ABC and GCB DLBCLs Display Unique Biologically Distinct and Clinically Relevant Epigenetic Signatures.

Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 619-619
Author(s):  
Rita Shaknovich ◽  
Huimin Geng ◽  
Nathalie Johnson ◽  
Leandro Cerchietti ◽  
Maria E Figueroa ◽  
...  

Abstract Abstract 619 Diffuse Large B-cell Lymphoma (DLBCL) is a complex biological entity with heterogeneous genetic, biological and clinical features. Gene expression profiling studies have attempted to resolve some of this heterogeneity. For example, DLBCL patients harboring gene signatures associated with expression of germinal center B-cell genes (GCB) or activated B-cell genes (ABC) vary in their response rate to standard chemo-immunotherapy regimens. Since epigenetic gene regulation can play a fundamental role in determining the phenotype of normal and malignant tissues we asked whether ABC and GCB DLBCLs also display unique epigenetic signatures that might be clinically useful and further explain the biology of these tumors. For this purpose we examined the DNA methylation level of 50,000 cytosine residues distributed among 14,000 gene promoters in a cohort of 159 patients with DLBCL, all of whom were uniformly treated with R-CHOP, using the HELP assay and high-density oligonucleotide microarrays. For a subset of these patients (n=69), we also performed Affymetrix gene expression profiling. First, a Bayesian predictor of ABC/GCB subtypes was trained from a published expression profiling study of 203 DLBCL patients. The predictor was then applied to our cohort of 69 patients. At a probability cutoff of 0.9, 20 patients were classified as ABC, 40 were classified as GCB and 9 could not be classified (i.e. “type III DLBCL”). As expected from published studies, the differences in progression free survival (PFS) and overall survival (OS) of these ABC vs. GCB patients thus determined was highly significant, with p=0.0026 (log rank) and p=0.043 (log rank) respectively, with a much worse prognosis for ABC patients compared to GCB ones. We then asked whether the ABC and GCB subtypes could be predicted from the DNA methylation profiles of the same 69 patients. By performing a t-test we found that 239 genes were differentially methylated between ABC and GCB (p<0.0001) and also displayed >30% difference in methylation level. This DNA methylation signature was incorporated into a new Bayesian predictor, which we showed to predict ABC and GCB DLBCL subtypes from DNA methylation profiles with a 91% accuracy. Using a cross-validation procedure, we estimated that the classification performance on independent cases to be ∼87%. The predicted ABC and GCB cases retained the clinical predictive power of the gene expression profile when applied to the remaining 90 patients that did not have gene expression profiles, confirming its clinical relevance (difference in PFS p=0.0148, log rank). Gene set enrichment analysis showed that the ABC DNA methylation signature was enriched in genes involved in antigen dependent B and T-cell responses and in TNF inflammatory responses (p<1.01E-4 and <6.01E-4 respectively). A computational analysis of promoter DNA sequences of the genes involved in this signature revealed over-representation of binding sites for transcription factors including MYB, STAT5A, MAZ, and JUN and Sp1; many of these factors have a known role in B cell development and function. The role of Sp1 in these tumors is under further examination. Among the 239 genes that were differentially methylated and the 411 genes that were differentially expressed between ABC and GCB there was an overlap of 16 genes (greater than expected by chance Fisher Exact p=0.005). A predictor based on the methylation profiles of these 16 genes was on its own 92% accurate in identifying ABC vs. GCB cases among our cohort of DLBCLs. Although there was a general trend for inverse correlation in expression between the 239 differentially methylated genes, these 16 overlapping genes displayed marked and extreme inverse correlation. This was validated by single locus quantitative methylation sequencing (MassArray) and QPCR. The 16 genes included genes known to play critical roles in B-cell differentiation, proliferation and metabolism but not previously implicated in DLBCL. Gain and loss of function assays of a subset of these genes in ABC and GCB DLBCL cell lines show that they have tumor suppressor functions in DLBCL. Our results demonstrate for the first time that ABC and GCB DLBCLs are epigenetically distinct diseases; they identify new biological differences and candidate tumor suppressor genes between these tumors and demonstrate that a DNA methylation classifier can be used to distinguish GCB and ABC DLBCL subtypes. Disclosures: No relevant conflicts of interest to declare.

Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 355-355 ◽  
Author(s):  
Yair Herishanu ◽  
Patricia Perez-Gelen ◽  
Delong Liu ◽  
Angelique Biancotto ◽  
Berengere Vire ◽  
...  

Abstract Abstract 355 In-vitro studies suggest that chronic lymphocytic leukemia (CLL) cells depend on the tissue microenvironment. Different molecules and cell types have been reported to enhance the proliferation and survival of CLL cells. The presence of CLL cells in three distinct compartments: peripheral blood (PB), bone marrow (BM) and lymph node (LN), provides a unique opportunity to investigate the effects of the microenvironment on tumor cell biology in-vivo. To this effect, we used gene expression profiling (Affymetrix HU133 plus arrays) to compare purified CLL cells sampled from PB, BM, and/or LN from 24 previously untreated patients. Initially, an unsupervised hierarchical clustering of all samples appeared to be dominated by the effect of the individual patient. However, in 12 patients where all three sites had been sampled, we used a 3-level one-way ANOVA blocked by patients to estimate patient effect and tissue effect. Three principal components of the 36 samples revealed a clear separation of the tumor cells according to their compartment of origin. Furthermore, supervised analysis with a cutoff of >2-fold change and false discovery rate <0.2 identified 151 genes that discriminated between circulating and LN resident CLL cells (n=17), most of which were more highly expressed in LN, and 27 genes that were differentially expressed in BM as compared to PB cells (n=19). Among the genes upregulated in the lymph node many are readily recognized as related to cell proliferation (e.g. Cyclin D2 and c-MYC) or NF-κB signaling. However, to use an observer independent, unbiased discovery tool to query the gene list for the presence of functional gene signatures we used gene set enrichment analysis (GSEA) and identified several gene expression signatures that were preferentially expressed in LN resident cells: a proliferation signature characterized by E2F and c-MYC regulated genes, signatures related to B-cell receptor and NF-kB signaling were prominent (FDR for all <0.02, normalized enrichment scores 1.81-2.15). A gene expression based E2F score was highest in LN, followed by BM and weakest in PB. Increased nuclear accumulation of E2F1 and c-MYC in LN compared to PB CLL cells was confirmed by Western blotting in paired samples. In general these changes were more prominent in the IgVH unmutated CLL subtype as compared to IgVH mutated CLL cases. In particular, the proliferation E2F score was higher in LN biopsies of IgVH unmutated CLL than IgVH mutated CLL (P=0.04). The E2F score was also an excellent predictor of tumor progression measured as progression free survival (PFS) from diagnosis to treatment: patients with a high E2F score had a median PFS of 16.6 months compared to a PFS in excess of 10 years for patients with a low score (P=0.015). The acquired proliferation and activation signatures in CLL cells which are more prominent in LN resident CLL cells than in cells residing in the BM, suggests that the two microenvironment niches are not identical. Possible upstream cascades driving the signature of CLL cells in the tissue appear to be related to NF-kB and B-cell receptor activation. In conclusion: proliferation and cell activation signatures are acquired in the tissue and are more prominent in LN resident CLL cells than in the BM, suggesting that these two microenvironmental niches have different effects on tumor biology. The LN E2F proliferation signature was more prominent in IgVH unmutated CLL cells and correlated with clinical disease progression. Disclosures: No relevant conflicts of interest to declare.


2021 ◽  
Vol 39 (15_suppl) ◽  
pp. 10030-10030
Author(s):  
Jennifer Seelisch ◽  
Matthew Zatzman ◽  
Federico Comitani ◽  
Fabio Fuligni ◽  
Ledia Brunga ◽  
...  

10030 Background: Infant acute lymphoblastic leukemia (ALL) is the only subtype of childhood ALL whose outcome has not improved over the past two decades. The most important prognosticator is the presence of rearrangements in the Mixed Lineage Leukemia gene (MLL-r), however, many patients present with high-risk clinical features but without MLL-r. We recently identified two cases of infant ALL with high-risk clinical features resembling MLL-r, but were negative for MLL-r by conventional diagnostics. RNA sequencing revealed a partial tandem duplication in MLL (MLL-PTD). We thus aimed to determine if MLL-PTD, other MLL abnormalities, or other genetic or transcriptomic features were driving this subset of high-risk infant ALL without MLL-r. Methods: We obtained 19 banked patient samples from the Children’s Oncology Group (COG) infant ALL trial (AALL0631) from MLL wildtype patients as determined by FISH and cytogenetics. Utilizing deep RNA-sequencing, we manually inspected the MLL gene for MLL-PTD, while also performing automated fusion detection and gene expression profiling in search of defining features of these tumors. Results: 3 additional MLL-PTDs were identified, all in patients with infant T-cell ALL, whereas both index cases were in patients with infant B-cell ALL. Gene expression profiling analysis revealed that all five MLL-PTD infants clustered together. Eight infants (7 with B-cell ALL) were found to have Ph-like expression. Five of these 8 infants were also found to have an IKZF1/JAK2 expression profile; one of these five had a PAX5-JAK2 fusion detected. Two infants (including the one noted above) had novel PAX5 fusions, known drivers of B-cell leukemia. Additional detected fusions included TCF3-PBX1 and TCF4-ZNF384. Conclusions: MLL-PTDs were found in both B- and T-cell infant ALL. Though Ph-like ALL has been described in adolescents and young adults, we found a substantial frequency of Ph-like expression among MLL-WT infants. Further characterization of these infants is ongoing. If replicated in other infant cohorts, these two findings may help explain the poor prognosis of MLL-WT ALL when compared to children with standard risk ALL, and offer the possibility of targeted therapy for select infants.


2006 ◽  
Vol 130 (4) ◽  
pp. 483-520 ◽  
Author(s):  
Cherie H. Dunphy

Abstract Context.—Gene expression (GE) analyses using microarrays have become an important part of biomedical and clinical research in hematolymphoid malignancies. However, the methods are time-consuming and costly for routine clinical practice. Objectives.—To review the literature regarding GE data that may provide important information regarding pathogenesis and that may be extrapolated for use in diagnosing and prognosticating lymphomas and leukemias; to present GE findings in Hodgkin and non-Hodgkin lymphomas, acute leukemias, and chronic myeloid leukemia in detail; and to summarize the practical clinical applications in tables that are referenced throughout the text. Data Source.—PubMed was searched for pertinent literature from 1993 to 2005. Conclusions.—Gene expression profiling of lymphomas and leukemias aids in the diagnosis and prognostication of these diseases. The extrapolation of these findings to more timely, efficient, and cost-effective methods, such as flow cytometry and immunohistochemistry, results in better diagnostic tools to manage the diseases. Flow cytometric and immunohistochemical applications of the information gained from GE profiling assist in the management of chronic lymphocytic leukemia, other low-grade B-cell non-Hodgkin lymphomas and leukemias, diffuse large B-cell lymphoma, nodular lymphocyte–predominant Hodgkin lymphoma, and classic Hodgkin lymphoma. For practical clinical use, GE profiling of precursor B acute lymphoblastic leukemia, precursor T acute lymphoblastic leukemia, and acute myeloid leukemia has supported most of the information that has been obtained by cytogenetic and molecular studies (except for the identification of FLT3 mutations for molecular analysis), but extrapolation of the analyses leaves much to be gained based on the GE profiling data.


2015 ◽  
Vol 102 (2) ◽  
pp. 188-194 ◽  
Author(s):  
Kana Miyazaki ◽  
Motoko Yamaguchi ◽  
Hiroshi Imai ◽  
Kyoko Kobayashi ◽  
Satoshi Tamaru ◽  
...  

Epigenomics ◽  
2018 ◽  
Vol 10 (8) ◽  
pp. 1103-1119 ◽  
Author(s):  
Mariya A Smetanina ◽  
Alexander E Kel ◽  
Ksenia S Sevost'ianova ◽  
Igor V Maiborodin ◽  
Andrey I Shevela ◽  
...  

Author(s):  
David W. Scott

Diffuse large B-cell lymphoma (DLBCL) is the most common lymphoma worldwide and consists of a heterogeneous group of cancers classified together on the basis of shared morphology, immunophenotype, and aggressive clinical behavior. It is now recognized that this malignancy comprises at least two distinct molecular subtypes identified by gene expression profiling: the activated B-cell-like (ABC) and the germinal center B-cell-like (GCB) groups—the cell-of-origin (COO) classification. These two groups have different genetic mutation landscapes, pathobiology, and outcomes following treatment. Evidence is accumulating that novel agents have selective activity in one or the other COO group, making COO a predictive biomarker. Thus, there is now a pressing need for accurate and robust methods to assign COO, to support clinical trials, and ultimately guide treatment decisions for patients. The “gold standard” methods for COO are based on gene expression profiling (GEP) of RNA from fresh frozen tissue using microarray technology, which is an impractical solution when formalin-fixed paraffin-embedded tissue (FFPET) biopsies are the standard diagnostic material. This review outlines the history of the COO classification before examining the practical implementation of COO assays applicable to FFPET biopsies. The immunohistochemistry (IHC)-based algorithms and gene expression–based assays suitable for the highly degraded RNA from FFPET are discussed. Finally, the technical and practical challenges that still need to be addressed are outlined before robust gene expression–based assays are used in the routine management of patients with DLBCL.


Sign in / Sign up

Export Citation Format

Share Document