scholarly journals Recurrent Germline Variant in the Cohesin Complex Gene RAD21 Predisposes Children to Lymphoblastic Leukemia and Lymphoma

Blood ◽  
2021 ◽  
Vol 138 (Supplement 1) ◽  
pp. 3358-3358
Author(s):  
Anne Schedel ◽  
Ulrike Anne Friedrich ◽  
Rabea Wagener ◽  
Juha Mehtonen ◽  
Claudia Saitta ◽  
...  

Abstract Introduction: Cohesin complex genes are commonly mutated in cancer particularly in myeloid malignancies. Yet patients with germline mutations in cohesin genes, leading to cohesinopathies like Cornelia-de-Lange syndrome (CdLS) are generally not known to be tumor-prone. The complex plays a major role in chromosome alignment and segregation (Uhlmann, Nature Reviews Molecular Cell Biology, 2016), homologous recombination-driven DNA repair (Ström et al., Molecular Cell, 2004) and regulation of gene expression (Busslinger et al., Nature, 2017). To deepen the understanding of cohesin variants in cancer predisposition, we performed TRIO Sequencing in two independent pediatric cancer cohorts. Thereby, we identified a novel recurrent heterozygous germline variant in the cohesin gene RAD21 not described in CdLS patients , located in the binding domain of the cofactors WAPL and PDS5B . Methods: Whole exome sequencing (WES) in a TRIO (child-parent datasets) setting was carried out in two independent, unselected cancer cohorts (TRIO-D, n=158 (Wagener et al., European Journal of Human Genetics, 2021) and TRIO-DD, n=60). To investigate the oncogenic potential of the novel RAD21 variant molecular and functional assessment was performed focusing on potential implications on the complex. Results: The newly identified RAD21 variant at amino acid position 298 resulting in a Proline to Serine (p.P298S) and a Proline to Alanine exchange, respectively, (p.P298A) is only rarely mutated in the general population (gnomAD database n=118,479; RAD21 p.P298S MAF <10 -6 and RAD21 p.P298A MAF <10 -5). While both patients did not show any signs of CdLS, they both have a remarkable family history of cancer. Patient 1 (13y) was diagnosed with T-cell acute lymphoblastic leukemia (T-ALL) whose father had died from breast cancer (41y), while patient 2 (2y) presented with precursor B-cell lymphoblastic lymphoma (pB-LBL) whose uncle had died from pediatric cancer of unknown subtype (8y). To assess the influence of RAD21 p.P298S/A on the binding capacity of the complex, RAD21 variants and the wildtype (WT) were cloned and transfected into HEK293T cells, respectively. Immunoprecipitation analysis of RAD21 with the cofactors WAPL and PDS5B showed no differential binding between the WT and the variants, suggesting that RAD21 p.P298S/A does not impact the formation of the complex. Nevertheless, on a transcriptional level 83 genes were significantly differentially expressed in RAD21 p.P298S and p.P298A compared to the wildtype (fc>1.5, adj. p-value <0.05) with enrichment of genes in p53 signaling pathways. We further observed an increased number of γH2AX and 53BP1 co-localized foci compared to the WT (p≤0.01; Student's t-test). In line, following ionizing radiation, primary patients' samples showed increased cell cycle arrest at G2/M cell-cycle stage compared to a healthy control (p.P298S: p=0.0049 [6Gy]; p=0.0026 [10Gy]; p.P298A: p=0.0054 [6Gy]; p=0.0006 [10Gy]; Student's t-test). For cross-validation of the germline variant RAD21 p.P298S/A and its potential role in pediatric lymphoblastic malignancies, we analysed a third cohort of 150 children with relapsed ALL (IntReALL) for RAD21 p.P298S/A. We again identified RAD21 p.P298A in a boy (12y) with B-cell precursor acute lymphoblastic leukemia. To compare our data to a non-pediatric cancer setting, a cohort of 2300 young adults (<51 years) with cancer was mined (MASTER program). Here, one patient carrying RAD21 p.P298A with a solid tumor was identified. Therefore, amongst all cohorts, RAD21 p.P298S/A was found to be enriched in pediatric vs. adult cancers (3/479 vs. 1/2299; Fisher's exact test; p=0.018). Conclusion: Taken together, we present for the first time the potential role of RAD21 germline variants in pediatric lymphoblastic malignancies. This may shed new light on the many roles of the cohesin complex and its implication outside the typical syndromal presentation. Disclosures No relevant conflicts of interest to declare.

Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 3734-3734
Author(s):  
Sinisa Dovat ◽  
Chunhua Song ◽  
Xiaokang Pan ◽  
Yali Ding ◽  
Chandrika S. Gowda ◽  
...  

Abstract IKZF1 (Ikaros) encodes a kruppel-like zinc finger protein that is essential for normal hematopoiesis and acts as a tumor suppressor in acute lymphoblastic leukemia (ALL). The deletion and/or mutation of Ikaros is associated with the development of human T-cell and B-cell acute lymphoblastic leukemia (B-ALL) with poor outcome. In vivo, Ikaros binds DNA and regulates gene expression by chromatin remodeling. Since there is a paucity of known genes that are regulated by Ikaros, the molecular mechanisms through which Ikaros exerts its tumor suppressor function remain unknown. Here we describe studies that identify the targets and mechanisms of Ikaros-mediated epigenetic regulation in human B-ALL. We used chromatin immunoprecipitation coupled with next generation sequencing (ChIP-seq) to identify target genes that are bound by Ikaros in vivo in human B-ALL, and to define epigenetic patterns associated with Ikaros binding. ChIP-seq revealed a large set of Ikaros target genes that contain a characteristic Ikaros binding motif. The largest group of genes that are direct Ikaros targets included genes that are essential for cell cycle progression. These included CDC2, CDC7, CDK2 and CDK6 genes whose deregulation is associated with malignant transformation. The strong binding of ikaros to the promoters of cell cycle-promoting genes was confirmed by quantitative immunoprecipitation in primary leukemia cells. To establish whether Ikaros directly regulates transcription of the cell cycle-promoting genes, their expression was measured in B-ALL cells that were transduced with either a retroviral vector that contains Ikaros, or a control vector. Target gene expression was monitored by qRT-PCR. Ikaros strongly repressed transcription of the cell cycle-promoting genes, which resulted in cell cycle arrest. Global epigenetic profiling using ChIP-seq suggested that Ikaros represses cell cycle-promoting genes by inducing epigenetic changes that are consistent with repressive chromatin. High-resolution epigenetic profiling of the upstream regulatory elements of the cell cycle-promoting genes targeted by Ikaros showed that increased Ikaros expression results in the formation of heterochromatin, which is characterized by the presence of the H3K9me3 histone modification and associated transcriptional repression. Functional analysis revealed that phosphorylation of Ikaros by the oncogenic protein. Casein kinase II (CK2), impairs its function as a transcriptional repressor of the cell cycle-regulating genes. Inhibition of CK2 by specific inhibitors enhances Ikaros-mediated repression of the cell cycle-regulating genes resulting in cessation of cellular proliferation and cell cycle arrest in vitro and in vivo in a B-cell ALL preclinical model. This was associated with increased Ikaros binding and the formation of heterochromatin at upstream regulatory elements of the cell cycle-promoting genes. Our results provide evidence that Ikaros functions as a repressor of cell cycle-promoting genes in B-ALL by directly binding their promoters and inducing the formation of heterochromatin with characteristic H3K9me3 histone modifications Ikaros repressor function is negatively regulated by CK2 kinase in B-cell ALL. Inhibition of CK2 enhances Ikaros mediated-repression of cell cycle-promoting genes resulting in an anti-leukemia effect in a preclinical model of B-cell ALL. Presented data identified the mechanism of action of CK2 inhibitors and demonstrated their efficacy in B-cell ALL preclinical model. Results support the use of CK2 inhibitors in Phase I clinical trial. Supported by National Institutes of Health R01 HL095120 and a St. Baldrick’s Foundation Career Development Award (to S.D.). Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 5283-5283
Author(s):  
Chuang Jiang ◽  
Jiabi Qian ◽  
Wenge Hao ◽  
Wei LIU ◽  
Shuhong Shen ◽  
...  

Abstract Background: Thanks to the total therapy and systemic basic-translation research, the overall survival rate in children with acute lymphoblastic leukemia (ALL) has dramatically improved to almost 90% over these past few decades. FOXO1 gene belongs to the forkhead family of transcription factors, which play roles in myogenic growth and differentiation. Translocation of FOXO1 with PAX3 has been reported in pediatric alveolar rhabdomyosarcoma. In B-cell precursor ALL, two cases with FOXO1 fusions have been identified already, while its function on ALL remains unknown. Here, we report a novel MEIS1-FOXO1 fusion gene in a case with B-ALL. Methods: Flowcytometery, karyotype, RT-PCR and fluorescence in were employed, MEIS1-FOXO1 was identified as novel fusion gene in a case of pediatric BCP-ALL. Using IL-3 dependent BaF3 cells as study model to test the leukemia transformation potential of MEIS1-FOXO1. Results: A novel MEIS1-FOXO1 fusion was identified in one cease of pediatric B-ALL. Panel next generation sequencing (NGS) showed that the leukemia clone had concurrent NRASG12D, TP53R273H, WHSC1E1099K, ABCC1R1166X, PHGR1H37P, HOXA3P219L and DSTP4606L somatic mutation. This patient was enrolled in CCCG-ALL2015 clinical trial (ChiCTR-IPR-14005706) and achieved completed remission and low minimal residual disease (MRD) level (MRD<0.01%) at day 19 from induction therapy. Functional study showed that MEIS1-FOXO1 fusion gene can potentiate BaF3 cells growth independent of IL3 supplement, as compared to those without MEIS1-FOXO1 fusion transduction. In the meanwhile, we have found that MEIS1-FOXO1 fusion gene can drive cells into S-phase with concurrent decreased G0/G1 phase, which might be its oncogenic role in leukemogenesis. Using qPCR methods, we have found that MEIS1-FOXO1 fusion gene altered the cell cycle related genes expression. Conclusions: Integrating the FOXO1-fusion reports, our data have added more evidence to underline the role of FOXO1 deregulation in the pathogenesis of acute lymphoblastic leukemia. Novel fusion of MEIS1-FOXO1 can potentiate B-ALL via cell cycle entry. Detailed mechanisms involved into the MEIS1-FOXO1 should be further investigated. Figure. Figure. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 3534-3534 ◽  
Author(s):  
Nathalie Y Rosin ◽  
Stefan Koehrer ◽  
Ekaterina Kim ◽  
Susan O'Brien ◽  
William G. Wierda ◽  
...  

Abstract Abstract 3534 Acute lymphoblastic leukemia (ALL) is a highly heterogeneous disease. B-cell acute lymphoblastic leukemia (B-ALL) is characterized by uncontrolled proliferation of immature lymphoid blasts with suppression of normal hematopoiesis. Phosphoinositide 3-kinases (PI3K) transmit activation signals from diverse transmembrane receptors, leading to generation of phosphatidylinositol- 3,4,5-trisphosphate (PIP3) which promotes proliferation, differentiation, migration, and survival in lymphocytes and various other cell types. A knockout mouse model of the PI3K isoform p110δ demonstrates a unique role of p110δ (PI3Kδ) in B cell receptor (BCR) signaling. This is corroborated by clinical efficacy of the PI3Kδ inhibitor GS-1101 in mature B cell malignancies, especially in chronic lymphocytic leukemia (CLL). In contrast to mature B cell malignancies, expression and function of PI3Kδ in B-ALL has not been well characterized. We therefore analyzed PI3Kδ expression and effects of the PI3Kδ inhibitor GS-1101 in B-ALL. To screen efficacy of GS-1101 in B-ALL subsets, we performed viability and proliferation assays, using a panel of B-ALL cell lines, derived from different B-cell development stages (Pro-B: REH, RS4;11, Nalm-20, Nalm-21, TOM-1; Pre-B: Nalm-6, Kasumi-2, KOPN-8, SMS-SB, RCH-ACV, 697; Mature: Tanoue, Ball-1 unknown: CCRF-SB). A key downstream effector of PI3K is the serine/threonine kinase Akt, whose phosphorylation is used as a common readout of PI3K activation status. Western Blot analysis of the 15 cell lines showed almost identical levels of phospho-Akt (Ser473) in all tested cell lines, suggesting constitutive PI3K activity. To investigate the ability of GS-1101 to inhibit B-ALL cell proliferation, we performed cell growth experiments. Among the pre-B cell lines 4 of 6 showed a marked decrease in proliferation, 2 other pre-B cell lines showed a minor decrease. In contrast, none of the pro-B or mature B-ALL cell lines were affected by GS-1101. To explore the effects of GS-1101 on cell cycle of B-ALL cells, cell lines were treated with GS-1101 at concentrations ranging from 0.5μM to 5μM. In accordance with the cell growth experiments, G1 phase arrest and reduced numbers of S phase cells were detected in pre-B cell lines after GS-1101 treatment, but not in the pro-B or mature B cell lines. Next, we examined GS-1101 effects on metabolism of B-ALL cells via XTT (sodium 2,3,-bis(2-methoxy-4-nitro-5-sulfophenyl)-5-[(phenylamino)-carbonyl]-2H-tetrazolium inner salt) staining. Cell lines were treated with GS-1101 concentrations between 0.1μM and 5μM for 3 days prior to XTT measurement. Pre-B cells showed a significant (p-value <0.0001) decrease in normalized absorbance compared to the control (without treatment) indicating a decrease in cellular viability. Finally, preliminary co-culture experiments of primary B-ALL samples and KUSA-H1 bone marrow stromal cells revealed significantly reduced B-ALL cell viability after GS-1101 treatment, signifying that GS-1101 can overcome microenviromental-mediated B-ALL cell protection; this is similar to that in other B cell malignances. In summary, these experiments demonstrate that GS-1101 inhibits growth, cell cycle progression and metabolic activity of pre-B ALL cells. Validation of these data with primary patient samples is ongoing. Disclosures: Lannutti: Gilead Sciences Inc: Employment.


Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 3962-3962
Author(s):  
Jason Saliba ◽  
Joanna Pierro ◽  
Nikki Ann Evensen ◽  
Anita Qualls ◽  
Natasha Belsky ◽  
...  

Abstract Introduction: While the outcome for children with acute lymphoblastic leukemia (ALL) has improved dramatically, the prognosis for those who relapse remains poor. One of the most common alterations found at relapse is the p.E1099K missense change within the SET domain of NSD2, a histone methyltransferase that di-methylates histone 3 lysine 36 (H3K36). NSD2 has 3 isoforms, two of which, Type II (canonical) and REIIBP (C-terminal), contain the SET domain, and another, Type I (N-terminal), that does not. The p.E1099K mutation leads to increased enzymatic activity, but pathways leading to a clonal advantage are unknown in ALL. Methods: We used short hairpin RNAs (shRNAs) to target knockdown of two combinations of NSD2 isoforms: shI/II targets Types I and II, shII/RE targets Type II and REIIBP. Three different B-cell lines (Reh, 697, and KOPN-8) with 2 wildtype (WT) copies of NSD2 were stably transduced with shII/RE. Two B-Cell lines, RS4;11 and RCH-ACV, heterozygous for the NSD2 p.E1099K mutation, were transduced with shI/II and shII/RE. As a control, each B-cell line was stably transduced with a scrambled non-targeting (NT) shRNA. NSD2 knockdown was confirmed by Western Blots. Cell lines were treated for 5 days with chemotherapy agents commonly used in pediatric ALL treatments (mercaptopurine (MP), cytarabine, methotrexate, prednisone, and doxorubicin). Cytotoxicity was assessed by CellTiter- Glo® and significance between IC50s was determined by ANOVA and post hoc Tukey test. Cell proliferation was measured by cell counting with trypan blue. Cell cycle progression in RS4;11 lines was monitored with Edu staining and flow cytometry with and without exposure to MP. Results: Similar to previously reported results, knockdown of NSD2 in the 3 WT B-cell lines had no effect on cell proliferation. However, shI/II reduced growth by 40% in RS4;11 and 20% in RCH-ACV, while shII/RE decreased proliferation by 45% in RS4;11 and 55% in RCH-ACV when compared to their NT control. In RS4;11, both shI/II and shII/RE led to a similar 10% decrease in cells progressing through S phase compared to NT, which could be due to either a slower progression through cell cycle or less cells entering the cell cycle. Knockdown of NSD2 resulted in sensitivity to 6MP compared to NT in both RS4;11 and RCH-ACV lines. RS4;11 shII/RE had an IC50 3.2-fold more sensitive ( p<.01) and the RS4;11 shI/II IC50 was 1.25-fold more sensitive (NS) versus the NT control. Similarly, RCH-ACV shII/RE had an IC50 3.4-fold more sensitive (p<.01) and the RCH-ACV shI/II IC50 was 2.6-fold more sensitive (p<.01) compared to the NT control. No significant changes in drug sensitivity were noted for the 3 WT NSD2 knockdown B-cell lines compared to their NT controls. During a 120 hour exposure to MP, 34% more RS4;11 shII/RE cells were arrested in the G phase than NT controls, while 26% more RS4;11 shI/II cells were arrested in G phase relative to NT controls. This result indicates MP exposure leads to a reduced percentage of knockdown cells able to progress through the cell cycle. Overall, simultaneously reduced expression of Type II and REIIBP had a greater effect of on cell proliferation and MP response compared to the co-reduction of Types I and II NSD2 in the p.E1099K heterozygous cell lines. Conclusion: The p.E1099K mutation confers a growth advantage and resistance to MP, a cornerstone of ALL therapy. Concurrent reduction of Type II and REIIBP expression by shII/RE resulted in the largest impact on proliferation and MP sensitivity. Both of these isoforms include the SET domain containing the p.E1099K mutation, which indicates one or both isoforms could be responsible for changes in the chromatin state and other possible alterations that lead to a clonal advantage. Based on our findings, determining the mechanism of resistance to MP imparted by NSD2 p.E1099K is now a top priority. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 429-429
Author(s):  
Charles G Mullighan ◽  
Letha A Phillips ◽  
Xiaoping Su ◽  
Jing Ma ◽  
Christopher B Miller ◽  
...  

Abstract Relapsed acute lymphoblastic leukemia (ALL) is the fourth most common pediatric malignancy and carries a dismal prognosis. To gain insights into the genetic alterations responsible for relapse, we performed genome-wide analysis of matched diagnosis and relapse samples from 61 pediatric ALL patients, including 47 B-progenitor and 14 T-ALL cases. All samples were flow sorted as required to ensure at least 80% tumor cell purity prior to DNA extraction. DNA copy number abnormality (CNA) and loss-of-heterozygosity (LOH) data was generated using Affymetrix SNP 6.0 arrays (1.87 million markers) for 47 diagnosis-relapse pairs, and 500k arrays for the remainder. Remission marrow samples were also examined for 48 cases. Analysis of diagnostic samples identified 10.8 CNAs per B-ALL case and 7.4 per T-ALL case. The most frequent target of CNAs at diagnosis were deletions of genes involved in B-lymphoid development (49% of B-ALL cases, involving PAX5 and IKZF1 in 12 cases each), CDKN2A/B in 36% of B-ALL and 71% of T-ALL, and ETV6 (11 B-ALL cases). At relapse, we observed a striking degree of change in the number, extent, and nature of CNAs. In B-ALL the average number of CNAs increased from 10.8/case at diagnosis to 14.0/case at relapse (P=0.0005), with the majority of this change due to an increase in deletions. By contrast, no difference in the number of CNAs was observed between diagnosis and relapse in T-ALL. Most relapse samples (54 of 61) harbored some of the CNAs present at diagnosis, and had identical or similar antigen receptor locus deletions, indicating a common clonal origin. Nevertheless, 92% of relapse cases exhibited significant changes in CNAs, including the acquisition of new lesions (34%), loss or alteration of lesions present at diagnosis (12%), or both acquisition of new lesions and loss of diagnosis lesions (46%). In the cases where a clear clonal relationship existed, almost half of the relapse clones were derived from a pre-leukemic cell and not from the clone predominating at diagnosis. The most frequent targets of relapse-acquired CNAs were CDKN2A/B, ETV6, and regulators of B-lymphoid development. 18 cases developed new CDKN2A/B deletions, with 70% showing bi-allelic loss; 11 developed new ETV6 lesions; and 15 developed new or more extensive CNAs involving B-lymphoid regulators. In contrast to diagnosis where PAX5 is most frequently involved, at relapse Ikaros and related gene family members were most common (IKZF1, 8 cases; IKZF2, 2 cases; IKZF3, 1 case). Other CNAs previously identified at diagnosis were also detected as new lesions at relapse, including deletions of ADD3, ATM, BTG1, FHIT, KRAS, NF1, PTCH, TBL1XR1, TOX and WT1, suggesting that these lesions contribute to treatment resistance. To further define the biological pathways most frequently altered by relapse–acquired CNAs, the genes within altered regions were categorized into 148 different biological pathways and each cases was then assessed for overrepresentation of these pathways. This analysis identified cell cycle and B-cell transcriptional regulatory pathways as the most significantly targeted pathways at relapse. To determine whether CNAs identified at relapse were present at low levels at diagnosis or were acquired during therapy, we developed qualitative genomic PCR assays for deletions involving ADD3, C20orf94, DMD, ETV6, IKZF2 and IKZF3, and tested corresponding diagnosis and relapse samples. This analysis detected evidence of the relapse clone at diagnosis in 7 of 10 cases tested, indicating that in the majority of cases, CNAs that emerge in the predominant clone at relapse are present at low levels at diagnosis and are selected for during treatment. These results provide critical insights into the spectrum of genetic lesions that underlie ALL relapse. Although our data are limited to a single class of mutations (CNAs), they demonstrate that no single genetic lesion or alteration of a single pathway is responsible for relapse. Instead, a diversity of mutations appear to contribute to relapse with the most common alterations targeting key regulators of tumor suppression, cell cycle control, and lymphoid/B cell development. Notably, few lesions involved genes with roles in drug import, metabolism, export and/or response, (an exception being the glucocorticoid receptor gene NR3C1) suggesting that that the mechanism of relapse is more complex than simple “drug resistance”.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 874-874
Author(s):  
Maike Buchner ◽  
Lars Klemm ◽  
Chen Zhengshan ◽  
Huimin Geng ◽  
Markus Muschen

Abstract Abstract 874 Background: Despite initial responsiveness of primary Philadelphia chromosome positive acute lymphoblastic leukemia (Ph+ALL) to tyrosine kinase inhibition (TKI), the majority of patients will relapse and develop TKI-resistant disease. Foxm1 belongs to the forkhead box transcription factor family and is a key regulator of malignant growth by promoting cell cycle and survival through increased DNA damage repair. Foxm1 has been implicated in the progression and chemoresistance in a wide range of solid tumors, including hepatocellular carcinoma and breast cancer. Foxm1 is expressed in dividing cells and regulates the expression of critical regulators for G2/M entry of the cell cycle including Cdc25B, cyclin-B1, Plk-1 and Aurora B kinase. In addition it decreases protein stability of p27kip and p21cip and regulates the expression of antioxidant defense machinery of the cell, e.g. by superoxide dismutase expression. Results: We compared Foxm1 expression levels in Ph+ ALL patient samples and CD19+ B cell precursors from healthy donors and found 12-fold higher levels in the leukemic cells (p=0.011). More importantly, Foxm1 levels at the time of diagnosis in a clinical trial for patients with high risk acute lymphoblastic leukemia (ALL) were predictive of poor outcome (COG P9906; n=207). Comparative analysis of microarray data from matched sample pairs at diagnosis and relapse revealed a significant upregulation of Foxm1 in the relapse samples (n=42; p=0.0025). To further study the role of Foxm1 in Ph+ ALL, we developed a genetic model for inducible inactivation of Foxm1 in Ph+ ALL. To this end, B cell precursors of Foxm1fl/fl mice were transformed with BCR-ABL1 and transduced with a tamoxifen (4-OHT)-inducible Cre. Interestingly, 4-OHT-mediated deletion of Foxm1 resulted in reduced cell viability and an arrest in G0/G1 with a significant decrease of the S-phase of the cell cycle following deletion of Foxm1. The ability to form colonies in vitro was significantly decreased by deletion of Foxm1. In addition, Foxm1−/− ALL cells revealed a strikingly higher sensitivity towards TKI-treatment (Imatinib dose-response curve) compared to the control cells. As a potential therapeutic agent to pharmacologically inhibit Foxm1 function, we evaluated the effects of a previously described ARF peptide that binds and inhibits Foxm1 function. We treated TKI-resistant (BCR-ABL1T315I) and TKI-sensitive patient-derived xenograft Ph+ ALL cells with various ARF peptide concentrations and found significant growth inhibition after 72h (IC50 16.8±4.3μM, n=4), regardless of TKI responsiveness. In addition, treatment of ARF peptide in combination with TKI reduced the viability from 65.7%±1.7 after TKI treatment alone (10μM) to 19%±0.8 after 48h (TKI 10μM ARF peptide 12μM). Ph+ ALL cells treated with similar concentrations of a mutated ARF control peptide revealed 77%±0.9 viable cells and ARF peptide treatment alone decreased the viability to 29.6%±0.4. Hence treatment with the ARF peptide alone induces apoptosis in patient-derived Ph+ ALL cells and enhances the effect of TKI, which confirms the findings of the ALL mouse model for human Ph+ ALL xenografts. In a complementary approach, we used the natural antibiotic Thiostrepton, which functions via Foxm1 blockade. To test the ability of Thiostrepton as a potential anti-leukemia agent, we studied patient-derived TKI-resistant (BCR-ABL1T315I) Ph+ ALL cells. Treatment of these patient derived Ph+ALL cells induced cytotoxicity in nanomolar concentrations of Thiostrepton along with a significant downregulation of Foxm1 protein levels. By contrast, Non-BCR-ABL1 tumor cells including lymphoma cells were not responsive to Thiostrepton treatment at similar concentrations. Conclusion: Our analyses reveal that Foxm1 is a valid therapeutic target for the treatment of TKI sensitive and resistant Ph+ ALL, including BCR-ABL1T315I. We show that Foxm1 has a crucial function in Ph+ ALL and impacts a) leukemia proliferation, b) colony formation, and c) TKI-resistance. These findings identify Foxm1 a rational target for combination therapy with TKI or as a single agent for TKI-resistant Ph+ ALL. Disclosures: No relevant conflicts of interest to declare.


2009 ◽  
Vol 206 (8) ◽  
pp. 1739-1753 ◽  
Author(s):  
Daniel Trageser ◽  
Ilaria Iacobucci ◽  
Rahul Nahar ◽  
Cihangir Duy ◽  
Gregor von Levetzow ◽  
...  

B cell lineage acute lymphoblastic leukemia (ALL) arises in virtually all cases from B cell precursors that are arrested at pre–B cell receptor–dependent stages. The Philadelphia chromosome–positive (Ph+) subtype of ALL accounts for 25–30% of cases of adult ALL, has the most unfavorable clinical outcome among all ALL subtypes and is defined by the oncogenic BCR-ABL1 kinase and deletions of the IKAROS gene in &gt;80% of cases. Here, we demonstrate that the pre–B cell receptor functions as a tumor suppressor upstream of IKAROS through induction of cell cycle arrest in Ph+ ALL cells. Pre–B cell receptor–mediated cell cycle arrest in Ph+ ALL cells critically depends on IKAROS function, and is reversed by coexpression of the dominant-negative IKAROS splice variant IK6. IKAROS also promotes tumor suppression through cooperation with downstream molecules of the pre–B cell receptor signaling pathway, even if expression of the pre–B cell receptor itself is compromised. In this case, IKAROS redirects oncogenic BCR-ABL1 tyrosine kinase signaling from SRC kinase-activation to SLP65, which functions as a critical tumor suppressor downstream of the pre–B cell receptor. These findings provide a rationale for the surprisingly high frequency of IKAROS deletions in Ph+ ALL and identify IKAROS-mediated cell cycle exit as the endpoint of an emerging pathway of pre–B cell receptor–mediated tumor suppression.


2021 ◽  
Vol 22 (15) ◽  
pp. 8166
Author(s):  
Natalia-Del Pilar Vanegas ◽  
Paola Fernanda Ruiz-Aparicio ◽  
Gloria Inés Uribe ◽  
Adriana Linares-Ballesteros ◽  
Jean-Paul Vernot

Leukemic cell growth in the bone marrow (BM) induces a very stressful condition. Mesenchymal stem cells (MSC), a key component of this BM niche, are affected in several ways with unfavorable consequences on hematopoietic stem cells favoring leukemic cells. These alterations in MSC during B-cell acute lymphoblastic leukemia (B-ALL) have not been fully studied. In this work, we have compared the modifications that occur in an in vitro leukemic niche (LN) with those observed in MSC isolated from B-ALL patients. MSC in this LN niche showed features of a senescence process, i.e., altered morphology, increased senescence-associated β-Galactosidase (SA-βGAL) activity, and upregulation of p53 and p21 (without p16 expression), cell-cycle arrest, reduced clonogenicity, and some moderated changes in stemness properties. Importantly, almost all of these features were found in MSC isolated from B-ALL patients. These alterations rendered B-ALL cells susceptible to the chemotherapeutic agent dexamethasone. The senescent process seems to be transient since when leukemic cells are removed, normal MSC morphology is re-established, SA-βGAL expression is diminished, and MSC are capable of re-entering cell cycle. In addition, few cells showed low γH2AX phosphorylation that was reduced to basal levels upon cultivation. The reversibility of the senescent process in MSC must impinge important biological and clinical significance depending on cell interactions in the bone marrow at different stages of disease progression in B-ALL.


Sign in / Sign up

Export Citation Format

Share Document