scholarly journals TMOD-31. NOVEL GENETIC CHARACTERIZATION OF MURINE MEDULLOBLASTOMA CELL LINES

2020 ◽  
Vol 22 (Supplement_2) ◽  
pp. ii234-ii234
Author(s):  
Sarah Rumler ◽  
Mark Maienschein-Cline ◽  
Melline Fontes Noronha ◽  
Fei Song ◽  
Andrew Lock ◽  
...  

Abstract Pre-clinical research in medulloblastoma (MB) is conducted either with human-derived cell lines or tumors arising in genetically engineered mice (GEM). Murine MB cell lines are not readily available. To begin in vitro immuno-oncology MB research, we identified murine MB cell lines from research laboratories to determine their fidelity to human cell lines and GEM models. Using PubMed, we identified murine MB cell lines of potential interest. Seven cell lines form two independent laboratories were identified. We characterized the cell lines’ behavior in culture and performed next generation sequencing (NGS) on four of them (two from each group) to determine the extent to which these cell lines resemble both human and GEM MB models. DNA and RNA were extracted from MB cell lines, quality and quantity assessed. Whole-exome sequencing was performed and post-capture libraries sequenced. The cell lines identified were derived from either FVB/N TP53 null mice carrying the N-MYC oncogene (GTML3 and GTML5) or C57Bl6/J TP53 null heterozygous ptch (-/+) mice (MM1 and MM3). GTML cells grow in suspension and form clusters. MM cells are adherent, growing in monolayers. Whole exome sequencing of the 4 lines revealed similarities between both GEM and human MB cell lines, demonstrating 25% of sixteen genes known to be affected in both GEM tumors and human MB cell lines. Furthermore, the four cell Iines shared five deleterious mutations previously not described in MB, suggesting they might play a role in MB tumorigenesis: Pde4d, Whrln, Olr156, RIKEN cDNA D630045J12, and Olfr159. These are the first data to describe these murine MB cell lines and demonstrates 1) they resemble both GEM tumors and human MB cell lines at the genome level, and 2) they offer the potential to identify heretofore unrecognized mechanisms in MB tumorigenesis.

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.


2021 ◽  
Vol 21 (1) ◽  
Author(s):  
Zhiwei Wang ◽  
Yinghao Guo ◽  
Yun Jin ◽  
Xiaoxiao Zhang ◽  
Hao Geng ◽  
...  

Abstract Background Patient-derived organoids (PDO) have been proposed as a novel in vitro method of drug screening for different types of cancer. However, to date, extrahepatic biliary tract carcinoma (eBTC) PDOs have not yet been fully established. Methods We collected six samples of gallbladder carcinoma (GBC) and one sample of extrahepatic cholangiocarcinoma (eCCA) from seven patients to attempt to establish eBTC PDOs for drug screening. We successfully established five GBC and one eCCA PDOs. Histological staining was used to compare structural features between the original tissues and cancer PDOs. Whole exome sequencing (WES) was performed to analyze the genetic profiles of original tissues and cancer PDOs. Drug screening, including gemcitabine, 5-fluorouracil, cisplatin, paclitaxel, infigratinib, and ivosidenib, was measured and verified by clinical effects in certain cases. Results Different PDOs exhibited diverse growth rates during in vitro culture. Hematoxylin and eosin staining demonstrated that the structures of most cancer PDOs retained the original structures of adenocarcinoma. Immunohistological and periodic acid-schiff staining revealed that marker expression in cancer PDOs was similar to that of the original specimens. Genetic profiles from the four original specimens, as well as paired cancer PDOs, were analyzed using whole exome sequencing. Three of the four PDOs exhibited a high degree of similarity when compared to the original specimens, except for GBC2 PDO, which only had a concordance of 74% in the proportion of single nucleotide polymorphisms in the coding sequence. In general, gemcitabine was found to be the most efficient drug for eBTC treatment, as it showed moderate or significant inhibitory impact on cancer growth. Results from drug screening were confirmed to a certain extent by three clinical cases. Conclusions Our study successfully established a series of eBTC PDOs, which contributed to the field of eBTC PDOs. Additional enhancements should be explored to improve the growth rate of PDOs and to preserve their immune microenvironment.


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.


BMC Cancer ◽  
2019 ◽  
Vol 19 (1) ◽  
Author(s):  
Pablo Fernández-Navarro ◽  
Pilar López-Nieva ◽  
Elena Piñeiro-Yañez ◽  
Gonzalo Carreño-Tarragona ◽  
Joaquín Martinez-López ◽  
...  

Abstract Background Acute T-cell lymphoblastic leukaemia (T-ALL) is an aggressive disorder derived from immature thymocytes. The variability observed in clinical responses on this type of tumours to treatments, the high toxicity of current protocols and the poor prognosis of patients with relapse or refractory make it urgent to find less toxic and more effective therapies in the context of a personalized medicine of precision. Methods Whole exome sequencing and RNAseq were performed on DNA and RNA respectively, extracted of a bone marrow sample from a patient diagnosed with tumour primary T-ALL and double negative thymocytes from thymus control samples. We used PanDrugs, a computational resource to propose pharmacological therapies based on our experimental results, including lists of variants and genes. We extend the possible therapeutic options for the patient by taking into account multiple genomic events potentially sensitive to a treatment, the context of the pathway and the pharmacological evidence already known by large-scale experiments. Results As a proof-of-principle we used next-generation-sequencing technologies (Whole Exome Sequencing and RNA-Sequencing) in a case of diagnosed Pro-T acute lymphoblastic leukaemia. We identified 689 disease-causing mutations involving 308 genes, as well as multiple fusion transcript variants, alternative splicing, and 6652 genes with at least one principal isoform significantly deregulated. Only 12 genes, with 27 pathogenic gene variants, were among the most frequently mutated ones in this type of lymphoproliferative disorder. Among them, 5 variants detected in CTCF, FBXW7, JAK1, NOTCH1 and WT1 genes have not yet been reported in T-ALL pathogenesis. Conclusions Personalized genomic medicine is a therapeutic approach involving the use of an individual’s information data to tailor drug therapy. Implementing bioinformatics platform PanDrugs enables us to propose a prioritized list of anticancer drugs as the best theoretical therapeutic candidates to treat this patient has been the goal of this article. Of note, most of the proposed drugs are not being yet considered in the clinical practice of this type of cancer opening up the approach of new treatment possibilities.


2017 ◽  
Vol 32 (10) ◽  
pp. 867-870 ◽  
Author(s):  
Hannah Song ◽  
Sina Haeri ◽  
Hannes Vogel ◽  
Marjo van der Knaap ◽  
Keith Van Haren

Objective: We describe 2 male siblings with a severe, prenatal phenotype of vanishing white matter disease and the impact of whole exome sequencing on their diagnosis and clinical care. Methods: The 2 children underwent detailed clinical characterization, through clinical and laboratory testing, as well as prenatal and postnatal imaging. Biobanked blood from the 2 siblings was submitted for whole exome sequencing at Baylor Laboratories. Results: Both male children had abnormal prenatal neuroimaging and suffered precipitous, fatal neurologic decline. Neuropathologic findings included subependymal pseudocysts, microcalcifications, and profound lack of brain myelin and sparing of peripheral nerve myelin. A novel homozygous mutation in the EIF2B3 gene (c.97A>G [p.Lys33Glu]) was found in both children; both parents were heterozygous carriers. The family subsequently conceived a healthy child via in vitro fertilization with preimplantation mutation screening. Conclusion: These histories expand the prenatal phenotype of eIF2b-related disorders and poignantly illustrate the impact that unbiased genomic sequencing can have on the diagnosis and medical decision making for families affected by childhood neurodegenerative disorders.


2020 ◽  
Vol 4 (12) ◽  
Author(s):  
Stephen I Stone ◽  
Daniel J Wegner ◽  
Jennifer A Wambach ◽  
F Sessions Cole ◽  
Fumihiko Urano ◽  
...  

Abstract Insulin-mediated pseudoacromegaly (IMPA) is a rare disease of unknown etiology. Here we report a 12-year-old female with acanthosis nigricans, hirsutism, and acromegalic features characteristic of IMPA. The subject was noted to have normal growth hormone secretion, with extremely elevated insulin levels. Studies were undertaken to determine a potential genetic etiology for IMPA. The proband and her family members underwent whole exome sequencing. Functional studies were undertaken to validate the pathogenicity of candidate variant alleles. Whole exome sequencing identified monoallelic, predicted deleterious variants in genes that mediate fibroblast growth factor 21 (FGF21) signaling, FGFR1 and KLB, which were inherited in trans from each parent. FGF21 has multiple metabolic functions but no known role in human insulin resistance syndromes. Analysis of the function of the FGFR1 and KLB variants in vitro showed greatly attenuated ERK phosphorylation in response to FGF21, but not FGF2, suggesting that these variants act synergistically to inhibit endocrine FGF21 signaling but not canonical FGF2 signaling. Therefore, digenic variants in FGFR1 and KLB provide a potential explanation for the subject’s severe insulin resistance and may represent a novel category of insulin resistance syndromes related to FGF21.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 69-69
Author(s):  
Michelle Perugini ◽  
Saumya E Samaraweera ◽  
Anna L Brown ◽  
Nik Cummings ◽  
Silke Danner ◽  
...  

Abstract BACKGROUND: We have previously shown that one target of hyper-methylation in AML is the promoter of the tumour suppressor and stress-response mediator Growth Arrest and DNA Damage inducible 45A (GADD45A) (GADD45AmeHI; 42% of AML). In mice Gadd45a has recently been shown to play a critical role in HSC stress responses. Gadd45a deficiency leads to enhanced HSC self-renewal, DNA damage accumulation in HSC, increased susceptibility to leukemogenesis, and impairment in HSC apoptosis after genotoxic exposure (Chen et al, Blood 2014). These findings suggest that hypermethylation of the GADD45A gene may play an important role in the altered properties of HSC, leukaemic initiation and progression. Promoter hypermethylation of this gene defines a patient group with poor survival on standard therapy (Perugini et al, Leukaemia 2012). To explore further the molecular basis of the GADD45AmeHI patient group weperformed genetic profiling of diagnosis samples using a Sequenom multiplex mutation panel, or using whole exome sequencing for broader coverage (n=95 patients).Sequenom MassARRAY was used for quantitative detection of GADD45A promoter methylation in patient samples. For a cohort of matched diagnosis and relapse samples we used CpG methylation data for GADD45A determined by ERRBS (Akalin et al, PLoSGenetics 2012). Response to cytotoxic drugs and assessment of drug combinations with 5-Aza-deoxycytidine (decitabine, DAC) and anthracycline (Daunorubicin, DNR) was performed in AML cell lines, and with primary leukemic cell populations. RESULTS: The association of the GADD45AmeHI patient group with poor outcome was validated in an independent AML patient cohort of 48 patients from the Alfred Hospital, Melbourne, Australia (p=0.003; HR3.35). Whole exome sequencing and Sequenom multiplex analysis of 95 AML patients revealed a striking co-occurrence of the GADD45AmeHI phenotype with mutations in IDH1, IDH2, and TET2 (p<0.0001, Fisher’s exact test, Fig. 1). To test the prediction that GADD45A hypermethylation may be an important factor for relapse we investigated GADD45A promoter DNA methylation levels in paired diagnosis and relapse samples. In a paired analysis of 39 patients we show that relapse samples display a significant increase in GADD45A promoter CpG methylation (p=0.035, paired t-test). This is consistent with emergence in many patients following chemotherapy of a chemoresistant clone that has increased GADD45A methylation and reduced GADD45A activity. We next tested whether reactivation of GADD45A expression in GADD45AmeHI patient samples could be achieved through the use of hypo-methylation agents, and whether this is beneficial for response to chemotherapy. DAC treatment has been reported to induce DNA demethylation and GADD45A reactivation in primary AML samples (Klco et al, Blood 2013), and we observe reduced GADD45A promoter methylation and increased expression following DAC treatment of the GADD45AmeHI AML cell line (Mv4;11), consistent with DNA methylation-induced gene silencing of GADD45A. DAC pre-treatment of the GADD45AmeHI AML cell lines MOLM13 and Mv4;11, and three primary AML samples (GADD45AmeHI), resulted in increased GADD45A expression and increased DNR sensitivity. CONCLUSIONS: DNA methylation of the GADD45A proximal promoter marks a large percentage of AML patients at diagnosis including the majority of those with IDH1/2 and TET2 mutations (collectively these occur in 28% of AML (Network CGAR, N Engl J Med, 2013)), and is an independent predictor of poor outcome in two independent patient cohorts. Our data shows that silencing of GADD45A through increased promoter CpG methylation maybe an important early event in leukaemogenesis associated with impaired TET2 activity. Based on recent studies describing the properties of Gadd45a-deficient murine HSC we suggest reduced GADD45A activity in this subset of patients may contribute to the properties of pre-leukaemic HSC that have been associated with IDH1/2 mutation and reported to display clonal expansion, resistance to chemotherapy, and ultimately a high risk of relapse. In vitro drug experiments suggest that a priming schedule of DAC followed by DNR may provide a successful tailored treatment strategy for GADD45AmeHI patients, in combination with GADD45A expression as a biomarker predicting increased DNR sensitivity. Fig 1: Co-association of GADD45AmeHI with IDH1/2 and TET2 mutations in 95 AML patients Fig 1:. Co-association of GADD45AmeHI with IDH1/2 and TET2 mutations in 95 AML patients Disclosures No relevant conflicts of interest to declare.


2015 ◽  
Author(s):  
Mirna Jarosz ◽  
Michael Schnall-Levin ◽  
Grace X. Y. Zheng ◽  
Patrick Marks ◽  
Sofia Kyriazopoulou-Panagiotopoulou ◽  
...  

2019 ◽  
Vol 18 (8) ◽  
pp. 1460-1471 ◽  
Author(s):  
Sunetra Das ◽  
Rupa Idate ◽  
Kathryn E. Cronise ◽  
Daniel L. Gustafson ◽  
Dawn L. Duval

2019 ◽  
Vol 21 (Supplement_3) ◽  
pp. iii54-iii54
Author(s):  
C Verheul ◽  
T V Kers ◽  
A Van Der Ploeg ◽  
M Van Der Kaaij ◽  
M Aghababazadeh ◽  
...  

Abstract BACKGROUND Despite considerable scientific efforts, endogenous in vitro isocitrate dehydrogenase (IDH)-mutated glioma models remain scarce. Availability of these models is key to understanding underlying molecular mechanisms and vital for the development of new therapeutic interventions. We established and characterized a set of seven cell lines derived from IDH1-mutated gliomas and utilized them to investigate IDH-mutant glioma drug-response in vitro. MATERIAL AND METHODS Fresh tumor material was collected directly from the operating room, mechanically and enzymatically dissociated and cultured under serum-free culture conditions. IDH mutation status was verified at several passages with Sanger sequencing, as well as with whole exome sequencing. D-2-hydroxyglutarate (D2-HG) levels were determined with mass-spectrometry. Genome-wide methylation profiling was performed using the Infinium MethylationEpic BeadChip array. Cell viability was measured using the ATP-based CellTiter-Glo assay. Cell proliferation was determined through cell counting. Drug screens were performed with an FDA-approved anti-cancer drug set from the NIH as well as two IDH-mutant specific inhibitors. RESULTS Over the last decade our lab processed over 800 glioma samples from all WHO grades and IDH mutational status. Optimisation of culture conditions led to the establishment of seven IDH1-mutant glioma cell lines. The IDH1-mutation was present during all tested passages in these cell lines. Whole exome sequencing showed a high concordance between tumor and cell lines with regard to driver gene mutations. Similarly, copy-number changes based on genome-wide methylation data also show a close resemblance between the parental tumor and resulting cell lines. The Heidelberg methylation profiler classified each cell line and its parental tumor as IDH mutant glioma. When exposed to an IDH1-mutant specific inhibitor, all cell lines showed a concentration-dependent decrease of D2-HG levels. The inhibitors showed little to no effect on viability up to 10uM during 8 days, however, long term treatment up to 4 weeks revealed a decrease in cell doubling time in 2 of 6 cultures. Drug screen results either alone or in combination with an IDH1-mutant specific inhibitor identified several interesting candidates that are currently followed up in additional experiments. CONCLUSION We established a unique set of patient-derived IDH1-mutant glioma cell lines that closely resemble their respective tumors and can be used to identify new therapeutic strategies.


Sign in / Sign up

Export Citation Format

Share Document