scholarly journals High Mir-17-92 Expression Is Associated with in Vitro chemoresistance in Burkitt Lymphoma

Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 1586-1586
Author(s):  
Thomas Ippolito ◽  
Rodney R. Miles ◽  
Vladimir Rodic ◽  
Cory Mavis ◽  
Francisco J. Hernandez-Ilizaliturri ◽  
...  

Abstract Background: Copy number gain and/or MYC induction leads to increased expression of miRs in the miR-17~92 cluster. Development of Burkitt lymphoma (BL) is associated with translocation of MYC resulting in MYC expression and frequent recurrent copy number gains of chromosome 13q containing the miR-17~92 cluster of miRs. MiRs in the miR-17-92 cluster, including miR19 and miR17, have been associated with B-cell lymphomagenesis. Copy number gain of 13q and high expression of miR-17 have also been associated with possible inferior outcome in children with BL. MiR-17~92 miRs can decrease expression of the PI3K inhibiting protein PTEN leading to activation of the PI3K/AKT pathway, a pathway implicated in BL lymphomagenesis. These miRs also target the pro-apoptotic protein BIM, suppression of which has also been associated with chemoresistance in BL. Objectives: Investigate the in vitro effect of altered miR-17-92 expression on chemoresponsiveness in BL cells. Methods: MiR expression was determined by qPCR. The miR-17-92 locus was deleted in Raji cells using custom CRISPR-Cas9 lentiviral vectors. Single cell-derived clones, R5 and R24, were established and locus deletion determined by genomic DNA PCR. Protein expression and Caspase cleavage was assessed by western blotting. Apoptosis induction was determined by flow cytometry for Annexin V-propidium iodide staining. Cell viability following exposure to chemotherapy was determined by AlamarBlue assay. SCID mice were inoculated with Raji or R24 cells by tail vein injection and survival determined by Kaplan-Meier analysis. Hind limb paralysis was used as an end-point for survival. Antagomir targeting miR17 was transduced into Raji cells by electroporation. Results: Chemotherapy resistant Raji 4RH cells exhibit increased miR17 and miR19 expression and increased PI3K/AKT pathway activation compared to parental Raji. R5 and R24 Raji KO lines exhibit decreased miR17 and miR19 expression and increased expression of miR targets PTEN and BIM. When xenografted into SCID mice, R24 cells demonstrated a significant prolongation in survival compared to Raji (Raji vs R24: median 29 days vs not reached at 150 days, P<0.05) (Figure 1B). A significant decrease in [IC50] in R24 cells compared to Raji was observed following 48h exposure to doxorubicin (Raji vs. R24: 65±23nM vs 12±2nM), dexamethasone (1579±10e5nM vs 49±57nM), etoposide (571±106nM vs 106±22nM) or cisplatin (2.5±0.7uM vs 0.96±0.3uM) (Figure 1A). Raji R24 cells also demonstrate an increase in apoptosis induction following exposure to doxorubicin or etoposide. To investigate the effect of direct miR targeting, Raji cells were transfected with an antagomir targeting miR17 resulting in decreased viability following exposure to doxorubicin or cisplatin compared to Raji cells transfected with a control oligomer (Figure 1C). Antagomir transfected Raji cells express higher levels of PTEN and BIM compared to control Raji cells. Conclusion: High expression of miR17-92 cluster miRs is associated with in vitro chemotherapy resistance. Knockout of the MIR17HG gene in BL cells results in impaired proliferation, impaired in vivo engraftement with prolonged survival in SCID mice and increased in vitro chemoresponsiveness associated with an increase in expression of PTEN and BIM. Targeting of miR17 using an antagomir approach resulted in increased PTEN and BIM and increased chemoresponsiveness. These findings highlight the role of MYC-associated miRs in in vitro chemoresistance in BL cells warranting continued investigation as a possible therapeutic target for relapsed/refractory BL. Disclosures No relevant conflicts of interest to declare.

2018 ◽  
Vol 128 (5) ◽  
pp. 1428-1437 ◽  
Author(s):  
Ryohei Otani ◽  
Akitake Mukasa ◽  
Masahiro Shin ◽  
Mayu Omata ◽  
Shunsaku Takayanagi ◽  
...  

OBJECTIVEChordoma is a slow-growing but clinically malignant tumor, and the prognosis remains poor in many cases. There is a strong impetus to develop more effective targeted molecular therapies. On this basis, the authors investigated the potential of Brachyury, a transcription factor involved in notochord development, as a candidate molecular target for the treatment of chordoma.METHODSBrachyury gene copy number and expression levels were evaluated by quantitative polymerase chain reaction in 27 chordoma samples, and the transcriptomes of Brachyury high-expression tumors (n = 4) and Brachyury low-expression tumors (n = 4) were analyzed. A chordoma cell line (U-CH2) was used to investigate the signaling pathways that regulate Brachyury expression.RESULTSAll chordoma specimens expressed Brachyury, and expression levels varied widely. Patients with higher Brachyury expression had significantly shorter progression-free survival (5 months, n = 11) than those with lower expression (13 months, n = 16) (p = 0.03). Somatic copy number gain was confirmed in 12 of 27 (44%) cases, and copy number was positively correlated with Brachyury expression (R = 0.61, p < 0.001). Expression of PI3K/Akt pathway genes was upregulated in Brachyury high-expression tumors, and suppression of PI3K signaling led to reduced Brachyury expression and inhibition of cell growth in the U-CH2 chordoma cell line.CONCLUSIONSActivation of the PI3K/Akt pathway and Brachyury copy number gain are strongly associated with Brachyury overexpression, which appears to be a key event in chordoma growth regulation. These findings suggest that targeting Brachyury and PI3K/Akt signaling may be an effective new approach for treating chordoma.


Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 1922-1922
Author(s):  
Joost Kluiver ◽  
Eugenia Haralambieva ◽  
Debora de Jong ◽  
Tjasso Blokzijl ◽  
Susan Jacobs ◽  
...  

Abstract We previously demonstrated a high expression of primary-microRNA BIC (pri-miRNA-155) in Hodgkin lymphoma (HL) and lack of expression in most non-Hodgkin lymphoma subtypes including some Burkitt lymphoma (BL) cases. Recently, a high expression of BIC was reported in BL in comparison to pediatric leukemia and normal peripheral blood samples. In this study we extended our series of BL cases and cell lines for BIC expression by RNA in-situ hybridization (ISH) and quantitative (q)RT-PCR. Both BIC RNA-ISH and qRT-PCR revealed no or only low levels of BIC in 25 BL tissues, including 7 Epstein-Barr virus (EBV) positive cases, compared to HL and normal controls. In agreement with these findings, Northern blotting revealed absence of miR-155 in BL tissues. EBV negative and EBV latency type I BL cell lines also showed very low BIC and miR-155 expression levels as compared to HL cell lines. Higher levels of BIC and miR-155 were detected in in vitro transformed lymphoblastoid EBV latency type III BL cell lines. An association of latency type III infection and induction of BIC was supported by consistent expression of BIC in 11 and miR-155 in 2 posttransplantation lymphoproliferative disorder (PTLD) cases. In summary, we demonstrated that expression of BIC and miR-155 is not a common finding in BL. Expression of BIC and miR-155 in 3 latency type III EBV positive BL cell lines and in all primary PTLD cases suggests a possible role for EBV latency type III specific proteins in the induction of BIC expression.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 5240-5240
Author(s):  
Xue Wu ◽  
Fei Wang ◽  
Guohua Xia ◽  
Yanping Chen ◽  
Baoan Chen

Abstract Objective: In this study, we investigated the influence of adriamycin and wogonin to the expression of miR-155 in Burrkit lymphoma in vitro. We focused on exploring the efficiency and pathway of wogonin to induce the apoptosis of lymphoma cells through the transcription and translation level of miR-155 compared with ADM and seeking for the evidence of the synergistic effect between wogonin and ADM. Methods: (1) Use drugs to interfere the growth of the Raji cells (Adriamycin (ADM, chemical name:doxorubicin), wogonin, ADM+wogonin), observe the cells through the microscope. (2) The apoptosis of Raji cells was tested by FCM. (3) The transcription of miRNA-155 of Raji cells was tested by real-time PCR. (4) The translation of miRNA-155 of Raji cells was tested by Western blot. Results: (5) After being disposed for 24 hours,the experimental groups developed to apoptosis.The statistic significance existed between groups of experiment and control and groups of each experiment.The effect of inducing apoptosis from strong to weak was group of ADM+wogonin,ADM and wogonin. (6) After being disposed for 24 hours,the transcription level of miRNA-155 of the experimental groups was lower than the control group.Compared with ADM,the transcription level of miRNA-155 of wogonin group seemed to be stronger,while the combination of the two drugs was the weakest. (7) After being disposed for 24 hours,the translation level of p85α of the experimental groups was lower than the control group. Compared with ADM, the translation level of p85α of wogonin seemed to be stronger, while the combination of the two drugs was the weakest. Conclusion: The Chinese materia medica preparation wogonin could induce the apoptosis of lymphoma cells and enhance the effect of ADM through PI3K/AKT pathway. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 3916-3916 ◽  
Author(s):  
Karl-Heinz Heider ◽  
Elinborg Ostermann ◽  
Herbert Lamche ◽  
Zaruhi Kuepcue ◽  
Alexander Jacobi ◽  
...  

Abstract Abstract 3916 Introduction: Treatment of B-cell malignancies with rituximab, an antibody specific for the B-cell antigen CD20, in combination with chemotherapy has been established as standard clinical practice during recent years. Despite the impressive clinical benefit of rituximab-chemotherapy combination treatment, there remains a substantial need for improved treatment modalities. Here we describe two novel Fc-engineered antibodies to CD37, mAb 37.1 and mAb 37.2, which mediate potent pro-apoptotic and ADCC activity against malignant B-cells. The tetraspanin CD37 is predominantly expressed on the cell surface of normal and malignant B-cells and therefore qualifies as target for antibody mediated tumor therapy. Methods: MAb 37.1 is an Fc-engineered, mouse-human chimeric IgG1-type of antibody which binds CD37 with low nanomolar affinity as determined by FACS scatchard analysis. MAb 37.2 is a humanized version derived from mAb 37.1. Affinity to Fc-receptors was determined by BIAcore analysis. ADCC and pro-apoptotic activity was determined using Ramos Burkitt lymphoma cells and primary CLL cells. ADCC assays were performed using PBMCs from healthy donors as effector cells. Apoptosis induction was determined by AnnexinV/PI staining. The effect in blood from healthy individuals was assessed in whole blood assays ex vivo. In vivo anti-tumor efficacy was assessed in a Ramos xenograft model in immuno-compromised mice. Results: Both CD37 antibodies are able to induce apoptosis of Ramos Burkitt lymphoma cells and primary CLL cells in vitro. Direct comparison of mAb 37.1 to Rituximab demonstrated superior apoptosis induction both on Ramos and primary CLL cells. The pro-apoptotic activity of mAb 37.1 is not dependent on cross-linking of the antibody, representing a novel mode of CD37-specific apoptosis induction. Phenotypic analysis of mAb 37.1 treated Ramos cells revealed early aggregation of tumor cells resembling homotypic aggregation. In order to further improve the cytotoxic potential of the antibodies an Fc-engineering approach was chosen to enhance binding to FcgRIIIa. By applying a two amino acid substitution in the Fc-region of the antibody the affinities of mAb 37.1 and mAb 37.2 to human FcgRIIIa was increased by 40 to 80-fold, depending on the FcgRIIIa allotype, as compared to the parental antibody. This increase directly translates into a strongly enhanced ADCC activity compared to the parental antibody as detected in an in vitro ADCC assay with Ramos and primary CLL cells, resulting in superior ADCC activity compared to Rituximab. The potent B-cell specific depleting activity of mAb 37.1 and mAb 37.2 was confirmed with naïve B-cells and spiked Ramos Burkitt lymphoma cells in vitro in whole blood conditions, whereas no significant effects on T-cells and monocytes were observed. Rituximab, which was used as comparator antibody in parallel, showed no significant reduction of viable B-cells in this assay format. Finally, the in vivo anti-tumor effect of CD37 mAbs was assessed using a Ramos xenograft. Treatment of established subcutaneous tumors with doses ranging from 2.5 to 25 mg/kg in a twice weekly treatment schedule resulted in tumor growth inhibition up to 100%. Conclusion: MAb 37.1 and mAb 37.2 represent novel, Fc-engineered antibodies specific for CD37 with potent pro-apoptotic and enhanced ADCC activity against lymphoma cell lines and primary CLL cells. Based on these data mAb 37.1 and mAb 37.2 are considered as promising candidates for treatment of patients with CD37-positive B-cell malignancies. Disclosures: Heider: Boehringer Ingelheim: Employment. Ostermann:Boehringer Ingelheim: Employment. Lamche:Boehringer Ingelheim: Employment. Kuepcue:Boehringer Ingelheim: Employment. Jacobi:Boehringer Ingelheim: Employment. Konopitzky:Boehringer Ingelheim: Employment. Hirt:Boehringer Ingelheim: Employment. Reichardt:Boehringer Ingelheim: Employment. Borges:Boehringer Ingelheim: Employment.


Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 3487-3487
Author(s):  
Qian Xu ◽  
Zheng Zhang ◽  
Lei Zhao ◽  
Yun Qin ◽  
Yicheng Zhang ◽  
...  

Abstract Introduction CRISPR/Cas9 system is a highly efficient genomic editing system. However, application of CRISPR/Cas9 system in vivo is somewhat limited for delivery methods and safety concerns. Extracellular vesicles (EVs) are released by exocytosis which results in consistent surface membrane proteins with original cells and capable to deliver several kinds of materials in vivo. However, EVs can be absorbed by other organs or tissues besides tumors, leading to possible off-target effects. Therefore, it is imperative to enhance the targeting property of EVs in vivo. Recently, clinical advances have confirmed that CAR-T cells possess great tropism and multiple choices of target. This may be an effective means to improve EVs' selective accumulation in tumors. In this proof-of-principle study, we exploit the target precision of CAR to establish an EV-derived tropism-delivery platform for CRISPR/Cas9 system as a novel therapeutic strategy for malignant diseases. Methods We established CD19-CAR-293T cell line by CAR lentivirus transfection. Raji cells (Burkitt lymphoma) were chosen as target tumor cells for it's CD19 positive. For the isolation of EVs from CD19-CAR-293T and 293T cells, culture medium was harvested, centrifuged and filtered through 0.45μm filters to remove the dead cells and large microvesicles. Purified EVs were examined by (NTA). The plasmids of CRISPR/Cas9 system targeting human cMyc gene were constructed by standard protocols and electroporated into EVs using Gene Pulser Xcell Electroporation System (BioRad). EVs were labelled with lipophilic fluorescent dye for visualization of confocal images. Uptake of EVs in Raji cells was analyzed using confocal microscope. To observe the biodistribution of EVs, Raji-bearing xenograft NOD/SCID mice were generated. When the tumors reached 200mm3, labelled EVs were intracardially injected to mice. Biofluorescence images were obtained using IVIS imaging system. Results NTA showed that a size distribution of approximately 100nm to 450nm for two types of EVs (Fig. 1A). The modifications didn't affect the physical properties. We used a fixed number ratio of Raji cells to EVs in incubation to compare the uptake efficiency between normal EVs and CD19-CAR-EVs. After one hour of incubation with Raji cells, uptake efficiency of the CD19-CAR-EVs was higher than that of the normal EVs, as indicated by signal intensity in the confocal microscope images (Fig. 1B). To confirm whether CD19-CAR-EVs could target tumors in vivo, we intracardially injected the same number of labelled EVs or CD19-CAR-EVs to Raji xenograft NOD/SCID mice and observed at 0h and 24h post-injection. As results, the CD19-CAR-EVs significantly accumulated in tumor sites compared to liver, kidney and other tissues. In contrast, the normal EVs were distributed throughout the body (Fig. 1C). Ex vivo images exhibited that compared with the EV group, the CD19-CAR-EVs accumulated more in tumors and less in liver, kidney and other tissues (Fig. 1C). The tumor/whole-body fluorescence intensity ratios were calculated in the EV and CD19-CAR-EV groups (Fig. 1D). At 24h post-injected, the ratios in the CD19-CAR-EV group were higher than that in EV group (P<0.05). Twelve days after the subcutaneous xenograft models were generated, CD19-CAR-EVs loaded with CRISPR/Cas9 system targeting human cMyc were injected intracardially or intratumorally (i.t.) to mice with interval of 4 days for 5 times. Mice injected with EVs only, with plasmid of cMyc sgRNA/Cas9 only, and with Cas9 loaded CD19-CAR-EVs without sgRNA were used as control groups. Tumor sizes were compared among groups at 18 days post the first EVs injection. Tumors from intracardially injected CD19-CAR-EVs+cMyc sgRNA/Cas9 group were smaller than those from EVs+cMyc sgRNA/Cas9 group. And the tumors from intratumoral injection of CD19-CAR-EVs+cMyc sgRNA/Cas9 were the smallest ones among groups (Fig. 1E). Conclusion In this study, we take advantages of CAR's affinity to tumor cells and EVs' delivery capacity, and generated CD19-CAR-EVs capable of delivering CRISPR/Cas9 system. This CAR-EV-derived tropism-delivery system can be administered systemically or locally and accumulated selectively in tumor tissues to achieve gene editing in vivo. With evolving repertoires of CARs and multiplex genome engineering abilities of CRISPR/Cas9, this approach is with great potential for biotechnological and therapeutic applications. Disclosures No relevant conflicts of interest to declare.


2018 ◽  
Vol 7 (11) ◽  
pp. 408 ◽  
Author(s):  
Chien-Feng Li ◽  
Ting-Ting Liu ◽  
Jui-Chu Wang ◽  
Shih-Chen Yu ◽  
Yen-Yang Chen ◽  
...  

The lipid-metabolizing enzymes remain underexplored in gastrointestinal stromal tumors (GISTs). Through transcriptomic reappraisal, hydroxysteroid 11-beta dehydrogenase-1 (HSD11B1) was identified as a top-upregulated, progression-associated gene. To validate the clinical relevance of HSD11B1, the informative results of Sanger sequencing (n = 58), mRNA quantification by branched-chain DNA in situ hybridization assay (n = 70), copy number assay by fluorescent in situ hybridization (n = 350), and immunohistochemistry (n = 350) were correlated with clincopathological variables, KIT/PDGFRA/BRAF genotypes, and disease-free survival (DFS). HSD11B1 was stably silenced to explore its oncogenic function. HSD11B1 mRNA varied between high-risk and non-high-risk groups (p = 0.009) and positively correlated with HSD11B1 immunoexpression (r = 0.783, p < 0.001). HSD11B1 copy-number gain (CNG), including polysomy (5.4%) and amplification (12%), associated with HSD11B1 overexpression (p < 0.001). Predominantly involving the homodimer interface-affecting exon 6 or exon 7, missense HSD11B1 mutations (17.2%) were related to high risk (p = 0.044), age ≥70 years (p = 0.007), and shorter DFS among relapsed cases (p = 0.033). CNG was related to unfavorable KIT/PDGFRA/BRAF genotypes (p = 0.015), while HSD11B1 overexpression was preferential in non-gastric cases (p < 0.001). Both abnormalities strongly associated with risk levels (both p < 0.001) and shorter univariate DFS (both p < 0.0001), and HSD11B1 CNG remained prognostically independent (p < 0.001) with a 3-fold increased hazard ratio. In vitro, HSD11B1 knockdown significantly inhibited proliferation and caused G2/M arrest. In conclusion, HSD11B1 overexpression may occur owing to CNG, confer a pro-proliferative function, and predict a worse prognosis in GISTs.


2007 ◽  
Vol 25 (18_suppl) ◽  
pp. 3037-3037
Author(s):  
N. M. Reddy ◽  
F. Hernandez-Ilizalituri ◽  
J. Knight ◽  
M. S. Czuczman

3037 Background: Lenalidomide is a thalidomide analogue with immunomodulatory effects. We previously demonstrated that lenalidomide enhances the biological activity of rituximab. In our current work we further studied the effects of combining lenalidomide with IDEC114, a primatized anti-CD80 monoclonal antibody, which is undergoing clinical testing in B-cell lymphoma. Methods: Raji cells were exposed in vitro to lenalidomide (10μg/ml) or DMSO over five days. Changes in DNA synthesis were determined by [3H]-thymidine uptake. For ADCC and CMC assays, lenalidomide or control exposed Raji cells were labeled to 51Cr and then exposed to IDEC114 or isotype control and PBMC’s or human serum. For in vivo studies, 6–8 week old SCID mice were inoculated with 1×106 Raji cells via tail vein injection and after a period of 72 hours animals were divided into four cohorts. Lenalidomide was administered intraperitoneally (i.p) at 0.5mg/kg/dose on days +3, +4, +8, +9, +13, +14, +18 and +19. IDEC114 was administered via tail vein injection at 10mg/kg/dose on days +5, +10, +15 and +20. Difference in survival between treatment groups was performed by Kaplan-Meier analysis. Results: In vitro exposure of Raji to lenalidomide for five consecutive days enhanced the anti-proliferative effects of IDEC114 when compared to control. In addition, an improvement in IDEC114-associated ADCC was observed in lenalidomide-exposed Raji cells. In vivo treatment of SCID mice with lenalidomide in combination with IDEC114 led to prolongation of survival (44 days) compared to either biological agent alone (p<0.01). Conclusions: Our current research demonstrates that Lenalidomide when added to IDEC114 has augmented in vitro antitumor activity (i.e, antiproliferation and ADCC) and synergistic effects in vivo (i.e., prolongation of survival). We hypothesize and currently are evaluating whether the improvement in in vivo antitumor activity of IDEC114, when combined with Lenalidomide, is secondary to potential changes in the tumor microenvironment and/or IMiD-primed upregulation of NK cells and ADCC. This promising unique combination of biologics warrants evaluation as a clinical trial. (Supported by USPHS grant PO1-CA103985 from the National Cancer Institute.) No significant financial relationships to disclose.


2012 ◽  
Vol 30 (15_suppl) ◽  
pp. 7534-7534 ◽  
Author(s):  
D. Ross Camidge ◽  
Margaret Skokan ◽  
Porntip Kiatsimkul ◽  
Barbara Helfrich ◽  
Nathan Schulte ◽  
...  

7534 Background: ALK rearranged NSCLC responds well to ALK inhibitors. Clinically, >15% cells showing rearrangements by break-apart FISH classifies tumors as ALK(+). Native ALK copy number gain has also been reported. We explored the significance of native and rearranged ALK copy number on crizotinib outcomes and whether >15% reflected a clear biological distinction in the frequency of ALK(+) cells. Methods: Copy number and genomic status of ALK assessed by FISH. A total of 1426 NSCLC clinical specimens, 174 ALK(+) and 1252 ALK(-) by standard criteria, and 26 NSCLC cell lines (2 ALK(+) and 24 ALK(-)) were investigated. Results: Native ALK gene mean copy number was significantly higher in ALK(-) than in ALK(+) cases (2.8, SD 0.93, range 1.2-11.4 vs. 1.8, SD 0.79; range 0.6 to 5.2; p<0.01). Frequency of native ALK copy number gain (≥3 copies/cell in ≥40% cells) was 19% in ALK(+) and 62% in ALK(-) tumors (p<0.001). In ALK(-) tumors, abundant focal amplification of native ALK was rare (0.8%); scanty amplification (<10% tumor cells) occurred in 1.1%, and duplication of the entire native ALK or of the ALK 3’ end occurred in 3.5%. Among ALK(-) cell lines, mean native ALK copy number ranged 2.1-6.9 and was not correlated with in vitro crizotinib sensitivity (IC50s 0.34-2.8 uM). In (+) patients, neither native or rearranged ALK copy number, nor percentage cell count correlated with maximal tumor shrinkage or PFS with crizotinib. Clinical specimens with 0-9%, 10-15%, 16-30%, 31-50% and >50% of ALK+ cells were found in 79.3%, 8.5%, 1.4%, 2.7% and 8.1% of cases, respectively. Conclusions: Lower native ALK copy number in ALK(+) NSCLC suggests ALK fusion occurs early, preceding chromosomal instability. Elevated native ALK copy number rarely reflects focal amplification and native ALK copy number increases alone are not associated with sensitivity to ALK inhibition in vitro. Neither native or rearranged copy number, nor positive cell count within ALK(+) tumors influences clinical benefit from ALK inhibition. As 8.5% of ALK(-) cases fall within 5% of the established >15% cell positive threshold, further investigation of ALK status by other diagnostic techniques in this subset may be warranted.


Oncogenesis ◽  
2020 ◽  
Vol 9 (11) ◽  
Author(s):  
Leibo Xu ◽  
Junlong Lin ◽  
Wanyu Deng ◽  
Weixin Luo ◽  
Yipei Huang ◽  
...  

Abstract EZH2, a histone methyltransferase, has been shown to involve in cancer development and progression via epigenetic regulation of tumor suppressor microRNAs, whereas BMI1, a driver of hepatocellular carcinoma (HCC), is a downstream target of these microRNAs. However, it remains unclear whether EZH2 can epigenetically regulate microRNA expression to modulate BMI1-dependent hepatocarcinogenesis. Here, we established that high EZH2 expression correlated with enhanced tumor size, elevated metastasis, increased relapse, and poor prognosis in HCC patients. Further clinical studies revealed that EZH2 overexpression was positively correlated to its gene copy number gain/amplification in HCC. Mechanistically, EZH2 epigenetically suppressed miR-200c expression both in vitro and in vivo, and more importantly, miR-200c post-transcriptionally regulated BMI1 expression by binding to the 3′-UTR region of its mRNA. Furthermore, miR-200c overexpression inhibits the growth of HCC cells in vivo. Silencing miR-200c rescued the tumorigenicity of EZH2-depleted HCC cells, whereas knocking down BMI1 reduced the promoting effect of miR-200c depletion on HCC cell migration. Finally, combination treatment of EZH2 and BMI1 inhibitors further inhibited the viability of HCC cells compared with the cells treated with EZH2 or BMI1 inhibitor alone. Our findings demonstrated that alteration of EZH2 gene copy number status induced BMI1-mediated hepatocarcinogenesis via epigenetically silencing miR-200c, providing novel therapeutic targets for HCC treatment.


2021 ◽  
Vol 23 (Supplement_6) ◽  
pp. vi219-vi219
Author(s):  
Cassandra Verheul ◽  
Federica Fabro ◽  
Ioannis Ntafoulis ◽  
Cecile Beerens ◽  
Youri Hoogstrate ◽  
...  

Abstract INTRODUCTION The search for effective therapies for gliomas is progressively moving towards patient-specific medicine. In order to test patient-tailored therapies, it is vital to develop protocols for reliable establishment of patient-derived glioma cultures. We present a method for reliable culture establishment, with a 95% success rate in 114 consecutive high-grade samples. METHODS Cell cultures were established from either traditionally-resected tumor tissue or ultrasonic surgical aspirator (CUSA) derived tissue fragments, and expanded in serum-free culture, with selection of astrocytic populations if required. Cultures were started from single cells or small tumor fragments of 0.5-3mm (3D). Whole exome and RNA sequencing were carried out with the Illumina Novaseq and HiSeq platforms. Methylation profiling was performed with the Infinium MethylationEPIC array. Cultures and tumors were compared through analysis of single nucleotide polymorphisms and copy number profiles with the Infinium Global Screening Array. Intra-tumoral heterogeneity in cultures was investigated with single-cell transcriptomic sequencing (SORT-seq). We studied tumor-initiating potential by orthotopic injection of cultures in NOD-SCID mice. RESULTS Cultures started from single cells were established from CUSA material more efficiently (92%) than from traditional resection material (70%). 3D-derived cultures had a higher overall efficiency (95% for CUSA, 85% for traditional resection material). We confirmed high concordance in driver mutations, copy number and methylation profiles between tumors and derived cultures. Transcriptomics analysis, comparing tumors and derived cultures, revealed high consistency in gene expression distribution as demonstrated by correlation analysis (r=0.88). Singe-cell RNA-seq shows increased heterogeneity in CUSA derived-cultures, and decreased heterogeneity with passaging over time. Cultures faithfully produce tumors after orthotopic injection in NOD-SCID mice. CONCLUSION We present a highly successful method for the establishment of glioma cultures from patient material, with CUSA-derived cultures revealing greater heterogeneity. Cultures faithfully represent important molecular characteristics of parental tumors and can be used to test potential therapies in vitro.


Sign in / Sign up

Export Citation Format

Share Document