Novel Germline Genetic Variants Associated with Familial Chronic Lymphocytic Leukemia (CLL)

Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 465-465
Author(s):  
Jennifer R Brown ◽  
Michael S Lawrence ◽  
Megan Hanna ◽  
Bethany Tesar ◽  
Petar Stojanov ◽  
...  

Abstract Abstract 465 CLL is among the most heritable of all cancers. To understand the genetic basis of this heritability, we have undertaken a comprehensive genomic analysis of familial CLLs including copy number analysis, gene expression profiling (GEP) and whole exome sequencing (WES). First, we examined whether familial and sporadic cases differ in the spectrum of acquired somatic mutations by WES of tumor and germline DNA of 36 familial CLLs (from 31 affected families). Compared to 55 sporadic CLLs, we observed that the somatic mutation rate in the familial CLLs was similar (mean 0.89 mutations/Mb (range 0.29–3.06) for the sporadics vs mean 0.97/Mb (range 0.11–3.78) for the familials, p=0.40). We also examined the spectrum of somatic mutations by testing for enrichment of 9 recently identified putative tumor drivers in our large CLL sequencing study (reported elsewhere in this meeting). We observed a similar distribution of these recurrent CLL mutations among the 36 familial CLLs as the 55 sporadic CLLs. These results were further confirmed by genotyping of the CLL driver mutations in an additional 32 familial and 67 sporadic CLLs. Collectively, these studies suggest that while the predisposing germline events may differ between familial and sporadic CLL, the spectrum of mutations and pattern of mutagenesis appear similar in the established CLL tumors. We therefore proceeded to examine the genetic characterization of germline DNA to identify predisposing loci, which we hypothesized might be enriched in a familial disease context. We first examined germline copy number variations (CNVs), which have not been previously characterized in this disease. We used high resolution Affymetrix 6.0 SNP arrays to study both tumor and germline DNA of 58 individuals representing 44 different families with CLL and lymphoproliferative disorders (LPDs). We identified two families (A and B) with autosomal dominant inheritance of CLL who carried distinct germline CNVs that affect genes previously implicated in CLL. Members of Family A carried a 525 kb germline deletion targeting DLEU7 at 13q14, but not affecting DLEU2, miR-15a, or miR-16–1. Importantly, by examining the tumor genome from these family members, we observed a uniform loss of the second allele of DLEU7 in 2/2 available CLLs from this family, suggesting an acquired “second hit” of a tumor suppressor gene. These findings underline the complexity of the most common somatically acquired copy number aberration (CNA) in CLL, 13q14 deletion, by demonstrating the role of additional regions other than the heavily investigated miRNA cluster. Members of Family B carried a 720 kb germline gain of 6p25 affecting the IRF4 gene, previously implicated in CLL through the identification of a GWAS risk allele located in the 3' UTR of IRF4, as well as the recent description of a recurrent somatic mutation affecting 1.5% of CLL cases. In Family B, the coding regions of the four genes located in this 6p gain, namely IRF4, DUSP22, EXOC2 and HUS1B, were sequenced, and no somatic mutations or novel SNPs were identified. However, the 6p gain in Family B represents an allele-specific enrichment of the haplotype carrying the GWAS risk SNP and, as previously described for that allele, results in lower expression of IRF4 in the two CLLs tested in this family. GEP further identified a signature associated with 6p gain that preserved low expression of IRF4 and showed high expression of KLF6. These results demonstrate that germline CNVs may facilitate the “path to cancer” by providing either an allelic deletion of a tumor suppressor or an amplification of a risk allele. As most familial CLL cases have not been accounted for by known SNPs or germline CNVs, we have initiated an in depth analysis of the WES germline results from familial cases compared to both sporadic CLL patients and normal individuals. Candidate variants have been filtered to exclude all SNPs described in the 1000 Genomes project and to focus on highly conserved sites. Thus far we have found that rare germline variants in patients with familial CLL contain a rich source of loci with relevance to B cell biology. Studies in progress are focused on further analysis of informative families and functional analyses of candidate variants. These comprehensive genomic analyses are expected to identify multiple cooperating genetic mechanisms that contribute to CLL pathogenesis, including CNVs and somatic and germline mutations. Disclosures: No relevant conflicts of interest to declare.

Blood ◽  
2020 ◽  
Vol 136 (Supplement 1) ◽  
pp. 25-26
Author(s):  
Michelle Nash ◽  
Adrianna Vlachos ◽  
Marcin W. Wlodarski ◽  
Jeffrey Michael Lipton

Background: Diamond Blackfan anemia (DBA) is a rare inherited bone marrow failure syndrome characterized by anemia, congenital anomalies and a predisposition to cancer. Patients usually present during infancy or early childhood, but can also be diagnosed as adults. In the vast majority of cases DBA is due to a mutation in a gene encoding a small or large subunit-associated ribosomal protein (RP) leading to RP haploinsufficiency. In a study of 702 patients enrolled in the DBA Registry (DBAR), the observed to expected ratio for acute myeloid leukemia (AML) was 28.8 and for myelodysplastic syndrome (MDS), 352.1 (Vlachos et al, Blood, 2018). The average age of onset for MDS in the DBA cohort was 26 years, compared to 60-70 years in the general population. Evolving clonal hematopoiesis (CH) with age has been observed as a precursor to MDS, with CH rarely observed in individuals younger than 40 years of age. Thus we hypothesized that the young age at the development of MDS in DBA would be presaged by evolving CH. Objective: The primary objective was to perform whole exome sequencing (WES) specifically screening for previously reported somatic mutations in 56 genes associated with CH (Jaiswal et al, NEJM, 2014). Design/Method: A total of 69 samples were analyzed from 65 patients, mostly targeting patients older than 18 years (median age 30 years). Multiple samples were run on patients who had available samples in the DBAR Biorepository to determine rate of acquisition of mutations. 468 age- and sex-matched healthy controls were made available from GeneDx who performed the WES for the study. We used a threshold for variant calling of minimum 5% with a minimum of 2 variant reads. Results: Three of the 65 DBA patients (5%) were found to have somatic mutations in STAG1, U2AF1, SF3B1, and DNMT3A at 8, 20, 41, and 70 years, respectively (Table 1). The patient who was 20 years of age had a sample in the DBAR biorepository from when he was age 8 years which was found to have a different somatic mutation (STAG1) than was found at present (U2AF1). This patient did go on to develop MDS at the age of 21 years. In comparison, of the 468 controls, 4 (0.8 %) had a somatic mutation in SF3B1, LUC7L2, DNMT3A, and LUC7L2 at ages 12, 31, 33 and 40 years, respectively. Conclusion: Patients with DBA show more somatic mutations as compared to controls (p<0.05). This early acquisition of mutations may be the driving force for their developing MDS at an earlier age than that of the general population. Further studies with more sensitive methods are warranted to accurately determine the prevalence of somatic CH mutations and their potential association with the development of myelodysplastic syndrome in these patients. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 823-823 ◽  
Author(s):  
Hideki Makishima ◽  
Kenichi Yoshida ◽  
Thomas LaFramboise ◽  
Bartlomiej P Przychodzen ◽  
Matthew Ruffalo ◽  
...  

Abstract Somatic mutations constitute key pathogenetic elements in MDS. Unbiased whole exome sequencing (WES) and deep NGS led to discovery of new somatic mutations and also to the recognition of i) tremendous diversity of mutations and their combinations; ii) individual intra-tumor heterogeneity and clonal hierarchy. Chromosomal lesions further increase the complexity of molecular defects. While in MDS molecular defects are acquired in order, observations made in AML highlight the importance of ancestral events; e.g., t(8;21), inv16 or t(15;17) and other lesions that are used as the basis for nosological sub-classification. Thus, it is the identity of individual ancestral events or their classes rather than the spectrum of secondary events or the distribution of mutations, that will allow for molecular, functionally-relevant and diagnostically useful classification within MDS. This would explain why only a few somatic mutations have been found to be prognostically important, as their position in the clonal hierarchy has not been accounted for. With this in mind, we applied WES (N=206) and targeted deep NGS (N=836) and studied 100 samples serially with analyses focused on ancestral events. Globally, through WES we identified and validated 2386 mutational events in 1458 genes. Of these, 112 genes were mutated at significant frequencies (q<0.05); groups of affected genes involved in splicing, transcription, DNA methylation, histone modification, and others were distinguished. On average, 9 somatic events per MDS case, 10.7 in secondary AML, and 12.5 in MDS/MPN were found. Resequencing in combination with SNP-array karyotyping provided information on variant allelic frequency (VAF) adjusted for corresponding zygosity of mutations; 99% of cases displayed clear intra-tumor heterogeneity due to multiple clones defined by hierarchically acquired somatic mutational patterns. Using cross-sectional analyses, the highest mean VAF could be interpreted as consistent with the ancestral nature of the mutations, as seen for instance in a proportion of TET2 and SF3B1 mutant cases. In contrast, the lowest mean VAF indicated secondary events, as occur in NPM1 and RAS pathway mutations. Similar conclusions were made based on cross-sectional analyses showing a similar distribution of ancestral but not secondary events in MDS and sAML. All gene mutations were categorized into those that are predominantly ancestral and those that are facultatively secondary. The most frequent founder mutations were identified (TET2, DNMT3A, SF3B1, ASXL1, TP53, U2AF1, RUNX1, SRSF2) and used to sub-classify approximately 80% of patients, with the remainder containing more infrequent ancestral mutations. While in a combined fashion (as both founder and secondary events) many of these mutations were not predictive of prognosis, they gained relevance when only cases affected by ancestral mutations were used for prognostication. Thus some of the mutations, when present as secondary events may not be predictive. Founding mutations may determine subsequent clinical and molecular features. While other frequently affected genes, SF3B1 or ASXL1, are not associated with a significant increase in the number of concomitant mutations, cases with TET2 mutations showed significantly more frequent mutations per case than those with wild-type TET2 (14.6 vs. 9.1; p=0.001). Moreover, ancestral TET2 mutations were associated with concomitant mutations due to high C-to-T transitions, possibly because reduced 5-hydroxymethylcytosine might create the specific mutator milieu. Most important is the association not of any type, but of ancestral mutations with certain pathomorphologic features and outcomes. Founding TET2 mutations are associated with MPN/MDS while secondary TET2 mutations are present in MDS. Ancestral DNMT3A mutations determine a rapid progression to AML, whereas subclonal DNMT3A mutations are also found in high-risk MDS. RAS pathway mutations are ancestral in CMML and also secondarily positive in the late stage of MDS (sAML). Specific ancestral events may determine subsequent mutational events, and while both types of mutation may affect the clinical phenotype, the initial events are less diverse and more subtype-specific. In conclusion, WES clarified the distinct landscape and ordering of the somatic mutational spectrum in MDS. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 4332-4332
Author(s):  
Sarina Sulong ◽  
Julie Irving ◽  
Marian Case ◽  
Lynne Minto ◽  
Nick Bown ◽  
...  

Abstract Genetic alterations including chromosomal translocation, promoter hypermethylation, somatic mutation and gene deletion are believed to play a key role in the leukemogenic process in childhood acute lymphoblastic leukemia (ALL). The p16INK4a (CDKN2A/MTS1/p16/INK4a) gene located on chromosome 9p21 is a tumor suppressor gene whose product can block cell division during the G1/S phase of the cell cycle. Inactivation of p16INK4a in ALL can occur by deletion, promoter hypermethylation or somatic mutation. However, published reports are inconsistent in terms of both incidence and route of p16INK4a inactivation suggesting that a detailed analysis of all possible modes of inactivation in a large cohort is essential to clarify the status of this gene in leukemogenesis. In this study, we report the findings of a comprehensive analysis of p16INK4a in 115 DNA samples with childhood ALL (86 cases at presentation and 29 cases at relapse) in which a combination of techniques including, fluorescence in situ hybridization (FISH), mapping arrays, denaturing high performance liquid chromatography (dHPLC) and methylation specific-PCR (MSP) were used to assess the mode of inactivation of this gene. Data from a genome-wide screening in 86 presentation cases and 20 of 29 relapse cases using Affymetrix Mapping 10K and/or 50K single nucleotide polymorphism (SNP) microarray technique showed loss of heterozygosity (LOH) at the p16INK4a locus in 21% (22/106) of cases (14 at presentation and 8 at relapse), 14 (8 at presentation and 6 at relapse) with an associated loss of copy number and 8 (6 at presentation and 2 at relapse) with a normal copy number, indicative of acquired isodisomy (AID). FISH analysis on 19 of the 22 confirmed that those cases with LOH and copy number loss had either p16INK4a homozygous (n=6) or hemizygous (n=6) deletion and those with LOH associated with AID (n=7) retained 2 copies. Mutation and methylation analyses were performed on those cases identified to have one p16INK4a allele or retention of both alleles. Partial methylation of p16INK4a was found in only 1 case. Mutational screening by dHPLC of the coding region revealed a somatic mutation, H83Y, in a subpopulation of leukemic blasts in one patient at relapse. Three common SNPs were identified including A148T in exon 2 and 500C&gt;G and 540 C&gt;T in the 3′ UTR. These data show that mutation and hypermethylation of p16INK4a are rare events in childhood ALL but that homozygous and hemizygous deletion is relatively common. The loss of only one p16INK4a allele in this latter group, without evidence for mutation or hypermethylation of the remaining one suggests that p16INK4a may be haploinsufficient in ALL. The finding that LOH on 9p locus is common but in nearly 40% of these cases is associated with AID with intact p16INK4a, suggests the existence of another tumor suppressor gene or oncogene in this region, which may have importance in leukemogenesis.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 2432-2432
Author(s):  
Jennifer R Brown ◽  
Bethany Tesar ◽  
Megan Hanna ◽  
Megan Ash ◽  
Stacey M Fernandes ◽  
...  

Abstract Abstract 2432 Chronic lymphocytic leukemia (CLL) is one of the most familial of all cancers but the genetic basis of this heritability remains poorly characterized. Families with very strong inheritance of CLL have been described in the literature, and recently the occurrence of CLL in one such family was associated with a polymorphism in the DAPK gene. Here we report the genomic characterization of a family in which CLL appears to be inherited in a Mendelian autosomal dominant manner. Within this family, five of eleven siblings of the first generation were affected, and one of those affected siblings had five children, of whom three were also affected (the second generation). The children of the second generation are currently aged 20–30 and hence too young to know whether they will develop CLL. We performed high-density single-nucleotide polymorphism (SNP) array analysis and gene expression profiling on tumor and germline DNA from four of the offspring of the second generation, as well as six of their children. Analysis of the SNP array data revealed a significant germline amplification of 6p, spanning 0–720 Mb and encompassing a known copy number variant (CNV) region but significantly larger than the CNV region. This amplification was found in both affected individuals with samples available from the second generation, and was transmitted by each of them to one of their two children in the third generation. This amplification was absent from the two unaffected members of the second generation, their children, or any of the other 189 individuals with CLL who were analyzed in our high-density SNP array dataset. None of the unaffected individuals with or without the amplification had evidence of monoclonal B cell lymphocytosis (MBL) by highly sensitive flow cytometry. These unaffected individuals also lacked any PCR-detectable oligoclonal or monoclonal immunoglobulin heavy chain gene rearrangement suggestive of MBL. The region of amplification contains four protein-coding genes: EXOC2, DUSP22, HUS1B and IRF4. We sequenced the coding regions of these four genes and the 5` and 3` UTRs of IRF4 in all family members, but found no somatic mutations in this family. All four genes were also sequenced in 92 other familial CLLs, identifying no somatic mutations. We then analyzed our gene expression profiling data to assess whether any genes in this region were altered in the affected individuals with the amplification. This analysis revealed a significant 1.74X increase in IRF4 expression in the CLLs with the amplification compared to those without (q value < 0.001). By Western blotting, we confirmed that IRF4 protein was increased approximately two-fold in amplified compared to non-amplified samples. These data suggest that the amplification may target IRF4, which has been previously implicated in CLL by a genome wide association study that identified a tag SNP in its 3` UTR as a CLL risk allele. Further analysis of our SNP data demonstrated allele specific amplification in this region, and mass-spectrometric genotyping confirmed enrichment of the CLL risk allele in the individuals with amplification. We conclude that amplification of IRF4 carrying the risk allele for CLL appears likely to be the culprit predisposing to CLL in this family. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 4591-4591 ◽  
Author(s):  
Timothy Geoffrey Bowler ◽  
Matthias Bartenstein ◽  
Kerry A. Morrone ◽  
Mersedeh Rohanizadegan ◽  
Rachel M. Kessel ◽  
...  

Abstract Introduction Familial MDS is a rare disease and has been associated with mutations in multiple genes including GATA2. Mixed Lineage Leukemia 3 (MLL3) encodes a histone methylase that is a tumor suppressor and implicated in poor prognosis in MDS and AML. It occurs at high frequency and across multiple tissue types in genomic surveys of somatic mutations in cancer, including reports of childhood AML and MDS. To investigate the genetic basis of MDS we analyzed the sequence variation in familial and non familial cases of MDS. Methods Exome sequencing of blood and germline tissue of a young patient with MDS with her mother and brother was performed. The patient presented with pancytopenia at the age of 15 and had monolobated megakaryocytes, macrocytosis and dysplastic appearance on bone marrow exam. Her mother and younger brother have milder variants of similar disease and have unexplained cytopenias without any nutrient deficiencies or chronic diseases. Data was also compared to other exomes performed by us and by other groups, by Illumina next generation sequencing. The raw data was processed using standard techniques (Illumina preprocessing, Burroughs Wheeler Alignment using GATKv3 via Genesifter). The lists of high quality mutations were compared to each other and shared mutations were identified. The presence of a selection of these mutations in MLL3 were validated by PCR and direct Sanger sequencing. Predicted mutations were cross-referenced to the COSMIC and TCGA databases. Results Exome sequencing revealed a intronic mutation in p53 (C>T at Chr 17 7579596) in all affected family members. We also identified a shared germline variant affecting BCL6B and a previously identified mutation in NOX5. BCL6B was homozygous for an insertion of an extra CAG at Chr 17:6928019 causing p.S235G change. BCL6B is a functional tumor suppressor, involved in early B cell development and COSMIC database lists for presence of mutations in breast, CNS, endometrium, large intestinal cancers. NOX5 was homozygous for a 6 base pair deletion at Chr 15:69,238,301 causing p.EP150del alteration. NOX5 encodes a NADPH oxidase that has been implicated in hairy cell leukemia, esophageal and prostate cancer and it thought to contribute to increased tumor proliferation. Functional studies are underway to understand the role of these variants. Interestingly, exome sequencing also identified missense mutations in the MLL3 gene (p.R284Q, p.R526P). Surprisingly, sequencing of unrelated MDS patients in our institution also identified many highly scoring, identical mutations in MLL3 (p.Y987*, p.T316S, p.P860S, p.G892R, p.D348N, p.E765G). Similar mutations have been reported at a high frequency in childhood leukemia (Leukemia. 2014 Jun;28(6); T insertion at chr7:151945072 (rs150073007)) as well as in a case of familial AML (Blood: 121 (8), 2013; T insertion at codon 817 in exon 14). These mutations occurred in clusters and match paralogous areas of the genome. Conventional PCR from within regions matching pseudogene copies appear to show heterozygous mutations. We then devised long range PCR (covering between 3 to 20 Kb) from non-conserved regions and determined that these mutant calls are false positives. Many of these mutations are identical those in catalogues of SNPs, somatic mutations in cancer are implicated in disease processes. Examination of the TCGA database reveals that MLL3 mutation has been reported at a high frequency in not only liquid tumors but also solid tumors such as melanoma (24%), lung adenocarcinoma (19%), pancreatic cancer (6%) and others. Examination of the mutations reveals that the false positive calls may be responsible for many of these calls. Conclusions We have determined novel genetic variants in a family with MDS and cytopenias. Functional studies are underway to determine the significance of these variants. More interestingly, we have determined that there is a high rate of false positive variation calls within the MLL3 gene. This is caused by systematically misattributing homologous or unassembled areas of the genome to MLL3. The significance of MLL3 as an oncogene has been misrepresented by this phenomenon. Long range PCRs from unique genomic regions are recommended to test for these mutations. The psuedogene effect can be the source of systematic bias that affects many other genes and has misrepresented their degree of natural polymorphism and correlation with malignancy. Disclosures No relevant conflicts of interest to declare.


2020 ◽  
Author(s):  
Yoo-Jin Kim ◽  
SeungHyun Jung ◽  
Eun-Hye Hur ◽  
Eun-Ji Choi ◽  
Kyoo-Hyung Lee ◽  
...  

Abstract Background: Recent advancements in next-generation sequencing (NGS) technologies allow the simultaneous identification of targeted copy number alterations (CNAs) as well as somatic mutations using the same panel-based NGS data. We investigated whether CNAs detected by the targeted NGS data provided additional clinical implications, over somatic mutations, in myelodysplastic syndrome (MDS). Methods: Targeted deep sequencing of 28 well-known MDS-related genes was performed for 266 patients with MDS. Results: Overall, 215 (80.8%) patients were found to have at least one somatic mutation; 67 (25.2%) had at least one CNA; 227 (85.3%) had either a somatic mutation or CNA; 160 had somatic mutations without CNA; and 12 had CNA without somatic mutations. Considering the clinical variables and somatic mutations alone, multivariate analysis demonstrated that sex, revised International Prognostic Scoring System (IPSS-R) and NRAS and TP53 mutations were independent prognostic factors for overall survival. For AML-free survival, these factors were sex, IPSS-R, and mutations in NRAS, DNMT3A, and complex karyotype/TP53 mutations. When we consider clinical variables along with somatic mutations and CNAs, genetic alterations in TET2, LAMB4, U2AF1, and CBL showed additional significant impact on the survivals. Conclusions: Our study suggests that the concurrent detection of somatic mutations and targeted CNAs may provide clinically useful information for the prognosis of MDS patients.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 2907-2907
Author(s):  
Lijian Yu ◽  
Haesook T. Kim ◽  
Siddha Kasar ◽  
Parul Benien ◽  
Wei Du ◽  
...  

Abstract Many studies have shown that deletion at chromosome 17p targeting the TP53 gene, or del(17p), is associated with poor prognosis in chronic lymphocytic leukemia (CLL). Despite this, not all del(17p) CLL progresses rapidly to treatment and has short survival. We hypothesized that other coexisting genetic aberrations may contribute to the poor clinical outcome and heterogeneity of del(17p) cases. To assess this, we analyzed copy number alterations (CNAs) using Affymetrix SNP array data from 200 CLL patients (55 with del(17p)), and somatic mutation profile by whole exome sequencing (WES; Illumina) in 168 patients (53 with del(17p)). Ninety-nine patients were studied with both SNP arrays and WES, 39 of whom had del(17p). Analysis of copy number showed that del(17p) CLL had a median of 11 CNA events, mostly copy number losses, compared to 2 events in WT CLL (p=1.1E-12). Both the number of CNA events (p=7.9E-10) and the total length of copy number gain (p=1.6E-4) or loss (p=1.3E-8) were associated with shorter overall survival (OS), even when controlling for co-existing del(17p). Using GISTIC analysis, we discovered three significantly deleted regions specific to del(17p), namely large chromosomal deletions at 3p, 4p, and 9p. These novel recurrent deletions were rarely seen in wild type CLL and the presence of any of these deletions with del(17p) was strongly associated with shorter OS compared to del(17p) alone. We were able to evaluate complex karyotype (CKT) determined by stimulated metaphase cytogenetics in a subset of the cohort, and found that 17/27 (63%) evaluable del(17p) CLLs had CKT, while only 13/58 (22%) WT CLLs did (p=0.0005). Analysis of OS showed that del(17p) and CKT together had worse OS than either individually, although individually they each conferred OS worse than WT (p<0.0001). Next we tested whether the number and size of CNAs predicts progression to treatment in 138 patients who were treatment naïve at sampling (n=23 for del(17p)), 65 of whom progressed to treatment after sampling (n=16 for del(17p)). We found that increasing number of CNA events (p=9E-6), total length of losses (p=4E-4), and total length of gains (p=2E-4) were all predictive of need for future treatment in WT as well as del(17p) CLL. Those treatment naive del(17p) patients who remained untreated had a median of 4 CNAs (n=7), compared to 12.5 for those who went on to treatment (p=0.013). Turning our attention to somatic mutation analysis, del(17p) CLLs had higher numbers of total somatic mutations (21 vs 18, p=0.0046), and nonsynonymous mutations (16 vs 13, p=0.0059) than WT CLL, with no difference in subclonal mutations (12 vs 10, p=0.34). Increasing number of total mutations (p=0.0017) and nonsynonymous mutations (p=0.0003) were both associated with shorter OS, even when controlling for 17p deletion. No significant correlation was observed between number of CNAs and number of somatic mutations in a given CLL, suggesting different mechanisms involved in their causation. In the del(17p) CLLs, as expected, TP53 was the most commonly mutated gene, seen in 43/53 (81%) patients. Interestingly, most TP53 mutations (78%) were clonal, suggesting they occurred early in CLL development, while SF3b1 assessed for comparison was most commonly subclonal (68%). Patients with subclonal TP53 mutations had longer OS than patients with clonal TP53 mutations (p=0.041). We also explored the outcome of patients with loss of both TP53 alleles via 17p deletion and TP53 mutation. We found that biallelic loss was associated with more CNAs (median = 15 vs 9) and longer total length of CNAs than monoallelic loss. Patients with biallelic loss of TP53 showed a trend towards worse OS than the limited cohort with monoallelic loss (p=0.07). In addition to TP53 and other previously reported CLL drivers such as NOTCH1 (n=8, q=8E-5) and DDX3X (n=4, q=0.03), we report RPS15 (n=6, q=6E-5) and GPS2 (n=3, q=0.03) as novel significantly mutated genes in del(17p) CLL but not in WT CLL. All RPS15 mutations were clustered in a 15-amino acid region in the far carboxyl terminus of the gene, suggesting conserved functions. RNA sequencing (n=2) confirmed that the mutant alleles of RPS15 were expressed in the CLLs. We conclude that the genetic profile of del(17p) CLL differs significantly from WT CLL and show that worse OS in del(17p) CLL is associated with complex karyotype and biallelic inactivation of TP53 as well as increasing number of somatic mutations and novel CNAs. Disclosures No relevant conflicts of interest to declare.


2017 ◽  
Vol 35 (15_suppl) ◽  
pp. e12518-e12518
Author(s):  
Varun Bagai ◽  
Jeoffrey Schageman ◽  
Dumitru Brinza ◽  
Yanchun Li ◽  
Jian Gu ◽  
...  

e12518 Background: With recent advances in next-generation sequencing (NGS) technologies, it is now possible to detect somatic mutations with allele frequencies in blood samples as low as 0.1% from circulating tumor DNA. A natural extension to this achievement is adding the ability to simultaneously detect copy number variants and gene fusions. A panel such as this addresses a full repertoire of variant classes found to be linked with certain tumors and would enable researchers additional tools to profile cancer samples more dynamically thus enriching current diagnostic tool sets. Here, we present progress on such an approach and apply current NGS technology to achieve our goals. Methods: Samples were sequecned using the Ion S5™ system. Results: Using control samples, we can reproducibly demonstrate detection of ERBB2 (HER2/neu) gene amplifications with high statistical significance and as low as a 2 fold difference versus non-amplified loci in titration experiments. In addition, this ERBB2 gene amplification was detected in the context of a validated breast cancer somatic mutation panel in which no negative impact was exhibited and mutation detection specificity and sensitivity were both greater than 90%. Lastly, we developed an additional panel to detect gene fusions relevant to lung cancer. Using the titration approach above, the EML4-ALK fusion variant was shown to have a limit of detection near 1% with no negative impact on detection sensitivity and specificity when combined with the validated lung cfDNA somatic mutation panel. Conclusions: From the outcomes of these experiments, we have shown the ability to reproducibly and simultaneously detect copy number and gene fusion variants as well as somatic mutations at very low limits of detection in a cell free DNA background derived from blood samples.


Author(s):  
Jeffrey A. Thompson ◽  
Brock C. Christensen ◽  
Carmen J. Marsit

Bidirectional gene promoters affect the transcription of two genes, leading to the hypothesis that they should exhibit protection against genetic or epigenetic changes in cancer. Therefore, they provide an excellent opportunity to learn about promoter susceptibility to somatic alteration in tumors. We tested this hypothesis using data from genome-scale DNA methylation (14 cancer types), simple somatic mutation (10 cancer types), and copy number variation profiling (14 cancer types). For DNA methylation, the difference in rank differential methylation between tumor and tumor-adjacent normal matched samples based on promoter type was tested by Wilcoxon rank sum test. Logistic regression was used to compare differences in simple somatic mutations. For copy number alteration, a mixed effects logistic regression model was used. The change in methylation between non-diseased tissues and their tumor counterparts was significantly greater in single compared to bidirectional promoters across all 14 cancer types examined. Similarly, the extent of copy number alteration was greater in single gene compared to bidirectional promoters for all 14 cancer types. Furthermore, among 10 cancer types with available simple somatic mutation data, bidirectional promoters were slightly more susceptible. These results suggest that selective pressures related with specific functional impacts during carcinogenesis drive the susceptibility of promoter regions to somatic alteration.


2018 ◽  
Vol 19 (8) ◽  
pp. 2296 ◽  
Author(s):  
Jeffrey Thompson ◽  
Brock Christensen ◽  
Carmen Marsit

Bidirectional gene promoters affect the transcription of two genes, leading to the hypothesis that they should exhibit protection against genetic or epigenetic changes in cancer. Therefore, they provide an excellent opportunity to learn about promoter susceptibility to somatic alteration in tumors. We tested this hypothesis using data from genome-scale DNA methylation (14 cancer types), simple somatic mutation (10 cancer types), and copy number variation profiling (14 cancer types). For DNA methylation, the difference in rank differential methylation between tumor and tumor-adjacent normal matched samples based on promoter type was tested by the Wilcoxon rank sum test. Logistic regression was used to compare differences in simple somatic mutations. For copy number alteration, a mixed effects logistic regression model was used. The change in methylation between non-diseased tissues and their tumor counterparts was significantly greater in single compared to bidirectional promoters across all 14 cancer types examined. Similarly, the extent of copy number alteration was greater in single gene compared to bidirectional promoters for all 14 cancer types. Furthermore, among 10 cancer types with available simple somatic mutation data, bidirectional promoters were slightly more susceptible. These results suggest that selective pressures related with specific functional impacts during carcinogenesis drive the susceptibility of promoter regions to somatic alteration.


Sign in / Sign up

Export Citation Format

Share Document