scholarly journals Multi-omics molecular phenotyping of childhood acute lymphoblastic leukemia cell lines reveals novel drug vulnerabilities

Author(s):  
Matthias Stahl ◽  
Luay Aswad ◽  
Isabelle Leo ◽  
Elena Kunold ◽  
Frederik Post ◽  
...  

Abstract Acute lymphoblastic leukemia (ALL) is the most common childhood cancer. Although standard-of-care chemotherapeutics are sufficient for most ALL cases, there are subsets of patients with poor response who relapse in disease. The biology underlying differences between subtypes and their response to therapy has only partially been explained by genetic and transcriptomic profiling. To characterize ALL subtypes and identify novel pharmacologic vulnerabilities, we performed comprehensive multi-omic analyses of 49 widely-used childhood ALL cell lines, using proteomics, transcriptomics, and pharmacoproteomic characterization. This enabled us to characterize the functional impact of genetic fusions and cellular differentiation states. The proteomics data revealed differences in spliceosome and p53 levels not evident in the transcriptomics data and with improved correlation to drug sensitivity. Focusing on BCP-ALL cell lines, we connected the genotype, molecular phenotype, and functional phenotype with drug response data on 528 oncology drugs. Here, we identified the DAG-analog Bryostatin-1 as a novel therapeutic candidate in the MEF2D-HNRNPUL1 fusion high-risk subtype, for which this drug activated pro-apoptotic ERK signaling associated with molecular mediators of pre-B cell negative selection. Our data also forms an interactive online resource with navigable proteomics, transcriptomics, and drug sensitivity profiles at https://lehtio-lab.se/forall/.

Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 791-791 ◽  
Author(s):  
Jun J. Yang ◽  
Heng Xu ◽  
Deepa Bhojwani ◽  
Takaya Moriyama ◽  
Maoxiang Qian ◽  
...  

Abstract Acute lymphoblastic leukemia (ALL) in children is a prototype of cancer that can be cured by chemotherapy alone. However, the molecular mechanisms for anti-leukemic drug sensitivity and genetic basis of inter-patient variability in treatment response are not fully understood. Taking a genome-wide approach, we recently identified genetic variants in the ARID5B gene that strongly predispose children to developing ALL and also a high risk of relapse following therapy (J Clin Oncol 2012 30:751, Nat Genet 2009 41:1001). To understand the mechanisms by which ARID5B is linked to treatment outcome in childhood ALL, we sought to 1) characterize ARID5B expression in different genetic subtypes of ALL, 2) determine the effects of ARID5B expression on cytotoxicity of chemotherapeutic agents commonly used in ALL therapy, and 3) describe molecular pathways linking ARID5B to anti-leukemic drug sensitivity. In 567 children with newly diagnosed ALL treated at St. Jude Children’s Research Hospital (GSE33315), ARID5B expression was highest in cases with hyperdiploid karyotype (>50 chromosomes) and lowest in T-cell ALL and cases with MLL rearrangements. This pattern was validated in an independent cohort of 106 children from the Dutch Childhood Oncology Group (GSE13351). In 59 patients treated on the Children’s Oncology Group (COG) CCG1961 trial, lower ARID5B expression was associated with higher rates of relapse (P=0.01, GSE7440). Importantly, when we compared matched newly-diagnosed vs. relapsed ALL blasts from a cohort of 60 patients enrolled in COG trials (GSE28460), ARID5B expression was further downregulated at disease recurrence (P=0.0009). shRNA-mediated ARID5B knockdown in 3 ALL cell lines (Nalm6, SEM, and UOCB-1) substantially increased resistance to antimetabolites (an average of 5.16 and 35.3-fold increase in IC50 for methotrexate [MTX] and 6-mercaptopurine [6MP], respectively), with minimal effects on glucocorticoids, vincristine, asparaginase, and daunorubicin. Because cytotoxic effects of MTX and 6MP are highly dependent on the rate of cell proliferation, we postulate that ARID5B directly influences cell cycle entry. In all 3 cell lines, ARID5B knockdown led to significant blockade at the G1/S checkpoint, increasing the percent of cells in G0/G1 phase. At the molecular level, downregulation of ARID5B resulted in higher levels of p21 and reduction in phosphorylated Rb, consistent with the retention at G0/G1 phase. ARID5B expression was restricted to nucleus but affected both nuclear and cytoplasmic p21 expression in a time-dependent fashion. Interestingly, there was a highly significant negative correlation between p21 expression and MTX- and 6MP-induced apoptosis in all 3 ALL cell lines. Taken together, we hypothesize that lower expression of ARID5B impairs ALL cell cycling by upregulating p21, contributing to resistance to MTX and 6MP and eventually leukemia relapse. Finally, we compared global gene expression in ARID5B knockdown vs. control ALL cells, and via the Connectivity Map analysis we identified histone deacetylase (HDAC) inhibitors as promising agents for overcoming ARID5B-related drug resistance. Indeed, ARID5B knockdown cells were significantly more sensitive to panobinostat than controls, suggesting HDAC inhibitors as potential therapeutic options for patients with ARID5B-deficient and drug resistant ALL. Disclosures No relevant conflicts of interest to declare.


Cancers ◽  
2020 ◽  
Vol 12 (11) ◽  
pp. 3161
Author(s):  
Weiqiang Zhao ◽  
Ying Li ◽  
Chenjiao Yao ◽  
Guojuan Zhang ◽  
Kevin Zhao ◽  
...  

Leukemia-associated alternative splicing of IKZF1 can result in proteins with loss of one to four copies of its N-terminal zinc finger domains (N-ZnF). The best characterized pathogenic splice isoforms, Ik-6 and Ik-8, have been commonly found in BCR-ABL1+ acute lymphoblastic leukemia (ALL) and a subset of BCR-ABL1-like ALL. Infantile and childhood ALL that express these pathogenic IKZF1 isoforms have shown inferior clinical outcomes and can be resistant to tyrosine kinase inhibitors. Using ALL cell lines, we designed and validated a method to detect abnormal IKZF1 transcripts. In the SUP-B15 leukemia cell line, we noted novel IKZF1 transcripts that include both an Ik-6 splice and a transcript with a 14 base pair insertion at the C-terminus. There was also increased IKZF2 protein in SUP-B15 as compared to other ALL lines. Expression of Ik-6 could be suppressed by treatment with the pro-apoptotic type II histone deacetylase inhibitor givinostat. In 17 adult ALL samples, we noted the Ik-6 isoforms in 6 of 15 BCR-ABL1−, and 1 of 2 BCR-ABL1+ cases, with Ik-8 also expressed in one case. Cases with Ik-6 expression showed inferior survival as well as older age at presentation, lower expression of CD10 and more commonly a diploid karyotype.


Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 155-155
Author(s):  
Maria Rosaria Ricciardi ◽  
Chiara Gregorj ◽  
Fabiana De Cave ◽  
Paola Bergamo ◽  
Samantha Decandia ◽  
...  

Abstract The treatment of adult acute lymphoblastic leukemia (ALL) remains unsatisfactory. A potential hope is now given to Philadelphia-positive cases by targeted treatment modalities. Among other pathways involved in cell proliferation, we have recently demonstrated (Blood2007; 109:5473) the unfavorable role of ERK1/2 phosphorylation as an independent predictor of complete remission (CR) in adult ALL, suggesting the potential therapeutic value of other targeted therapies. The B-cell leukemia/lymphoma 2 (Bcl-2) family of proteins are important regulators of apoptosis and are frequently found aberrantly expressed, particularly in lymphoid malignancies. The role of Bcl-2 overexpression in tumorigenesis and chemoresistance prompted us to investigate whether the inhibition of the antiapoptotic function may result also in ALL in an attractive therapeutic strategy. In this study, we thus investigated the cell cycle and apoptotic effects of ABT-737 (kindly provided by Abbott Laboratories), a Bcl-2 (BH3) inhibitor, on both lymphoid leukemia cell lines and primary adult and childhood ALL cells. The lymphoid leukemia cell lines CEM and MOLT-4 were exposed to increasing concentrations of ABT-737 (from 0.1 to 1 μM) up to 72 hours. A dose- and time-dependent cell growth arrest and induction of apoptosis was found. In fact, measuring the subG0/1 peak at 48 hours, the levels of apoptosis increased in the CEM cell line from 14.1% (DMSO) to 34.4%, 64.5%, 86.5% and 98.6% at ABT-737 concentrations of 0.1, 0.25, 0.5 and 1 μM, respectively. Similarly, 48 hours of exposure to ABT-737 increased in MOLT-4 the Annexin V-positive cells from 7.2% to 64.2%. The effects of ABT-737 were then examined on primary blasts from 9 ALL patients (6 adults and 3 children). Bone marrow aspirates with a blast infiltration >70% were obtained at diagnosis from patients broadly characterized for clinical and biological parameters, as well as therapeutic response. ALL cells were cultured in vitro with ABT-737 (at increasing concentrations from 0.01 to 1 μM) for 24 hours. A significant decrease in viability was observed at 0.01 μM (p=0.008) with a remarkable dose-dependent increase of apoptosis. In fact, Annexin V-positive cells increased from a mean baseline value of 16.8% ± 8.8 to 43.6% ± 22.8 (p=0.04), 66% ± 21.3 (p=0.0001), 70.3% ± 26.9 (p=0.04), 74.6% ± 18.9 (p=0.03) and 76.2% ± 11.8 (p<0.0001) in the presence of ABT-737 at 0.01, 0.1, 0.25, 0.5 and 1 μM, respectively. A significant cell killing was demonstrated in all samples (9/9), including Ph-positive ALL. No significant cell cycle changes were instead detected even at higher concentration of ABT-737. In summary, our study shows for the first time a potent growth-inhibitory and pro-apoptotic activity of the Bcl-2 antagonist ABT-737, at nanomolar concentrations, on primary cells from adult and childhood ALL samples. These results prompt to further extend pre-clinical studies in the different biologically-defined subset of ALL and suggest a potential clinical development of a Bcl-2 family inhibitor in this disease.


2009 ◽  
Vol 33 (10) ◽  
pp. 1386-1391 ◽  
Author(s):  
Hiroaki Goto ◽  
Takuya Naruto ◽  
Reo Tanoshima ◽  
Hiromi Kato ◽  
Tomoko Yokosuka ◽  
...  

2018 ◽  
Vol 120 (5) ◽  
pp. 7428-7438 ◽  
Author(s):  
Nashwa El‐Khazragy ◽  
Amal Ali Elshimy ◽  
Safaa Shawky Hassan ◽  
Safa Matbouly ◽  
Gehan Safwat ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document