The Genomic Landscape of TEL-AML1+ (ETV6-RUNX1) Acute Lymphoblastic Leukaemia

Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 403-403
Author(s):  
Elli Papaemmanuil ◽  
Inmaculada Rapado ◽  
Anthony M Ford ◽  
Keiran Raine ◽  
Jonathan Hinton ◽  
...  

Abstract Abstract 403 TEL-AML1 (ETV6-RUNX1) fusions are common (∼25%) in paediatric B cell precursor acute lymphoblastic leukaemia (ALL) and associated with a very favourable prognosis. Our prior studies on monozygotic twins with concordant ALL and ‘backtracking' studies using archived neonatal blood spots established that ETV6-RUNX1 fusion is usually an early or initiating mutational event arising pre-natally. Subsequent studies, have provided strong evidence that additional genetic changes are essential for the clinical development of ALL. Twin studies suggest that these additional events are most likely to be post-natal and secondary to the ETV6-RUNX1 fusion (Bateman C et al, Blood 2010). To obtain a more in depth portrait of the composite genetic events that drive this subtype of ALL, we have carried out an extensive genomic analysis of 58 cases with diagnostic (leukaemic) cell DNA paired with matched, remission sample as a source of constitutive DNA. For all 58 cases, both leukemic and remission samples were subjected to massively parallel sequencing across all protein coding exons and a subset of selected non-coding RNAs. All cases were sequenced to at least 35x sequence coverage. Likely acquired mutations were called using algorithms developed in house for single base pair substitutions, small insertions and deletions. All putative somatic mutations were validated using conventional Sanger sequencing and to allow for estimation of allelic burden we further validated all somatic calls by massively parallel pyrosequencing. More than 700 mutations were confirmed to be somatically acquired. The majority of these mutations (>500) represent non-synonymous coding changes or stop-codons. Similar to other paediatric neoplasms, each patient harboured a modest number of total mutations (n=13.5, 95% CI: 9.6–17.4). Interestingly, recurrent mutations were present in 42 genes, however the majority (n=37) were only present in two cases each. Allelic burden estimates across all confirmed somatic variant positions in a patient sample, demonstrated evidence of sub-clonal events at presentation. This is being validated by quantitative PCR and single cell analysis. Mutation signatures were profiled in respect to occurrence, frequency and sequence context. To implement a detailed characterisation of these patterns, further sequencing of the entire genome is currently under way in selected samples. To further characterise the genomes of ETV6-RUNX1+ ALL and to identify novel cryptic rearrangements, whole genome paired-end sequencing at a physical depth of at least 16x was performed on 49 cases. Paired reads were mapped to build 37 of the human reference genome. Mapping of the breakpoints to base-pair resolution demonstrated that the ETV6-RUNX1 fusion is the result of complex rearrangements involving several chromosomal events. A further series of genomic rearrangements were defined and selected for validation by conventional PCR of the breakpoint junctions in the matched sample pairs. In summary, we report the identification of a novel spectrum of somatic mutations in ETV6-RUNX1+ ALL and present the first detailed characterisation of the genomic landscape of this ALL subtype. We provide new insights into the molecular pathology of ETV6-RUNX1+ ALL and discuss the potential implications of our findings both in understanding the underlying molecular mechanisms and also its clinical management. Disclosures: No relevant conflicts of interest to declare.

2019 ◽  
Vol 37 (15_suppl) ◽  
pp. e15670-e15670
Author(s):  
Jiazhou Ye ◽  
Yinguang Wang ◽  
Rong Liang ◽  
Xue Wu ◽  
Yang Shao ◽  
...  

e15670 Background: Development of hepatocellular carcinoma (HCC) is a complex process with accumulations of polygene abnormalities and multi-pathway misregulation. Hepatitis B virus (HBV) exposure can cause liver damage and promote hepatocarcinogenesis via various biological effects. We aimed to investigate the molecular mechanisms underlying the etiology of HBV-related HCC development, and provide new insights into novel molecular targets. Methods: 84 HBV-positive HCC patients from Guangxi Province, South China, who underwent hepatic resection, were enrolled in this study. Genomic alterations were analyzed in pair-matched tumor and adjacent normal tissue using a hybridization capture-based next-generation sequencing (NGS) assay targeting 422 cancer-relevant genes. Results: In total, 691 somatic mutations, 166 copy number variations and 10 gene fusions were detected in 81 (96.4%) of 84 tumor samples. The most commonly mutated gene is TP53 in this cohort (84% of the patients), which is much higher than its frequency in the reported overall HCC patients. TERT promoter has somatic mutations in 32% of the patients, reactivation of which has been implicated in multiple cancer types. Dysfunction in the cell cycle control pathway (TP53, RB1, CCND1, CDKN2A and CCNE1) was dominant, followed by PI3K/AKT cascade (PIK3CA, AKT3, MTOR, TSC1 and TSC2), while genes of WNT signaling pathway (CTNNB1, APC and AXIN2) were mutated at a lower frequency. In addition, 69 variants in 25 DNA damage repair (DDR) genes were identified in 37 (45.7%) patients. Patients with DDR mutations had a higher tumor mutation burden (TMB) than those without DDR mutations. Conclusions: This study revealed a unique genomic landscape of HBV-related HCC. Besides TP53 being the highest mutated gene, a significant fraction of patients was identified with TERT promoter mutations, suggesting that TERT may play a role in HBV-related hepatocarcinogenesis as a novel molecular marker. Furthermore, the most common biological processes affected by HBV status in HCC were cell cycle control, PI3K/AKT and WNT signaling pathways. The possible synergistic effects of HBV in hepatocarcinogenesis warrant further investigations.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 3769-3769
Author(s):  
Vikki Rand ◽  
Stephen Johnstone ◽  
Rachel E Crossland ◽  
Sarah Wilkinson ◽  
Andrew G Hall

Abstract Despite significant advancements in the treatment of paediatric B-cell acute lymphoblastic leukaemia (B-ALL), ALL remains one of the most challenging adult malignancies. Outcome of adult B-ALL is poor with only 40% 5-year event-free survival compared to >80% in children. B-ALL is characterised by the acquisition of chromosomal abnormalities and many are strong predictors of outcome. The difference in the prevalence of cytogenetic subtypes and specific genomic abnormalities observed between adult and childhood ALL suggests a difference in tumour biology that may contribute to the differences in patient outcome. Detailed analysis of the paediatric B-ALL genome have revealed a plethora of abnormalities targeting key pathways. Although specific alterations have been investigated in adult and adolescent B-ALL, studies of the genomic landscape remain scarce. In this study we set-out to define the genomic landscape of adolescent and adult relapsed B-ALL. Genomic backtracking analysis of sequential diagnostic and relapse samples revealed known and novel abnormalities that may play a role in chemoresistance and disease progression in these tumours. DNA was isolated from the diagnostic and relapse samples from 12 adolescents/adult patients (5 female and 7 male) diagnosed with B-ALL. Eight of the 12 patients had remission samples available. Known cytogenetic abnormalities were detected in 7 patients: high hyperdiploid, t(1;19), t(8;14) and t(4;11) rearrangements. Two cases were positive for the BCR-ABLfusion gene. The mean age at diagnosis was 36.8 years (range 16-59 years) of which 10 relapsed early, within 2 years of initial diagnosis. DNA for the 12 diagnostic, 12 relapse and 8 remission sample were hybridised to the Affymetrix SNP6.0 array to determine copy number abnormalities (CNAs). The mutational landscape was captured for 4 cases using the Agilent SureSelect Human All Exon V4+UTR kit and sequenced to depths of 200X. The incidences of the most prevalent abnormalities in paediatric B-ALL were determined in each adult/adolescent sample: CDKN2A/B 88% (21/24), IKZF1 20% (5/24), PAX5 8% (2/24), ETV6 0% (0/24), RB1 8% (2/24), BTG1 8% (2/24) and EBF1 17% (4/24). Deletions of CDKN2A/B were detected in all but one patient. In 9 cases the abnormality was seen at both diagnosis and relapse and one case had a de novo deletion at relapse. A further case had a sub-clone harbouring CDKN2A/B deletion at diagnosis that emerged as the dominant clone at relapse. Deletion of CDKN2A/Bhas been associated with poor overall survival and has been reported at high incidence in relapsed adult BCR-ABL1-ALL, but the association with prognosis and relapse in other subtypes has not been confirmed. Genomic backtracking analysis of the matched diagnostic and relapse samples identified, on average, 36 somatic mutations at relapse that were either not detected or were only detectable in a sub-clone at diagnosis. An average of 0.05 mutations per Mb were computationally predicted to be damaging to the function of the protein. Novel de novo mutations seen at relapse were identified in cancer-related genes: FAT4, CDCA7 and PVRL4. Sequencing at depth of >200X demonstrates the ability to detect mutations in the resistant clone which could be involved in disease progression. Mutations in the ATP-binding cassette transporter gene, ABCC9, were identified in a sub-clone at diagnosis at a variant frequency of 5% (13/268 reads) and at 43% (113/260) in the relapse sample. ABCC9is involved in drug resistance suggesting a potential role in chremoresistance in this patient. In conclusion, in-depth genomic analysis and whole-exome sequencing of matched diagnostic and relapse samples in adult/adolescent B-ALL has identified known and novel genomic abnormalities. Deletion of CDKN2A/B was prevalent in 11 of the 12 cases confirming the importance of this region in relapsed B-ALL. We have identified novel mutations in genes associated with chemoresistance and tumorigenesis: ABCC9, FAT4, CDCA7 and PVRL4. Our study provides the most comprehensive genetic portrait of adult relapsed B-ALL to date and is a significant step to defining the genetic causes of disease progression and chemoresistance. Disclosures No relevant conflicts of interest to declare.


2008 ◽  
Vol 136 (3-4) ◽  
pp. 187-192 ◽  
Author(s):  
Tatjana Stankovic ◽  
Eliot Marston

Acute lymphoblastic leukaemia (ALL) is the most common paediatric cancer. Despite cure rates approaching 80%, resistance to treatment and disease relapse remain a significant clinical problem. Identification of the genes and biological pathways responsible for chemoresistance is therefore crucial for the design of novel therapeutic approaches aiming to improve patient survival. Mutations in the membrane transporter P-glycoprotein genes, genetic variations in drug-metabolising enzymes and defects in apoptotic pathways are mechanisms of chemoresistance common to a wide spectrum of cancers and also play a role in paediatric ALL. In addition, several recent microarray studies have identified transcriptional profiles specifically associated with chemoresistance and pointed to a number of potentially novel therapeutic targets. These microarray studies have shown that genes discriminating between clinically responsive and resistant leukaemias tend to be involved in cellular processes such as regulation of cell cycle, proliferation, and DNA repair. Here we review the outcomes of these microarray studies and also present our own investigations into apoptotic resistance to DNA double strand breaks (DSBs) in paediatric ALL. We present stratification of paediatric ALL by the profile of DNA damage response following ionising radiation (IR) in vitro. This approach allows classification of ALL tumours at presentation into IR-apoptotic sensitive and IR-apoptotic resistant. Furthermore, apoptotic resistant leukaemias exhibit abnormal response of NFkB pathway following irradiation and inhibition of this pathway can sensitise leukaemic cells to IR-induced DSBs.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 4268-4268
Author(s):  
Vollter Anastas ◽  
Weixin Wang ◽  
Susan Price ◽  
Zhen Zhao ◽  
V. Koneti Rao ◽  
...  

Abstract RAS-Associated Autoimmune Leukoproliferative Disorder (RALD) is a chronic indolent condition that typically presents in childhood with monocytosis, lymphocytosis, autoimmune phenomena, splenomegaly, and variable lymphadenopathy. RALD is characterized by somatic mutations in KRAS or NRAS in hematopoietic cells (myeloid and lymphoid), resulting in missense mutations involving the 12th or 13thcodon. Despite the indolent course of RALD, it shares some overlapping clinical features and similar genetic defects with Juvenile Myelomonocytic Leukemia (JMML), which is characterized by canonical and non-canonical RAS-pathway mutations. All RALD patients have normal bone marrow karyotypes. While patients with JMML often harbor monosomy 7 or other karyotypic abnormalities, normal karyotypes are also not uncommon in JMML. Optimal therapeutic management of RALD and JMML differs significantly, necessitating accurate diagnostic discrimination of RALD and normal karyotype JMML. Recent JMML studies have characterized the genomic landscape of JMML, which involves additional recurrent mutations that are thought to cooperate with RAS-pathway mutations driving the malignant phenotype of JMML. We sought to evaluate the genomic landscape of RALD for the presence of non-RAS cooperating mutations reported in JMML. Using targeted sequencing for 54 genes associated with myeloid malignancies, amplicon libraries were constructed from peripheral blood mononuclear cells (PBMCs) collected from 11 patients diagnosed with RALD (6 with previously identified KRAS and 5 with previously identified NRAS mutations) and sequenced with an average read depth of ~3000x. Variants were identified using the Illumina miSeq reporter software and analyzed using Illumina Variant Studio. Variants were filtered based SIFT, PolyPhen, and CADD predictions, ClinVar annotations, and mutations previously identified in JMML. Over 90% of the mutated genes reported in JMML sequencing reports, were evaluated (including KRAS, NRAS, CBL, PTPN11, SH2B3, SETBP1, GATA2, RUNX1, ASXL1, EZH2, DNMT3A, and ZRSR2). Of note, only the KRAS or NRAS mutations, initially identified from Sanger sequencing, were identified as disease-contributing and no other known pathogenic mutations were detected in the RALD cohort. Variant allele frequencies (VAFs) of KRAS and NRAS mutations in RALD ranged from 32.3%-50.8% with a median VAF of 46.6%. In order to further investigate genes not included in the targeted sequencing panel that may be contributing to disease, whole exome sequencing (WES) was performed on PBMCs from 7 RALD patients (4 KRAS patients, 3 NRAS patients). Despite the expanded genomic sequencing in WES (mean coverage of 225x), only the single pathogenic KRAS or NRAS mutation, known to be disease-related, was identified. No other mutations that were previously identified in JMML were detected in the WES data set. Based on presence of autoimmune phenomena in RALD, a multiplex inflammatory cytokine immunoassay investigating GM-CSF, IFN-γ, IL-1β, IL-2, IL-4, IL-6, IL-8, IL-10, and IL-12p70 was used to quantify cytokine levels in the plasma of 9 RALD patients and 17 healthy volunteers. The levels of IL-2, IL-6, and IL-10 were found significantly increased in RALD patients. IL-2 was 4.6 fold higher in RALD than controls (median 0.696 pg/mL vs. 0.149 pg/mL; p=0.049). Similarly, IL-6 was 10 fold higher in RALD than controls (median 6.42 pg/mL vs. 0.63 pg/mL; p=0.035), and IL-10 was 18 fold higher in RALD than controls (median 26.9 pg/mL vs. 1.49 pg/mL; p=0.018). Overall, these findings provide evidence that RALD, in our patient cohort with an indolent clinical course, is characterized by single somatic KRAS or NRAS mutations with the absence of cytogenetic abnormalities and cooperating mutations that are associated with malignancy. This suggests that in the spectrum of so-called RASopathies, RALD represents a non-malignant proliferative hematopoietic disease that is clinically distinct from JMML. The altered plasma cytokine profile in RALD may play a role in the autoimmune features of the disease and be related to sustained activation of the RAS pathway in hematopoietic cells. Further investigation is imperative to distinguish these two overlapping entities characterized by RAS pathway mutations, and to better understand the spectrum of disease from indolence to aggressive malignancy. Disclosures No relevant conflicts of interest to declare.


Antioxidants ◽  
2021 ◽  
Vol 10 (10) ◽  
pp. 1616
Author(s):  
Abdul Mannan ◽  
Zacary P Germon ◽  
Janis Chamberlain ◽  
Jonathan R Sillar ◽  
Brett Nixon ◽  
...  

Acute lymphoblastic leukaemia (ALL) is the most common cancer diagnosed in children and adolescents. Approximately 70% of patients survive >5-years following diagnosis, however, for those that fail upfront therapies, survival is poor. Reactive oxygen species (ROS) are elevated in a range of cancers and are emerging as significant contributors to the leukaemogenesis of ALL. ROS modulate the function of signalling proteins through oxidation of cysteine residues, as well as promote genomic instability by damaging DNA, to promote chemotherapy resistance. Current therapeutic approaches exploit the pro-oxidant intracellular environment of malignant B and T lymphoblasts to cause irreversible DNA damage and cell death, however these strategies impact normal haematopoiesis and lead to long lasting side-effects. Therapies suppressing ROS production, especially those targeting ROS producing enzymes such as the NADPH oxidases (NOXs), are emerging alternatives to treat cancers and may be exploited to improve the ALL treatment. Here, we discuss the roles that ROS play in normal haematopoiesis and in ALL. We explore the molecular mechanisms underpinning overproduction of ROS in ALL, and their roles in disease progression and drug resistance. Finally, we examine strategies to target ROS production, with a specific focus on the NOX enzymes, to improve the treatment of ALL.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 1426-1426 ◽  
Author(s):  
Lisa Jane Russell ◽  
Lisa Jones ◽  
Amir Enshaei ◽  
Jamie Rutherford ◽  
Stefano Tonin ◽  
...  

Abstract Deregulated expression of the type I cytokine receptor, cytokine receptor-like factor 2 (CRLF2 -d), is observed in 5% of B-cell precursor acute lymphoblastic leukaemia (BCP-ALL). It occurs via three known aberrations; a cryptic reciprocal translocation with the immunoglobulin heavy chain locus (IGH); an interstitial deletion within PAR1, resulting in the P2RY8-CRLF2 fusion; rare but recurrent CRLF2 mutation (F323C). All three result in overexpression of CRLF2, however alone it is insufficient to cause overt leukaemia. Mutations of the Janus kinase (JAK) family genes and the IL7 receptor, are recurrent (50% of CRLF2-d) and together with CRLF2 -d result in transformation of mouse BaF3 cells. We noted that outcome data from different study groups were inconsistent, with patients being classified as either high or intermediate risk. Thus, in this largest study to date, our aim was to acertain whether differences in clinical and/or genetic features between patients with CRLF2-d and involvement of either IGH or P2RY8 may define them as different disease entities and partly explain the outcome heterogeneity. Among 160 CRLF2-d patients, chromosomal analysis confirmed a high incidence of additional somatic copies of chromosomes X (39/88, 44%) and 21 (23/88, 26%) and identified that aneuploidy of chromosomes 9 (5/88, 6%) and 17 (7/88, 8%) were recurrent in both groups. From comparison of patients with IGH-CRLF2 and P2RY8-CRLF2, we noted a higher frequency of IKZF1 (79% v 37% p<0.001) and BTG1 (50% v 8%, p<0.001) deletions and greater complexity (>3 additional copy number alterations (CNA)) by MLPA (SALSA MLPA P335 kit, MRC Holland, The Netherlands) (p=0.037) among IGH-CRLF2 patients. We verified that (48/160) 30% of CRLF2 -d patients were Down syndrome (DS). In addition to CNA within the immunoglobulin loci and the genes investigated by MLPA, SNP6.0 arrays (Affymetrix, Santa Clara, USA) (n=25) and subsequent FISH screening identified recurrent CNA in the histone cluster at 6p22.2 (7/25, 28%), VPREB1 (6/25, 24%), ADD3 (14/58, 24%), BTLA (4/25, 16%), SLX4IP (12/43, 28%), SERP2 and TSC22D1 (6/53, 11%), PBX3 (16/54, 30%). A novel finding was an interstitial deletion at Xp11.4 fusing USP9X to DDX3X (9/50, 18%). A total of 137 somatic structural variants (SV) were detected by whole genome sequencing of 11 patients [tandem duplication- (n=6), deletion- (n=104), inversion- (n=14) and translocation-type (n=13)] creating an average of 12.7 validated SV per patient. Mutations were identified at an incidence of 12.8 per patient, including mutations in JAK1 and JAK2 (40%) and IL7R, CACNA1D and USH2A (n=2). Mutations in kinase genes were common and unique to all patients, a number have not been previously reported (e.g. ERBB4, TTBK1 and STK38L). Of interest, 7 patients had ≥1 point mutation in gene(s) involved in cell adhesion (e.g. NPNT, ADAM29, ITGB7, COL3A1 and USH2A). There was a significant difference in the median age and white blood cell count (WBC) between P2RY8-CRLF2 and IGH-CRLF2 (4yrs v 14yrs, p<0.001 and WBC >50x106 /L - 25% v 44%, p=0.021). Overall IGH-CRLF2 patients had a worse outcome compared to P2RY8-CRLF2 (p=0.191) with outcome heterogeneity in relation to gender (IGH females, HR 3.75, p=0.003), number of CNA (IGH <3 genes deleted, HR 3.79, p=0.047) and treatment trial (IGH have worse outcome on ALL97, HR 3.15, p=0.014) being observed. There was no heterogeneity for age (p=0.458), WBC (p=0.331), cytogenetic risk (p=1.0), MRD (p=1.0) or DS-ALL (p=0.877). In summary we have confirmed differences in age and WBC, involvement of kinase gene mutations and chromosomal gains in CRLF2-d ALL. Other findings included recurrent deletions involving TSC22D1 and PBX3, transcription factors involved in other human cancers; SLX4IP, an uncharacterized gene, frequently deleted in childhood ALL; the novel involvement of USP9X, a deubiquitinase involved in cancer development. We also identified recurrent mutations in cell adhesion genes. We showed that IGH-CRLF2 patients had a worse outcome, with heterogeneity seen between gender, number of gene deletions and treatment trial. We have shown that there are genetic and clinical differences dependent on whether CRLF2 is deregulated by IGH or P2RY8, suggesting that they should be regarded as different clincial entities in future studies. Disclosures Campbell: 14M genomics: Other: Co-founder and consultant.


2011 ◽  
Vol 223 (03) ◽  
Author(s):  
M Morak ◽  
R Joas ◽  
S Fischer ◽  
A Attarbaschi ◽  
G Mann ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document