scholarly journals Identifying Candidate Druggable Targets in Canine Cancer Cell Lines Using Whole-Exome Sequencing

2019 ◽  
Vol 18 (8) ◽  
pp. 1460-1471 ◽  
Author(s):  
Sunetra Das ◽  
Rupa Idate ◽  
Kathryn E. Cronise ◽  
Daniel L. Gustafson ◽  
Dawn L. Duval
2015 ◽  
Author(s):  
Mirna Jarosz ◽  
Michael Schnall-Levin ◽  
Grace X. Y. Zheng ◽  
Patrick Marks ◽  
Sofia Kyriazopoulou-Panagiotopoulou ◽  
...  

Author(s):  
Chi Song ◽  
Shih-Chi Su ◽  
Zhiguang Huo ◽  
Suleyman Vural ◽  
James E Galvin ◽  
...  

Abstract Summary In this article, we introduce a hierarchical clustering and Gaussian mixture model with expectation-maximization (EM) algorithm for detecting copy number variants (CNVs) using whole exome sequencing (WES) data. The R shiny package ‘HCMMCNVs’ is also developed for processing user-provided bam files, running CNVs detection algorithm and conducting visualization. Through applying our approach to 325 cancer cell lines in 22 tumor types from Cancer Cell Line Encyclopedia (CCLE), we show that our algorithm is competitive with other existing methods and feasible in using multiple cancer cell lines for CNVs estimation. In addition, by applying our approach to WES data of 120 oral squamous cell carcinoma (OSCC) samples, our algorithm, using the tumor sample only, exhibits more power in detecting CNVs as compared with the methods using both tumors and matched normal counterparts. Availability and implementation HCMMCNVs R shiny software is freely available at github repository https://github.com/lunching/HCMM_CNVs.and Zenodo https://doi.org/10.5281/zenodo.4593371. Supplementary information Supplementary data are available at Bioinformatics online.


PLoS ONE ◽  
2011 ◽  
Vol 6 (6) ◽  
pp. e21097 ◽  
Author(s):  
Han Chang ◽  
Donald G. Jackson ◽  
Paul S. Kayne ◽  
Petra B. Ross-Macdonald ◽  
Rolf-Peter Ryseck ◽  
...  

2019 ◽  
Vol 17 (2) ◽  
pp. 119-129
Author(s):  
Tatsuya Deguchi ◽  
Kenji Hosoya ◽  
Yusuke Murase ◽  
Sung Koangyong ◽  
Sangho Kim ◽  
...  

2008 ◽  
Vol 69 (7) ◽  
pp. 938-945 ◽  
Author(s):  
William C. Kisseberth ◽  
Sridhar Murahari ◽  
Cheryl A. London ◽  
Samuel K. Kulp ◽  
Ching-Shih Chen

Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 4449-4449
Author(s):  
James W Murray ◽  
Christopher Fegan ◽  
Chris Pepper

Abstract Background: Understanding the pathology of Multiple Myeloma and the testing of therapeutic options has relied heavily on isogeneic cell lines due to the inability to sustain myeloma plasma cells in long-term in vitro culture. The cell lines MM.1S and MM.1R are well recognised in the field of myeloma research, providing a model of glucocorticoid drug resistance, primarily believed to be through variable expression of the glucocorticoid receptor NR3C1 but here we found no evidence of a genetic basis for this. Here we set out to examine the phenotype, function and genotype of the MM.1S and MM.1R cell lines in order to explore the origins of glucocorticoid drug resistance manifested by MM.1R cells and establish whether exome analysis could identify sub-clones with preferential sensitivity to molecular targeted inhibitors. Methods: MM.1S and MM.1R cell lines were purchased from ATCC. A 10-colour flow cytometry panel (CD38, CD138, CD19, CD45, CD56, CD49d, CXCR4, MMP-9, Ki-67, IL-6) was analysed on a BD LSR Fortessa flow cytometer and MM.1S subsets were sorted using a FACS Aria III. Telomere length was assessed using Single Telomere Length Analysis (STELA) and drug toxicity assays using Annexin V-FITC/PI staining. Bioinformatics of whole exome sequencing was carried out on the GATK platform and gene list analysis using Enrichr. PI3K isoforms were analysed by quantitative PCR and immunoblotting. Results: The MM.1S cell line demonstrated bimodal CD38 expression, with a 1.5 log difference in CD38 expression (p<0.0001) between the two populations (termed MM.1Sdim and MM.1Sbright). In contrast the MM.1R cell line was uniformly CD38bright, with expression a further 0.5 log higher than MM.1Sbright. When cell sorted subsets of MM.1S cells were subjected to increasing concentrations of Dexamethasone, the MM.1Sbright cells had a significantly higher LC50 than the MM.1Sdim cells (62nM v 29nM respectively; p<0.0001). In contrast, these subsets showed no significant difference in sensitivity to bortezomib (p=0.84. Furthermore, MM.1Sbright cells had a shorter doubling time than both MM.1Sdim (p=0.0001) and MM.1R (p=0.048). This was underscored by an increased proportion of MM.1Sbright cells in S-phase coupled with shorter mean telomere length when compared with MM.1Sdim and MM.1R (2.58 Kb v 3.29Kb v 3.2Kb respectively). We next subjected purified MM.1Sbright, MM.1Sdim and MM.1R cells to whole exome sequencing. The common clonal origin of the three cell lines was evident from the analysis but each line possessed unique genetic lesions. For example, MM.1Sbright had a FIP1L1-PDGFRA fusion mutation that was not present in the MM.1Sdim cells. This was associated with increased expression of the p110d isoform in MM.1Sdim cells. We therefore analysed the effects of the PI3Kd inhibitor, Idelalisib, on the two cell lines and showed that MM.1Sdim cells were more sensitive (p=0.003) to the effects of this agent. The specific nature of this response was confirmed by the fact that the pan p13K inhibitor PKI-402, was equipotent in both MM.1Sbright and MM.1Sdim cells (p=0.89). Conclusion: Analysis of two phenotypically distinct subsets within the MM.1S cell line revealed differences in function and genetics thereby confirming the sub-clonal architecture within this cell line. Intriguingly, our data point to the pre-existence of a dexamethasone resistant sub-clone the MM.1Sbright (CD38+) population. The subsequent production of the dexamethasone resistant cell line (MM.1R) allowed us to perform comparative genomics thereby identifying the genetic origins of dexamethasone resistance (selection) in MM.1Sbright cells and to track the subsequent clonal evolution (induction) in the MM.1R cells. Furthermore, we showed the potential for developing bespoke treatment plans based on the identification of cell signalling pathway mutations via genomic sequencing. By selective targeting of of these genetic lesions it may be possible to remove multiple sub-clones thereby diminishing the potential for clonal tiding and the development of drug resistance. In theory this could result in longer time to relapse and ultimately improved overall survival. Disclosures Fegan: Roche: Honoraria; Gilead Sciences: Honoraria; AbbVie: Honoraria.


Author(s):  
Han Chang ◽  
Donald G. Jackson ◽  
Paul S. Kayne ◽  
Petra B. Ross-Macdonald ◽  
Rolf-Peter Ryseck ◽  
...  

2019 ◽  
Vol 116 (45) ◽  
pp. 22730-22736 ◽  
Author(s):  
Luca Zammataro ◽  
Salvatore Lopez ◽  
Stefania Bellone ◽  
Francesca Pettinella ◽  
Elena Bonazzoli ◽  
...  

The prognosis of advanced/recurrent cervical cancer patients remains poor. We analyzed 54 fresh-frozen and 15 primary cervical cancer cell lines, along with matched-normal DNA, by whole-exome sequencing (WES), most of which harboring Human-Papillomavirus-type-16/18. We found recurrent somatic missense mutations in 22 genes (including PIK3CA, ERBB2, and GNAS) and a widespread APOBEC cytidine deaminase mutagenesis pattern (TCW motif) in both adenocarcinoma (ACC) and squamous cell carcinomas (SCCs). Somatic copy number variants (CNVs) identified 12 copy number gains and 40 losses, occurring more often than expected by chance, with the most frequent events in pathways similar to those found from analysis of single nucleotide variants (SNVs), including the ERBB2/PI3K/AKT/mTOR, apoptosis, chromatin remodeling, and cell cycle. To validate specific SNVs as targets, we took advantage of primary cervical tumor cell lines and xenografts to preclinically evaluate the activity of pan-HER (afatinib and neratinib) and PIK3CA (copanlisib) inhibitors, alone and in combination, against tumors harboring alterations in the ERBB2/PI3K/AKT/mTOR pathway (71%). Tumors harboring ERBB2 (5.8%) domain mutations were significantly more sensitive to single agents afatinib or neratinib when compared to wild-type tumors in preclinical in vitro and in vivo models (P = 0.001). In contrast, pan-HER and PIK3CA inhibitors demonstrated limited in vitro activity and were only transiently effective in controlling in vivo growth of PIK3CA-mutated cervical cancer xenografts. Importantly, combinations of copanlisib and neratinib were highly synergistic, inducing long-lasting regression of tumors harboring alterations in the ERBB2/PI3K/AKT/mTOR pathway. These findings define the genetic landscape of cervical cancer, suggesting that a large subset of cervical tumors might benefit from existing ERBB2/PIK3CA/AKT/mTOR-targeted drugs.


Sign in / Sign up

Export Citation Format

Share Document