Quantitative Whole Genome Analysis of Sequential Samples From Patients with B-CLL Identifies Novel Recurrent Copy Number Alterations Involving Critical B-Cell Transcription Factors

Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 3590-3590
Author(s):  
Adele Timbs ◽  
Sam Knight ◽  
Elham SadighiAkha ◽  
Adam Burns ◽  
Helene Dreau ◽  
...  

Abstract Abstract 3590 Genome-wide array or sequencing analyses are powerful tools for identifying genetic aberrations in cancers including leukaemias. However, the majority of these aberrations are likely to be random passenger events that do not drive clonal expansion. Currently, it is unknown whether cancers are maintained by a finite set of recurrent mutations similar for each patient or whether and to what extent malignancies are ‘personalised’, and also how molecular disease drivers evolve over time in the context of clinical intervention. The answers to these questions will determine whether future treatment modalities must be tailored according to individual and dynamic cancer characteristics. We hypothesized that differential quantitative high-resolution genome-wide array analysis of sequential samples from the same patients before treatment and at subsequent relapse would have the potential to identify emerging structural abnormalities with relevance to disease progression and/or response to treatment in a given patient. In order to test this hypothesis, we chose B-cell chronic lymphocytic leukaemia (CLL), because of its unique clinical characteristics, as our model to begin to evaluate the potential role of ‘companion diagnostics’ for this condition. We analysed DNA samples of 80 patients with CLL using a 1 million high resolution SNP array. On 34 of them, sequential pre-treatment and relapse samples were available. The raw data was analysed using the OncoSNP analysis tool designed in-house specifically for cancer samples as it enables quantification of copy number alterations (CNA) and copy neutral loss of heterozygosity (cnLOH) based on B-allele frequency plots in complex mixtures. This allows low levels of aberrations to be detected and for mosaic samples to be identified. Results were compared against the data from the Wellcome Trust Case Control Consortium, the DGA and germline DNA in selected cases. Large CNAs (>1Mb) and cnLOH (>5Mb) without deletions of 11q22.3 or 17p13.1 were identified in a third of patients. These patients had an intermediate clinical risk score that increased with the number of large CNAs. SNP array demonstrated clonal evolution in 32% of patients in the sequential sample cohort. These consisted of extension of the 13q abnormality (2), loss of the 13q deletion (1), a 10q23.1-q25.1 deletion (1), gain of 2pter-p14 (1), deletion of 2q33.1-q36.3, (1) a heterozygous deletion of 2q37.1 (1), gain of 8q22.2-qter (1), deletion of 8p (1), amplification of 8q (1), deletion of 8q (1), loss of 16p13.3 (2), mosaic deletion of 17q11.2 (1), an expansion of chromosomes carrying a 19p13.2-p13.11 gain and a 19p13.11 loss (1), deletions within 3p (1), conversion of a gain of 12p12.2-q21.31 to a copy neutral loss of heterozygosity (cnLOH) (1), deletion of 17pter-13.1 (1), increased proportion of chromosomes with the 7q33–34 deletion (1), expansion of cnLOH for 20q11.22-qter (1) and an increased number of cells with a deletion of 2q22.2-q24.1 (1) found at relapse. Importantly, most CNAs occurring at relapse were recurrent in the entire cohort implying that these are non-random events that are important in disease progression. Analysis of the minimal deleted region (MDR) of these recurrent and relapse associated CNAs revealed genes important in lymphoid development, such as NFκB2 and TRAP1 found in the alternative NFκB pathway and BLIMP1 involved in B-cell differentiation. There were also recurrent abnormalities in the region coding for SP140 which has been implicated in familial CLL. Known cancer genes were also affected by these recurrent and relapse-associated CNAs such as RND3, RIF1, RFXANK, and RHOT1, which are all members of the RAS pathway family. Using the OncoSNP program it was possible to determine that most of these emerging abnormalities were present in low numbers at diagnosis (fig. 1) suggesting that treatment does not induce the genetic alterations but may select for them. Fig .1 CNA and B-allele frequency plots of pre-treatment and relapse samples of a patient demonstrating clonal evolution. A low level of cnLOH can be seen in the B-allele frequency plot at diagnosis (a) at the end of chromosome 8 (dashed box) which becomes more apparent at relapse (b). Fig .1. CNA and B-allele frequency plots of pre-treatment and relapse samples of a patient demonstrating clonal evolution. A low level of cnLOH can be seen in the B-allele frequency plot at diagnosis (a) at the end of chromosome 8 (dashed box) which becomes more apparent at relapse (b). In conclusion, this is the first attempt to quantify CNAs in sequential leukaemia samples. The results demonstrate that recurrent and relapse associated CNAs affect genes important in B-cell development and cancer progression. Disclosures: No relevant conflicts of interest to declare.

Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 4092-4092
Author(s):  
Stephanie Poulain ◽  
Christophe Roumier ◽  
Elisabeth Bertrand ◽  
Aline Renneville ◽  
Sabine Tricot ◽  
...  

Abstract Background. Waldenstrom macroglobulinemia (WM) is a B-cell malignancy characterized by bone marrow (BM) infiltration of clonal lymphoplasmacytic cells, which produce a monoclonal immunoglobulin M. MYD88L265P mutation may be considered as a founder event because of it high frequency in WM. WM cells may acquire additional genetic hits that may potentially promote disease progression: CXCR4 or CD79B mutations, copy number variation,…TP53 is a tumor suppressor gene that functions as regulator influencing cellular responses to DNA damage. Little is known regarding TP53 alteration in WM. Our aim was to screen TP53 mutation in a large cohort of WM at diagnosis to analyze the genomic landscape of WM using targeted next generation sequencing (NGS) and genome wide single nucleotide polymorphism array (SNPa) and to identify clinical and biological characteristics. Method. BM samples of 125 WM (mean age: 67 years) were analyzed at diagnosis. Tumoral DNA was extracted following CD19 B cell selection. TP53 mutations were analyzed by targeted NGS to scan the coding exons of TP53. MYD88L265P, CD79A, CD79B, and CXCR4mutations were analyzed by sanger sequencing and/or NGS. Genome-Wide Human SNP Array 6.0 (Affymetrix chips) was performed in 62 cases. CN-LOH (copy neutral- loss of heterozygosity) and CNA (copy number aberration) were mapped using console 3.02 software (Affymetrix). Flow cytometry was performed to assess P53 and p21 expression after nutlin3a exposition to characterize functional mutant of TP53. Viability and cell growth of treated cells were determined using the MTS assay. Results. We have identified TP53 mutations using NGS in 7.3 % of WM (6 non-sense, 3 frameshift mutations located in the DNA binding domain) (TP53mut WM). The mutation load of TP53 varied from 13% to 98.9% (mean: 62.0%) using the variant allele frequency in NGS. We next examined the effects of nutlin-3a which is an mdm2 inhibitor on WM patients CD19+ cells genotyped for TP53 mutation. Nutlin-3a increased the expression of p53 and p21 in TP53Wild WM patients using flow cytometry (n=6). In contrast, in TP53MutWM cells, no significant variation of p53, p 21 and viability using MTS assay was observed suggesting the presence of functional mutation of TP53. The minimal deleted region of 17p in 17p deleted (TP53Del) samples was mapped using SNP array and contained 79 genes, among which was systematically comprised the loss of TP53. A high correlation between TP53 mutation and deletion 17p (p<106) was observed. One case of CN-LOH was observed at TP53 locus (1,6% of cases). Overall, we have identified alteration of TP53 locus including mutation, deletion and copy neutral loss of heterozigosity in 11, 2% of WM. Using SNP array, we found a relationship between deletion 17p, alteration of TP53 locus including mutation, UPD or del17p (TP53Alt) and TP53Mutand a greater frequency of genomic aberrations in WM compared toTP53wild (p=0.01, p=0.024 and p=0.06 respectively). A higher frequency of WM patients with more than 3 CNA identified by SNPa was observed in TP35Mut group (p=0.03) and del17p group (p<0.00001). No association was observed between TP53Mut and CXCR4 and MYD88mutations. We thoughtto identify clinical and biological characteristics of WM according to TP53Mutand/or Del17pfeatures. With a median follow-up of 5 years, 33 (26%) patients had died. 69% of cases were treated. Front line therapy included rituximab-based regimens in 76%, alkylating agent in 78%, fludarabine in 6%. The WM with TP53alteration, irrespective of TP53Mut or del17p, displayed features of adverse prognosis in regards to higher serum levels of b2m (89% versus 40%, p=0.012), and also greater IPSSWM score 2 and 3 (50% versus 30%, and 43% versus 30%, p=0.041, respectively). Importantly, the presence of TP53alteration, irrespective of TP5Mut or del17p, was associated to poor outcome in overall survival in our series, TP53alteration (p=0.003), del17p (p=0.002), and TP53Mut(p=0.015). TP53 alteration prognostic value was independent of CXCR4 or MYD88L265Pmutations. Conclusion: A low frequency of TP53 mutation was observed in WM at diagnosis. We identified a genomic signature associated to their presence. In addition, a pejorative prognostic value of TP53 mutation was observed in WM highlighted the need of new therapeutic in this sub group of WM. Disclosures Leleu: TEVA: Membership on an entity's Board of Directors or advisory committees; Novartis: Honoraria; LeoPharma: Honoraria; Pierre Fabre: Honoraria; Amgen: Honoraria; Bristol-Myers Squibb: Honoraria; Takeda: Honoraria; Celgene: Honoraria; Janssen: Honoraria.


Blood ◽  
2011 ◽  
Vol 117 (25) ◽  
pp. 6876-6884 ◽  
Author(s):  
Manuel G. Afable ◽  
Marcin Wlodarski ◽  
Hideki Makishima ◽  
Mohammed Shaik ◽  
Mikkael A. Sekeres ◽  
...  

Abstract In aplastic anemia (AA), contraction of the stem cell pool may result in oligoclonality, while in myelodysplastic syndromes (MDS) a single hematopoietic clone often characterized by chromosomal aberrations expands and outcompetes normal stem cells. We analyzed patients with AA (N = 93) and hypocellular MDS (hMDS, N = 24) using single nucleotide polymorphism arrays (SNP-A) complementing routine cytogenetics. We hypothesized that clinically important cryptic clonal aberrations may exist in some patients with BM failure. Combined metaphase and SNP-A karyotyping improved detection of chromosomal lesions: 19% and 54% of AA and hMDS cases harbored clonal abnormalities including copy-neutral loss of heterozygosity (UPD, 7%). Remarkably, lesions involving the HLA locus suggestive of clonal immune escape were found in 3 of 93 patients with AA. In hMDS, additional clonal lesions were detected in 5 (36%) of 14 patients with normal/noninformative routine cytogenetics. In a subset of AA patients studied at presentation, persistent chromosomal genomic lesions were found in 10 of 33, suggesting that the initial diagnosis may have been hMDS. Similarly, using SNP-A, earlier clonal evolution was found in 4 of 7 AA patients followed serially. In sum, our results indicate that SNP-A identify cryptic clonal genomic aberrations in AA and hMDS leading to improved distinction of these disease entities.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 1098-1098
Author(s):  
Samantha JL Knight ◽  
Elham Sadighi Akha ◽  
Adele Timbs ◽  
Tariq Enver ◽  
Andrew R Pettitt ◽  
...  

Abstract Abstract 1098 Poster Board I-120 Background B-cell chronic lymphocytic leukaemia (B-CLL) is the most common form of adult leukaemia in the Western World. It is a heterogeneous disease and important biological and clinical differences have been identified. However, the molecular mechanisms underlying emergence and maintenance of B-CLL after treatment remain elusive. Array based comparative genomic hybridization (aCGH) has revolutionized our ability to perform genome wide analyses of copy number variation (CNV) within cancer genomes. Single Nucleotide Polymorphism arrays (aSNP) provide genotyping and copy number variation data and detect regions of copy neutral Loss of Heterozygosity (cnLOH) with the potential to indicate genes involved in leukaemia pathogenesis. Both technologies are evolving rapidly and emerging platforms are thought to allow high resolution (HR) of abnormalities down to a single gene level. Aim The aim of the current study was therefore to test a HR-aCGH and a HR-aSNP platform for their ability to detect large and small CNVs and regions of cnLOH in B-CLL. More specifically, we wanted to: Method We used a high resolution 244K aCGH platform and a 1Mio SNP array in parallel to test and characterize enriched B-CLL peripheral blood samples (>80% CD19+;CD5+) from 44 clinically annotated patients collected at our institution. To distinguish CNVs seen commonly in the general population the results were compared with ‘in house’ control data sets and the Database of Genomic Variants (http://projects.tcag.ca/variation/). Results Our results show that large abnormalities, already noted by FISH, were reliably identified and the boundaries of abnormalities at 11q22.3, 13q14.2 and 17p could be defined more precisely. In addition, novel and recurrent CNVs within the sample set were identified (1p33; 3p24.3; 3p14.2; 4q12; 4q13.3; 6q21; 6q27; 8p22; 10q24; 11p15.4; 11q12; 11q13.4; 11q14.1; 11q22.1; 11q23.3; 13q14.11; 14q21.1; 15q15.1; 15q25.3; 17p13.3; 17q22; 18p11.32; 18p23; 19p13.13; 19p13.12; 19p13.32; 22q11.21; 22q11.22). Interestingly, some of these abnormalities contain single gene alterations involving oncogenes, chemokine receptors, kinases and transcription factors important in B cell development and differentiation. Assessment of smaller CNVs (less then 10 consecutive oligonucleotides) also revealed recurrent CNVs involving single genes that were clustered according to function and pathways. Comparison of paired pre-treatment and relapse samples showed differences in large CNVs in 6 out of the 14 pairs with the majority being losses within the relapse sample. In particular, relapse samples contained new losses within 2q33.1-2q37.1; 4q13.2-4q13.3; 5q31.3-5q34; 7q36.3; 10q23.1-10q25.1 11q12.3 and multiple losses within 13q14.1-13q14.3. Taken together, these data indicates that genomic instability plays a role in clonal evolution and selection after treatment in at least some patients. Analysis of a bigger cohort of matched pre-treatment and relapse samples is on-going. The importance of copy neutral LOH in B-CLL has been a subject of debate. Using the 1Mio HR-aSNP, we were able to detect multiple regions of cnLOH throughout the genome. Examination of the four regions that are known to have prognostic significance when deleted identified cnLOH involving 13q11-13q34(ter) and cnLOH of 13q21.1-q34(ter) outside the FISH region. Deletions of the 17p13.1 locus including the p53 gene confer poor prognosis in B-CLL and direct treatment decisions. Interestingly, we were able to identify cnLOH involving this region in 5% of samples. In addition, we also noticed cnLOH in 17p13.2 containing genes previously implicated in cancer. The exact pathogenetic and prognostic implications of these findings remain to be established. Conclusion Using HR-aCGH and HR-aSNP we have identified novel recurrent CNVs and regions of cnLOH in patients with B-CLL. Sequential analysis of the same patients over time suggests that at least in some patients, clonal complexity and dynamics are driven by genomic instability. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 1288-1288
Author(s):  
Charles Herbaux ◽  
Guillemette Marot ◽  
Elisabeth Bertrand ◽  
Natacha Broucqsault ◽  
Sylvie Zouitna-Galiègue ◽  
...  

Abstract Abstract 1288 Background. Approximately 30% of the patients who fulfil the criteria of Waldenström macroglobulinemia (WM) are diagnosed while asymptomatic, and will not require immediate therapy; these cases are called indolent WM (IWM). However, patients with a disease-related event will be considered for therapy, these cases are called symptomatic or aggressive WM (AWM). The physiopathology of these 2 groups remains unclear, and the mechanisms of progression have not been fully understood so far. We hypothesized that a gene signature that differentiates these two categories could be identified to better understand the underlying mechanisms of progression of WM. Methods. Seventeen patients diagnosed with WM (8 IWM and 9 AWM) were included in this study. We selected tumour cells from the bone marrow (BM) using mononuclear cell isolation, then B cell enrichment (B cell isolation kit, Myltenyi-Biotec, USA). The purity was confirmed by flow cytometry. Total RNA was extracted using the Trizol method. Gene expression profiling was performed using U133A arrays (Affymetrix, USA). Gene expression was normalized using the RMA algorithm. We ranked genes by fold-change of expression levels on a first series of 11 patients (5 IWM and 6 AWM) calculated with the ‘limma’ package in R. Next, we used a supervised classification to establish a gene expression profile to distinguish IWM from AWM. Therewith, we validated this profile on an independent set of 6 patients (3 IWM and 3 AWM). We then performed a pathway analysis using Ingenuity® analysis software. We confirmed gene expression deregulation with qRT-PCR on 3 candidate genes in the first series of patients. Genome-wide detection of copy number alteration and loss of heterozygosity were performed on 13 of the 17 WM cases, using the Genome-Wide Human SNP Array 6.0 (Affymetrix, USA). Finally, we investigated the functional consequences of the deregulation of these candidate genes in BCWM1 and MWCL1, both B cell lines originated from WM. Survival was studied using a colorimetric method with MTS (Promega, USA). Proliferation was analyzed using incorporation of a nucleoside analog (EdU) into DNA during active DNA synthesis (Invitrogen, USA). Results. The differential analysis has identified 82 probes, corresponding to 48 genes, significantly deregulated and capable of differentiating samples from IWM and AWM in an unsupervised classification. Moreover, with a supervised classification, this gene expression profile accurately classified 94% of the 17 WM samples, including the 6 WM of the independent validation set. The two molecular networks that appeared to play a major role in the physiopathology of IWM versus AWM were the plasma cell differentiation pathway and the AKT pathway. We have then identified 3 key genes in those 2 pathways, BACH2 and CIITA on the one hand and PTEN, respectively. We have then confirmed the deregulation of these gene expression levels by qRT-PCR in 3 IWM and 4 AWM; these 3 genes were over-expressed in IMW relatively to AMW. BACH2 is a B-cell-specific transcription factor known to be a tumour suppressor gene. It was shown that BACH2 reduces proliferation and induces cell death when over-expressed in B lymphoma tumour cells. We have thus pharmacologically over-expressed BACH2 in BCWM1 and MWCL1 and significantly reduced the proliferation and the survival of the two cell-lines. Further studies using BACH2 specific overexpression with lentiviral infection are underway, in vitro. The data will be presented at ASH. In order to further study the mechanisms of deregulation of BACH2 in IWM and AWM, we have conducted a genome wide SNP array study of 13 patients. Among those, 7 patients (4 IWM and 3 AWM) demonstrate a deletion of long arm of chromosome 6 (del6q), the most frequent chromosomal abnormality in WM. BACH2 gene is located on the 6q15 locus. Interestingly, we found that 3 out of the 3 AWM had a del6q that took in the 6q15 region, whereas 3 out of 4 of the IWM had a del6q preserving the 6q15 region. Therefore, haploinsufficiency could participate in the under-expression of BACH2 in aggressive WM; this hypothesis will be verified by using DNA qRT-PCR of BACH2. Conclusion. To the best of our knowledge, we have identified for the first time a specific gene expression signature that differentiates IWM and AWM. We have exposed several genes from this dataset, including BACH2, which is a candidate to better understand the underlying mechanisms of progression of WM. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 297-297 ◽  
Author(s):  
Stephanie Poulain ◽  
Christophe Roumier ◽  
Charles Herbaux ◽  
Aline Renneville ◽  
Elisabeth Bertrand ◽  
...  

Abstract Abstract 297 Background. Current models of cancer progression are based on evolution and clonal selection i.e. evolution of tumour cells inducing expansion of cells that acquire genetic lesions over time, related to ongoing mechanisms of genomic instability. An initiating event may be followed by gene mutation, copy number alteration (CNA) or copy neutral loss of heterozygosity (CN-LOH) that drive tumor progression and emergence of mechanisms of resistance to drugs. WM is a lymphoproliferative disorder characterized by bone marrow (BM) infiltration of lymphoplasmacytic cells that secrete monoclonal IgM antibody. The clinical course is characterized either by an indolent or smoldering status or a symptomatic profile that needs chemotherapy to control the progression of tumour cells. The majority of patients (pts)evolved from indolent to symptomatic, and the mechanisms of progression of WM are not fully understood to date. We hypothesized that we could gain insights into clonal evolution underlying disease progression of WM on a paired serial analysis of WM samples using genome wide SNPa, that allow both the detection of LOH and CNA. Method. BM samples of 19 untreated pts with WM (12 males, mean age: 67 years, 11 symptomatic pts) were analysed. All patients had Genome-Wide Human SNP Array 6.0 (Affymetrix chips) on at least two sequential tumor samples > 6 months apart (42 samples from 19 patients with two to three points). Tumoral DNA was extracted following CD19 B cells selection. Paired samples (tumor/normal T lymphocytes) were used as an intra-individual reference to identify germline polymorphisms. Size, position and location of genes were identified with UCSC Genome Browser HG18 assembly, LOH and CNA using genotyping console 3.02 software (Affymetrix). FISH analysis was performed to detect deletion 6q; 13q14, 11q22, TP53, trisomy 4 and 12 chromosomal aberrations using Vysis probes. P53 and MYD88 mutation were analyzed by sanger sequencing. Results. At initial sampling, SNPa detected a total of 76 CNA genetic aberrations (range 0 – 24 per genome) including 22 gains and 54 losses; 85% of patients had MYD88 L265P mutation. During the follow-up of all indolent WM that remained indolent, we haven't observed any new genetic aberrations, gain or loss (either CNA or CN-LOH) (n=8, mean follow-up: 63 months, range: 16–107 months). Among the 11 remaining patients (mean follow-up: 44.2 months, range: 7–92 months), a clonal evolution was observed in 6 cases. Three of them were symptomatic and acquired CNA or CN-LOH during clinical evolution: one case with acquisition of several CN-LOH, including one at 3p22 involving MYD88 locus associated to a mutation, one case with an additional deletion 7q31 which was observed in a patient with a chromothripsis at diagnosis, one case with emergence of subclonal del17p. The two remaining patients evolved from smoldering to symptomatic with a major variation in CNA number. Finally, loss of CNAs displayed at diagnosis was observed in one patient after chemotherapy and during indolent follow-up. In 5 cases, no detectable CNA changes were identified between the initial and subsequent sample at relapse. Conclusion. Our results using high resolution SNP array support the hypothesis that a symptomatic WM disease will favour genetic clonal evolution of tumor cells. In our study, 3 distinct genotypic patterns were observed: (i) absence of genomic variation in stable smoldering WM disease or in symptomatic relapsed patient. (ii) unstable genotype in symptomatic or asymptomatic patients switching to symptomatic suggesting high risk tumors that are less stable and more prone to change with time. (iii) loss of abnormalities suggesting that chemotherapy eradicated the dominant clone or that a genetically distinct relapsed clone has emerged. This study refines our understanding of the dynamic genetic changes in progressive WM. Further confirmation of the role of some candidate genes is underway. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 4387-4387
Author(s):  
Marisol Betensky ◽  
Daria V. Babushok ◽  
Jacquelyn J. Roth ◽  
Philip J Mason ◽  
Jaclyn A. Biegel ◽  
...  

Abstract Acquired Aplastic Anemia (aAA) develops from the immune-mediated destruction of hematopoietic stem cells (HSCs) by auto-reactive T cells. Although the exact mechanisms remain incompletely defined, it is hypothesized that auto-reactive T cells target auto-antigens presented by human leukocyte antigens (HLA), leading to HSC destruction. Acquired genetic alterations resulting in clonal hematopoiesis may therefore emerge in patients with aAA as a means of evading this immune response. Well-recognized patterns of clonal cellular evolution in aAA include myelodysplastic syndrome (MDS) and paroxysmal nocturnal hemoglobinuria (PNH). Recent studies have identified acquired copy-number neutral loss of heterozygosity on the short arm of chromosome 6 (6p CN-LOH) as another recurrent abnormality. The HLA loci map to 6p, and deletions or LOH in this region result in the loss of alleles from one parental HLA haplotype. 6p CN-LOH may allow for immune escape and restoration of hematopoiesis in aAA by deleting the HLA haplotype to which the HLA-restricted auto-reactive T cell response is targeted. The effect of 6p CN-LOH on clinical outcome and further clonal evolution in aAA is not well described. Thus, the aim of our study was to investigate the frequency, evolution and clinical significance of clonal 6p CN-LOH in our institutional cohort of adult and pediatric patients diagnosed with aAA. Bone marrow (BM) or peripheral blood (PB) samples were collected for genome wide single nucleotide polymorphism (SNP) arrays (Illumina Human OMNI1 or 850K BeadChip), and analyzed for the presence of copy number abnormalities and loss of heterozygosity, with specific reference to 6p CN-LOH. Clinical outcome data were extracted from patient charts. High Resolution HLA typing information was obtained from patient charts or performed on a research basis. HLA allele frequency in our cohort was compared to that of published control populations from the National Marrow Donor Program. Quantitative comparison of HLA haplotype allele frequency in patients with 6p CN-LOH was performed by next generation sequencing to determine the identity of clonally deleted parental alleles. Our cohort was comprised of 71 patients with aAA and related conditions including PNH and hypoplastic MDS (AA (n=64), PNH (n=5), MDS (n=2); median age 14 years, range 8 months to 68 years; 52% females). A total of 68 patients (96%) had a SNP array performed. 6p CN-LOH (n=8) or acquired clonal deletion of 6p (n=1) was detected in 13% of these patients, with the prevalence significantly higher in adult versus pediatric patients (29% versus 8.3%, p < 0.045). In all patients, clones were first identified at least 6 months after initial diagnosis. The majority of patients with 6p CN-LOH (n=7) received immunosuppressive therapy (IST). Of these, 43% failed IST requiring additional therapy while the rest achieved only a partial response. Importantly, no patient with 6p CN-LOH receiving IST alone achieved complete remission at the time of analysis. The characteristics of 6p CN-LOH varied considerably in terms of the number of clones with different sized regions of LOH, the percentage of cells with LOH, and the specific HLA loci included in the LOH region. 6p CN-LOH patients in whom follow-up SNP analyses were performed exhibited stable clone size, with no new clonal abnormalities. Quantitative analysis of HLA alleles in 6p CN-LOH patients revealed many distinct functionally deleted HLA alleles. While no specific allele was recurrently lost in the 6p CN-LOH clones, several of these deleted alleles occurred at increased frequency in our overall aAA cohort compared to ethnicity-matched control populations. In conclusion, the results from our institutional aAA cohort confirm the prevalence rates of clonal 6p CN-LOH described previously in ethnically distinct populations, while demonstrating an increased frequency of 6p CN-LOH in adult versus pediatric aAA patients. None of the aAA patients with 6p CN-LOH in our cohort achieved a complete response to IST alone, suggesting that 6p CN-LOH may be a risk factor or marker predicting poor response to IST. Taken together, our findings underscore the critical importance of understanding the mechanisms underlying clonal evolution in aAA and their effects on disease pathogenesis and response to therapy. Future multi-institutional studies are needed to define optimal therapeutic approaches for aAA patients with 6p CN-LOH. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 971-971
Author(s):  
Motohiro Kato ◽  
Masashi Sanada ◽  
Itaru Kato ◽  
Yasuharu Sato ◽  
Junko Takita ◽  
...  

Abstract Abstract 971 NFκB is a tightly regulated transcription factor of lymphocyte activation, proliferation and development. Controlled activity of NF κ B signaling pathway plays critical roles in coordination of immune and inflammatory response. Constitutive NFκB activation is recognized as a key pathological feature in several subsets of B-cell malignant lymphoma, and it is well known that lymphoma frequently occurred in association with chronic inflammation. Recently, our group showed frequent inactivation of A20, a negative regulator of NF κ B, in B-lineage malignant lymphomas. However, the molecular mechanism underlying the aberrant NF κ B activation in lymphomagenesis has not fully understood. In this study, to clarify the genetic basis of the aberrant NFκB activation, we performed genome-wide analysis of copy number alterations as well as allelic imbalances of primary B-lineage lymphoma specimens using Affymetrix GeneChip 250K genomic microarray with the CNAG/AsCNAR algorithm. We also searched for possible mutations in CARD11, CYLD, IKK and TRAF family genes and IκB genes, to obtain comprehensive registry of lesions in genes regulating NFκB pathway. This study included 238 primary lymphoma samples, including 64 samples of diffuse large B-cell lymphomas (DLBCL), 52 of follicular lymphomas (FL), 35 of mantle cell lymphomas (MCL), and 87 of mucosa-associated tissue (MALT) lymphomas. Five Hodgkin lymphoma-derived cell line (KM-H2, L1236, HDLM2, RPMI1666, L540) was also analyzed. Through a genome-wide analysis, we identified that each histology type had a unique genomic signature, suggesting a distinctive underlying molecular pathogenesis for different histology types. In contrast to the fact that A20 mutation was highly associated with loss of heterozygosity at 6q23.3, mutations of CARD11 (5 cases of DLBCL, 2 cases of MALT lymphoma) and IκB family genes (2 cases of DLBCL and 1 cases of MALT lymphoma) had no association with copy number abnormality at the locus of the genes. In total, mutations and copy number alterations in genes regulating NFκB pathway were found in more than 40% of B-cell lymphomas, which underpinned the importance of aberrant NFκB activation in lymphomagenesis. To also assess the role of uncontrolled signaling of NFκB pathway in lymphomagenesis, we re-expressed wild-type A20 in two lymphoma-derived cell lines without normal functional A20 alleles (KM-H2 and L1236). In both cells, re-expression of wild-type A20 resulted in suppression of cell growth and induction of apoptosis, accompanied by down-regulation of NFκB activation. The A20-deficient KM-H2 stably generated tumors in immunodeficient mice, whereas the tumorigenicity was effectively suppressed by re-expression of A20. We further investigated the role of A20 inactivation during clonal expansion of lymphoma by competitive proliferation assays using A20-deficient lymphoma-derived cell lines with or without re-expression of A20. The proportion of A20-expressing cells gradually decreased during competitive cell culture, and A20-expressing cells outgrew control cells in NOG mice, indicating the importance of A20 inactivation in clonal evolution of lymphoma. We demonstrated that uncontrolled NFκB signaling caused by alterations of multiple genes is a common feature of B-lineage lymphomas. Considering the physiological function of NFκB activation induced upon a variety of upstream stimuli, our observations provide an intriguing insight into and the pathogenesis of lymphoma. Our study also indicated that NFκB inhibition may have a role in lymphoma therapeutics. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 1964-1964
Author(s):  
Jennifer Edelmann ◽  
Eugen Tausch ◽  
Johannes Bloehdorn ◽  
Thorsten Zenz ◽  
Kirsten Fischer ◽  
...  

Abstract Genomic abnormalities have strong prognostic impact in chronic lymphocytic leukemia (CLL). However, clonal evolution has been studied in a limited number of cases and not within the setting of current standard therapy. It was therefore our aim to study changes in the composition of copy number alterations (CNA) over time with and without the influence of chemo(immuno)therapy. Sequential samples of 92 patients enrolled on the CLL8 trial of the GCLLSG were analyzed by Affymetrix® 6.0 single nucleotide polymorphism (SNP) arrays. Since procurement of a relapse sample was a prerequisite for this study, the cohort was not representative for the CLL8 trial [21% CR (N=19), 63% PR (N=58), 13% non response (N=12), 3% missing response (N=3)]. 48 patients received Fludarabine / Cyclophosphamide (FC), 44 patients FC plus Rituximab (FCR). Samples were taken at 3 time points: pre-treatment [N=27], time of first treatment in CLL8 [N=92] and post treatment at relapse / progressive disease [N=74]. The median observation period between samples was 35 months [range: 6-127] for the pre-treatment vs. first treatment and 41 months [range: 5-87] for the first treatment vs. post treatment (relapse) comparisons. The majority of cases maintained genomic stability over time. This applied in particular for the comparison between pre-treatment and first treatment [N=21 of 27; 78%] but also for the comparison between first treatment and relapse [N=49 of 74; 66%]. The cohort was characterized by a high proportion of high-risk genomic abnormalities [30% del(11)(q22.3), 22% TP53 loss and/or mutation in the pre-treatment cohort; 36% del(11)(q22.3), 24% TP53 loss and/or mutation in the post treatment cohort]. The acquisition of novel clonal CNAs was associated with these: In the pre-treatment to first treatment comparison [N=27], only 6 [22%] cases had novel CNAs emerging over time and all of them carried an ATM loss by del(11)(q22.3). Eight cases with ATM and/or TP53 alteration and all cases without high-risk genomic aberrations [N=13] showed no evidence of clonal evolution. In the first treatment to relapse comparison [N=74], 25 [34%] cases had newly acquired CNAs at relapse and 20 of them [80%] carried high-risk genomic abnormalities [44% del(11)(q22.3), 36% TP53 loss and/or mutation]. Only 5 cases lacking ATM or TP53 alteration had newly acquired CNAs. In contrast, genomic stability was observed in 25 cases with high-risk genomic abnormalities [15 cases with del(11)(q22.3), 10 with TP53 loss and/or mutation) and 24 cases without high-risk abnormalities. No statistically significant increased incidence of clonal evolution was observed in IGHV unmutated cases [N=66 of 92] [24 (80%) cases with clonal evolution vs. 42 (70%) cases without clonal evolution, p=0.3]. Nine patients had samples available from all 3 time points. While mainly genomic stability could be observed prior to treatment, 3 cases acquired 3 novel CNAs each after therapy. Comparing both treatment arms in the post treatment cohort [N=74] revealed a higher incidence of clonal evolution after treatment with FCR [FCR: N=16 of 35, 46%; FC: N=9 of 39, 23%; p=0.04] at a similar median observation period [36 and 35 months, respectively]. Also, the mean number of newly acquired CNAs at relapse was higher in the FCR treated group [3.3 vs. 2.6]. With regard to response no statistically significant differences were observed between cases with and without clonal evolution [cases with clonal evolution: 6 (24%) CR, 17 (64%) PR, 2 (8%) non-response; without: 8 (17%) CR, 31 (66%) PR, 8 (17%) non-response, p=0.5]. Loss of clonal lesions was rare, occurring only under the selection pressure of therapy: 16 CNAs in 11 cases were not observed anymore at relapse. 3 of these CNAs were subclonal at time of first treatment [10-20% allelic burden] and might not yet have re-emerged after relapse. 5 CNAs in 4 cases were lost at relapse. The remaining 8 CNAs were del(13q) [N=5] and del(11q) [N=3] that were lost for a probably more advantageous del(13q) / del(11q) clone. The appearance of a more advantageous del(13q) / del(11q) clone was linked to a larger deletion size [N=3] or a larger discontinuous deletion very likely resulting from chromothripsis [N=3]. The results of this study support previous data of a high genomic stability in CLL cases lacking alterations of TP53 and/or ATM. However, application of chemo(immuno)therapy did slightly increase the number of cases acquiring novel clonal CNAs. Disclosures Stilgenbauer: Pharmacyclics, Janssen: Honoraria, Research Funding.


2007 ◽  
Vol 14 (2) ◽  
pp. 483-492 ◽  
Author(s):  
Yasuhiko Nagano ◽  
Do Ha Kim ◽  
Li Zhang ◽  
Jill A White ◽  
James C Yao ◽  
...  

Pancreatic endocrine tumors (PETs) are uncommon and the genetic alterations in these indolent tumors are not well characterized. Chromosomal imbalances are frequent in tumors but PETs have not been studied by high-density single nucleotide polymorphism (SNP) array. We used genome-wide high-density SNP array analysis to detect copy number alterations using matched tumor and non-neoplastic tissue samples from 15 patients with PETs. In our study, whole or partial loss of chromosomes 1, 3, 11, 22 was present in 40, 47, 53, 40% of tumors respectively, and gain of chromosomes 5, 7, 12, 14, 17, and 20 was present in 47, 60, 47, 53, 53, and 47% of tumors respectively. One tumor had loss of heterozygosity of chromosome 3 and another of chromosome 22 without copy number alterations, suggesting uniparental disomy due to non-disjunction and deletion or to chromosomal recombination. Chromosomal aberrations of the autosomal chromosomes were correlated with chromosomal loss or gain of other chromosomes (r>0.5, P<0.5). About 60% of PETs had high allelic imbalances (AI) defined by more than four chromosomal aberrations, and 40% of tumors had low AI. The PETs with high AI were larger: the mean tumor size with high AI was 5.4 ± 3.1 cm compared with 2.3 ± 1.3 cm for low AI (P = 0.03). Our study shows that genome-wide allelotyping is a powerful new tool for the analysis of AI in PETs.


Sign in / Sign up

Export Citation Format

Share Document