor prognosis. Treatment with FLT3 inhibitors frequently produces a clinical response, but the disease nevertheless often recurs. Recent studies have revealed system-wide gene expression changes in FLT3-mutant AML cell lines in response to drug treatment. Here we sought a systems-level understanding of how these cells mediate these drug-induced changes. Using RNAseq data from AML cells with an internal tandem duplication FLT3 mutation (FLT3-ITD) under six drug treatment conditions including quizartinib and dexamethasone, we identified seven distinct gene programs representing diverse biological processes involved in AML drug-induced changes. Based on the literature knowledge about genes from these modules, along with public gene regulatory network databases, we constructed a network of FLT3-ITD AML. Applying the BooleaBayes algorithm to this network and the RNAseq data, we created a probabilistic, data-driven dynamical model of acquired resistance to these drugs. Analysis of this model reveals several interventions that may disrupt targeted parts of the system-wide drug response. We anticipate co-targeting these points may result in synergistic treatments that can overcome resistance and prevent eventual recurrence. Full article
(This article belongs to the Special Issue Recent Developments in Cancer Systems Biology)
Show Figures

Figure 1

11 pages, 1990 KiB  
Article
Differential Interactome Proposes Subtype-Specific Biomarkers and Potential Therapeutics in Renal Cell Carcinomas
by Aysegul Caliskan, Gizem Gulfidan, Raghu Sinha and Kazim Yalcin Arga
J. Pers. Med. 2021, 11(2), 158; https://0-doi-org.brum.beds.ac.uk/10.3390/jpm11020158 - 23 Feb 2021
Cited by 8 | Viewed by 2403
Abstract
Although many studies have been conducted on single gene therapies in cancer patients, the reality is that tumor arises from different coordinating protein groups. Unveiling perturbations in protein interactome related to the tumor formation may contribute to the development of effective diagnosis, treatment [...] Read more.
Although many studies have been conducted on single gene therapies in cancer patients, the reality is that tumor arises from different coordinating protein groups. Unveiling perturbations in protein interactome related to the tumor formation may contribute to the development of effective diagnosis, treatment strategies, and prognosis. In this study, considering the clinical and transcriptome data of three Renal Cell Carcinoma (RCC) subtypes (ccRCC, pRCC, and chRCC) retrieved from The Cancer Genome Atlas (TCGA) and the human protein interactome, the differential protein–protein interactions were identified in each RCC subtype. The approach enabled the identification of differentially interacting proteins (DIPs) indicating prominent changes in their interaction patterns during tumor formation. Further, diagnostic and prognostic performances were generated by taking into account DIP clusters which are specific to the relevant subtypes. Furthermore, considering the mesenchymal epithelial transition (MET) receptor tyrosine kinase (PDB ID: 3DKF) as a potential drug target specific to pRCC, twenty-one lead compounds were identified through virtual screening of ZINC molecules. In this study, we presented remarkable findings in terms of early diagnosis, prognosis, and effective treatment strategies, that deserve further experimental and clinical efforts. Full article
(This article belongs to the Special Issue Recent Developments in Cancer Systems Biology)
Show Figures

Figure 1

28 pages, 11840 KiB  
Article
Comprehensive Profiling of Genomic and Transcriptomic Differences between Risk Groups of Lung Adenocarcinoma and Lung Squamous Cell Carcinoma
by Talip Zengin and Tuğba Önal-Süzek
J. Pers. Med. 2021, 11(2), 154; https://0-doi-org.brum.beds.ac.uk/10.3390/jpm11020154 - 23 Feb 2021
Cited by 21 | Viewed by 4077
Abstract
Lung cancer is the second most frequently diagnosed cancer type and responsible for the highest number of cancer deaths worldwide. Lung adenocarcinoma (LUAD) and lung squamous cell carcinoma (LUSC) are subtypes of non-small-cell lung cancer which has the highest frequency of lung cancer [...] Read more.
Lung cancer is the second most frequently diagnosed cancer type and responsible for the highest number of cancer deaths worldwide. Lung adenocarcinoma (LUAD) and lung squamous cell carcinoma (LUSC) are subtypes of non-small-cell lung cancer which has the highest frequency of lung cancer cases. We aimed to analyze genomic and transcriptomic variations including simple nucleotide variations (SNVs), copy number variations (CNVs) and differential expressed genes (DEGs) in order to find key genes and pathways for diagnostic and prognostic prediction for lung adenocarcinoma and lung squamous cell carcinoma. We performed a univariate Cox model and then lasso-regularized Cox model with leave-one-out cross-validation using The Cancer Genome Atlas (TCGA) gene expression data in tumor samples. We generated 35- and 33-gene signatures for prognostic risk prediction based on the overall survival time of the patients with LUAD and LUSC, respectively. When we clustered patients into high- and low-risk groups, the survival analysis showed highly significant results with high prediction power for both training and test datasets. Then, we characterized the differences including significant SNVs, CNVs, DEGs, active subnetworks, and the pathways. We described the results for the risk groups and cancer subtypes separately to identify specific genomic alterations between both high-risk groups and cancer subtypes. Both LUAD and LUSC high-risk groups have more downregulated immune pathways and upregulated metabolic pathways. On the other hand, low-risk groups have both up- and downregulated genes on cancer-related pathways. Both LUAD and LUSC have important gene alterations such as CDKN2A and CDKN2B deletions with different frequencies. SOX2 amplification occurs in LUSC and PSMD4 amplification in LUAD. EGFR and KRAS mutations are mutually exclusive in LUAD samples. EGFR, MGA, SMARCA4, ATM, RBM10, and KDM5C genes are mutated only in LUAD but not in LUSC. CDKN2A, PTEN, and HRAS genes are mutated only in LUSC samples. The low-risk groups of both LUAD and LUSC tend to have a higher number of SNVs, CNVs, and DEGs. The signature genes and altered genes have the potential to be used as diagnostic and prognostic biomarkers for personalized oncology. Full article
(This article belongs to the Special Issue Recent Developments in Cancer Systems Biology)
Show Figures

Figure 1

18 pages, 2925 KiB  
Article
Identification of Somatic Structural Variants in Solid Tumors by Optical Genome Mapping
by David Y. Goldrich, Brandon LaBarge, Scott Chartrand, Lijun Zhang, Henry B. Sadowski, Yang Zhang, Khoa Pham, Hannah Way, Chi-Yu Jill Lai, Andy Wing Chun Pang, Benjamin Clifford, Alex R. Hastie, Mark Oldakowski, David Goldenberg and James R. Broach
J. Pers. Med. 2021, 11(2), 142; https://0-doi-org.brum.beds.ac.uk/10.3390/jpm11020142 - 18 Feb 2021
Cited by 18 | Viewed by 4415
Abstract
Genomic structural variants comprise a significant fraction of somatic mutations driving cancer onset and progression. However, such variants are not readily revealed by standard next-generation sequencing. Optical genome mapping (OGM) surpasses short-read sequencing in detecting large (>500 bp) and complex structural variants (SVs) [...] Read more.
Genomic structural variants comprise a significant fraction of somatic mutations driving cancer onset and progression. However, such variants are not readily revealed by standard next-generation sequencing. Optical genome mapping (OGM) surpasses short-read sequencing in detecting large (>500 bp) and complex structural variants (SVs) but requires isolation of ultra-high-molecular-weight DNA from the tissue of interest. We have successfully applied a protocol involving a paramagnetic nanobind disc to a wide range of solid tumors. Using as little as 6.5 mg of input tumor tissue, we show successful extraction of high-molecular-weight genomic DNA that provides a high genomic map rate and effective coverage by optical mapping. We demonstrate the system’s utility in identifying somatic SVs affecting functional and cancer-related genes for each sample. Duplicate/triplicate analysis of select samples shows intra-sample reliability but also intra-sample heterogeneity. We also demonstrate that simply filtering SVs based on a GRCh38 human control database provides high positive and negative predictive values for true somatic variants. Our results indicate that the solid tissue DNA extraction protocol, OGM and SV analysis can be applied to a wide variety of solid tumors to capture SVs across the entire genome with functional importance in cancer prognosis and treatment. Full article
(This article belongs to the Special Issue Recent Developments in Cancer Systems Biology)
Show Figures

Figure 1

16 pages, 4640 KiB  
Article
Gene Regulatory Network of ETS Domain Transcription Factors in Different Stages of Glioma
by Yigit Koray Babal, Basak Kandemir and Isil Aksan Kurnaz
J. Pers. Med. 2021, 11(2), 138; https://0-doi-org.brum.beds.ac.uk/10.3390/jpm11020138 - 17 Feb 2021
Cited by 7 | Viewed by 3203
Abstract
The ETS domain family of transcription factors is involved in a number of biological processes, and is commonly misregulated in various forms of cancer. Using microarray datasets from patients with different grades of glioma, we have analyzed the expression profiles of various ETS [...] Read more.
The ETS domain family of transcription factors is involved in a number of biological processes, and is commonly misregulated in various forms of cancer. Using microarray datasets from patients with different grades of glioma, we have analyzed the expression profiles of various ETS genes, and have identified ETV1, ELK3, ETV4, ELF4, and ETV6 as novel biomarkers for the identification of different glioma grades. We have further analyzed the gene regulatory networks of ETS transcription factors and compared them to previous microarray studies, where Elk-1-VP16 or PEA3-VP16 were overexpressed in neuroblastoma cell lines, and we identify unique and common regulatory networks for these ETS proteins. Full article
(This article belongs to the Special Issue Recent Developments in Cancer Systems Biology)
Show Figures

Graphical abstract

27 pages, 2753 KiB  
Article
ETS-Domain Transcription Factor Elk-1 Regulates Stemness Genes in Brain Tumors and CD133+ BrainTumor-Initiating Cells
by Melis Savasan Sogut, Chitra Venugopal, Basak Kandemir, Ugur Dag, Sujeivan Mahendram, Sheila Singh, Gizem Gulfidan, Kazim Yalcin Arga, Bayram Yilmaz and Isil Aksan Kurnaz
J. Pers. Med. 2021, 11(2), 125; https://0-doi-org.brum.beds.ac.uk/10.3390/jpm11020125 - 14 Feb 2021
Cited by 8 | Viewed by 2988
Abstract
Elk-1, a member of the ternary complex factors (TCFs) within the ETS (E26 transformation-specific) domain superfamily, is a transcription factor implicated in neuroprotection, neurodegeneration, and brain tumor proliferation. Except for known targets, c-fos and egr-1, few targets of Elk-1 have been identified. [...] Read more.
Elk-1, a member of the ternary complex factors (TCFs) within the ETS (E26 transformation-specific) domain superfamily, is a transcription factor implicated in neuroprotection, neurodegeneration, and brain tumor proliferation. Except for known targets, c-fos and egr-1, few targets of Elk-1 have been identified. Interestingly, SMN, SOD1, and PSEN1 promoters were shown to be regulated by Elk-1. On the other hand, Elk-1 was shown to regulate the CD133 gene, which is highly expressed in brain-tumor-initiating cells (BTICs) and used as a marker for separating this cancer stem cell population. In this study, we have carried out microarray analysis in SH-SY5Y cells overexpressing Elk-1-VP16, which has revealed a large number of genes significantly regulated by Elk-1 that function in nervous system development, embryonic development, pluripotency, apoptosis, survival, and proliferation. Among these, we have shown that genes related to pluripotency, such as Sox2, Nanog, and Oct4, were indeed regulated by Elk-1, and in the context of brain tumors, we further showed that Elk-1 overexpression in CD133+ BTIC population results in the upregulation of these genes. When Elk-1 expression is silenced, the expression of these stemness genes is decreased. We propose that Elk-1 is a transcription factor upstream of these genes, regulating the self-renewal of CD133+ BTICs. Full article
(This article belongs to the Special Issue Recent Developments in Cancer Systems Biology)
Show Figures

Figure 1

21 pages, 4914 KiB  
Article
Circulating miR-1246 Targeting UBE2C, TNNI3, TRAIP, UCHL1 Genes and Key Pathways as a Potential Biomarker for Lung Adenocarcinoma: Integrated Biological Network Analysis
by Siyuan Huang, Yong-Kai Wei, Satyavani Kaliamurthi, Yanghui Cao, Asma Sindhoo Nangraj, Xin Sui, Dan Chu, Huan Wang, Dong-Qing Wei, Gilles H. Peslherbe, Gurudeeban Selvaraj and Jiang Shi
J. Pers. Med. 2020, 10(4), 162; https://0-doi-org.brum.beds.ac.uk/10.3390/jpm10040162 - 11 Oct 2020
Cited by 13 | Viewed by 3045
Abstract
Analysis of circulating miRNAs (cmiRNAs) before surgical operation (BSO) and after the surgical operation (ASO) has been informative for lung adenocarcinoma (LUAD) diagnosis, progression, and outcomes of treatment. Thus, we performed a biological network analysis to identify the potential target genes (PTGs) of [...] Read more.
Analysis of circulating miRNAs (cmiRNAs) before surgical operation (BSO) and after the surgical operation (ASO) has been informative for lung adenocarcinoma (LUAD) diagnosis, progression, and outcomes of treatment. Thus, we performed a biological network analysis to identify the potential target genes (PTGs) of the overexpressed cmiRNA signatures from LUAD samples that had undergone surgical therapy. Differential expression (DE) analysis of microarray datasets, including cmiRNAs (GSE137140) and cmRNAs (GSE69732), was conducted using the Limma package. cmiR-1246 was predicted as a significantly upregulated cmiRNA of LUAD samples BSO and ASO. Then, 9802 miR-1246 target genes (TGs) were predicted using 12 TG prediction platforms (MiRWalk, miRDB, and TargetScan). Briefly, 425 highly expressed overlapping miRNA-1246 TGs were observed between the prediction platform and the cmiRNA dataset. ClueGO predicted cell projection morphogenesis, chemosensory behavior, and glycosaminoglycan binding, and the PI3K–Akt signaling pathways were enriched metabolic interactions regulating miRNA-1245 overlapping TGs in LUAD. Using 425 overlapping miR-1246 TGs, a protein–protein interaction network was constructed. Then, 12 PTGs of three different Walktrap modules were identified; among them, ubiquitin-conjugating enzyme E2C (UBE2C), troponin T1(TNNT1), T-cell receptor alpha locus interacting protein (TRAIP), and ubiquitin c-terminal hydrolase L1(UCHL1) were positively correlated with miR-1246, and the high expression of these genes was associated with better overall survival of LUAD. We conclude that PTGs of cmiRNA-1246 and key pathways, namely, ubiquitin-mediated proteolysis, glycosaminoglycan binding, the DNA metabolic process, and the PI3K–Akt–mTOR signaling pathway, the neurotrophin and cardiomyopathy signaling pathway, and the MAPK signaling pathway provide new insights on a noninvasive prognostic biomarker for LUAD. Full article
(This article belongs to the Special Issue Recent Developments in Cancer Systems Biology)
Show Figures

Figure 1

16 pages, 3634 KiB  
Article
Sestrin2 Expression Has Regulatory Properties and Prognostic Value in Lung Cancer
by Hee Sung Chae, Minchan Gil, Subbroto Kumar Saha, Hee Jeung Kwak, Hwan-Woo Park, Balachandar Vellingiri and Ssang-Goo Cho
J. Pers. Med. 2020, 10(3), 109; https://0-doi-org.brum.beds.ac.uk/10.3390/jpm10030109 - 01 Sep 2020
Cited by 15 | Viewed by 3382
Abstract
Lung cancer remains the most dangerous type of cancer despite recent progress in therapeutic modalities. Development of prognostic markers and therapeutic targets is necessary to enhance lung cancer patient survival. Sestrin family genes (Sestrin1, Sestrin2, and Sestrin3) are involved in protecting cells from [...] Read more.
Lung cancer remains the most dangerous type of cancer despite recent progress in therapeutic modalities. Development of prognostic markers and therapeutic targets is necessary to enhance lung cancer patient survival. Sestrin family genes (Sestrin1, Sestrin2, and Sestrin3) are involved in protecting cells from stress. In particular, Sestrin2, which mainly protects cells from oxidative stress and acts as a leucine sensor protein in mammalian target of rapamycin (mTOR) signaling, is thought to affect various cancers in different ways. To investigate the role of Sestrin2 expression in lung cancer cells, we knocked down Sestrin2 in A549, a non-small cell lung cancer cell line; this resulted in reduced cell proliferation, migration, sphere formation, and drug resistance, suggesting that Sestrin2 is closely related to lung cancer progression. We analyzed Sestrin2 expression in human tissue using various bioinformatic databases and confirmed higher expression of Sestrin2 in lung cancer cells than in normal lung cells using Oncomine and the Human Protein Atlas. Moreover, analyses using Prognoscan and KMplotter showed that Sestrin2 expression is negatively correlated with the survival of lung cancer patients in multiple datasets. Co-expressed gene analysis revealed Sestrin2-regulated genes and possible associated pathways. Overall, these data suggest that Sestrin2 expression has prognostic value and that it is a possible therapeutic target in lung cancer. Full article
(This article belongs to the Special Issue Recent Developments in Cancer Systems Biology)
Show Figures

Figure 1

Review

Jump to: Editorial, Research

28 pages, 2153 KiB  
Review
Recent Advances in Integrative Multi-Omics Research in Breast and Ovarian Cancer
by Christen A. Khella, Gaurav A. Mehta, Rushabh N. Mehta and Michael L. Gatza
J. Pers. Med. 2021, 11(2), 149; https://0-doi-org.brum.beds.ac.uk/10.3390/jpm11020149 - 19 Feb 2021
Cited by 14 | Viewed by 4914
Abstract
The underlying molecular heterogeneity of cancer is responsible for the dynamic clinical landscape of this disease. The combination of genomic and proteomic alterations, including both inherited and acquired mutations, promotes tumor diversity and accounts for variable disease progression, therapeutic response, and clinical outcome. [...] Read more.
The underlying molecular heterogeneity of cancer is responsible for the dynamic clinical landscape of this disease. The combination of genomic and proteomic alterations, including both inherited and acquired mutations, promotes tumor diversity and accounts for variable disease progression, therapeutic response, and clinical outcome. Recent advances in high-throughput proteogenomic profiling of tumor samples have resulted in the identification of novel oncogenic drivers, tumor suppressors, and signaling networks; biomarkers for the prediction of drug sensitivity and disease progression; and have contributed to the development of novel and more effective treatment strategies. In this review, we will focus on the impact of historical and recent advances in single platform and integrative proteogenomic studies in breast and ovarian cancer, which constitute two of the most lethal forms of cancer for women, and discuss the molecular similarities of these diseases, the impact of these findings on our understanding of tumor biology as well as the clinical applicability of these discoveries. Full article
(This article belongs to the Special Issue Recent Developments in Cancer Systems Biology)
Show Figures

Figure 1

24 pages, 703 KiB  
Review
Current State of “Omics” Biomarkers in Pancreatic Cancer
by Beste Turanli, Esra Yildirim, Gizem Gulfidan, Kazim Yalcin Arga and Raghu Sinha
J. Pers. Med. 2021, 11(2), 127; https://0-doi-org.brum.beds.ac.uk/10.3390/jpm11020127 - 14 Feb 2021
Cited by 21 | Viewed by 5667
Abstract
Pancreatic cancer is one of the most fatal malignancies and the seventh leading cause of cancer-related deaths related to late diagnosis, poor survival rates, and high incidence of metastasis. Unfortunately, pancreatic cancer is predicted to become the third leading cause of cancer deaths [...] Read more.
Pancreatic cancer is one of the most fatal malignancies and the seventh leading cause of cancer-related deaths related to late diagnosis, poor survival rates, and high incidence of metastasis. Unfortunately, pancreatic cancer is predicted to become the third leading cause of cancer deaths in the future. Therefore, diagnosis at the early stages of pancreatic cancer for initial diagnosis or postoperative recurrence is a great challenge, as well as predicting prognosis precisely in the context of biomarker discovery. From the personalized medicine perspective, the lack of molecular biomarkers for patient selection confines tailored therapy options, including selecting drugs and their doses or even diet. Currently, there is no standardized pancreatic cancer screening strategy using molecular biomarkers, but CA19-9 is the most well known marker for the detection of pancreatic cancer. In contrast, recent innovations in high-throughput techniques have enabled the discovery of specific biomarkers of cancers using genomics, transcriptomics, proteomics, metabolomics, glycomics, and metagenomics. Panels combining CA19-9 with other novel biomarkers from different “omics” levels might represent an ideal strategy for the early detection of pancreatic cancer. The systems biology approach may shed a light on biomarker identification of pancreatic cancer by integrating multi-omics approaches. In this review, we provide background information on the current state of pancreatic cancer biomarkers from multi-omics stages. Furthermore, we conclude this review on how multi-omics data may reveal new biomarkers to be used for personalized medicine in the future. Full article
(This article belongs to the Special Issue Recent Developments in Cancer Systems Biology)
Show Figures

Figure 1

34 pages, 2251 KiB  
Review
Drug Repurposing for Triple-Negative Breast Cancer
by Marta Ávalos-Moreno, Araceli López-Tejada, Jose L. Blaya-Cánovas, Francisca E. Cara-Lupiañez, Adrián González-González, Jose A. Lorente, Pedro Sánchez-Rovira and Sergio Granados-Principal
J. Pers. Med. 2020, 10(4), 200; https://0-doi-org.brum.beds.ac.uk/10.3390/jpm10040200 - 29 Oct 2020
Cited by 26 | Viewed by 6933
Abstract
Triple-negative breast cancer (TNBC) is the most aggressive type of breast cancer which presents a high rate of relapse, metastasis, and mortality. Nowadays, the absence of approved specific targeted therapies to eradicate TNBC remains one of the main challenges in clinical practice. Drug [...] Read more.
Triple-negative breast cancer (TNBC) is the most aggressive type of breast cancer which presents a high rate of relapse, metastasis, and mortality. Nowadays, the absence of approved specific targeted therapies to eradicate TNBC remains one of the main challenges in clinical practice. Drug discovery is a long and costly process that can be dramatically improved by drug repurposing, which identifies new uses for existing drugs, both approved and investigational. Drug repositioning benefits from improvements in computational methods related to chemoinformatics, genomics, and systems biology. To the best of our knowledge, we propose a novel and inclusive classification of those approaches whereby drug repurposing can be achieved in silico: structure-based, transcriptional signatures-based, biological networks-based, and data-mining-based drug repositioning. This review specially emphasizes the most relevant research, both at preclinical and clinical settings, aimed at repurposing pre-existing drugs to treat TNBC on the basis of molecular mechanisms and signaling pathways such as androgen receptor, adrenergic receptor, STAT3, nitric oxide synthase, or AXL. Finally, because of the ability and relevance of cancer stem cells (CSCs) to drive tumor aggressiveness and poor clinical outcome, we also focus on those molecules repurposed to specifically target this cell population to tackle recurrence and metastases associated with the progression of TNBC. Full article
(This article belongs to the Special Issue Recent Developments in Cancer Systems Biology)
Show Figures

Graphical abstract

12 pages, 444 KiB  
Review
Systems Biology and Experimental Model Systems of Cancer
by Gizem Damla Yalcin, Nurseda Danisik, Rana Can Baygin and Ahmet Acar
J. Pers. Med. 2020, 10(4), 180; https://0-doi-org.brum.beds.ac.uk/10.3390/jpm10040180 - 19 Oct 2020
Cited by 15 | Viewed by 4002
Abstract
Over the past decade, we have witnessed an increasing number of large-scale studies that have provided multi-omics data by high-throughput sequencing approaches. This has particularly helped with identifying key (epi)genetic alterations in cancers. Importantly, aberrations that lead to the activation of signaling networks [...] Read more.
Over the past decade, we have witnessed an increasing number of large-scale studies that have provided multi-omics data by high-throughput sequencing approaches. This has particularly helped with identifying key (epi)genetic alterations in cancers. Importantly, aberrations that lead to the activation of signaling networks through the disruption of normal cellular homeostasis is seen both in cancer cells and also in the neighboring tumor microenvironment. Cancer systems biology approaches have enabled the efficient integration of experimental data with computational algorithms and the implementation of actionable targeted therapies, as the exceptions, for the treatment of cancer. Comprehensive multi-omics data obtained through the sequencing of tumor samples and experimental model systems will be important in implementing novel cancer systems biology approaches and increasing their efficacy for tailoring novel personalized treatment modalities in cancer. In this review, we discuss emerging cancer systems biology approaches based on multi-omics data derived from bulk and single-cell genomics studies in addition to existing experimental model systems that play a critical role in understanding (epi)genetic heterogeneity and therapy resistance in cancer. Full article
(This article belongs to the Special Issue Recent Developments in Cancer Systems Biology)
Show Figures

Figure 1

Back to TopTop