A three-gene signature based on MYC, BCL-2 and NFKBIA improves risk stratification in diffuse large B-cell lymphoma

Enrico Derenzini, Saveria Mazzara, Federica Melle, Giovanna Motta, Marco Fabbri, Riccardo Bruna, Claudio Agostinelli, Alessandra Cesano, Chiara Antonia Corsini, Ning Chen, Simona Righi, Elena Sabattini, Annalisa Chiappella, Angelica Calleri, Stefano Fiori, Valentina Tabanelli, Antonello Cabras, Giancarlo Pruneri, Umberto Vitolo, Alessandro Massimo Gianni, Alessandro Rambaldi, Paolo Corradini, Pier Luigi Zinzani, Corrado Tarella, Stefano Pileri, Enrico Derenzini, Saveria Mazzara, Federica Melle, Giovanna Motta, Marco Fabbri, Riccardo Bruna, Claudio Agostinelli, Alessandra Cesano, Chiara Antonia Corsini, Ning Chen, Simona Righi, Elena Sabattini, Annalisa Chiappella, Angelica Calleri, Stefano Fiori, Valentina Tabanelli, Antonello Cabras, Giancarlo Pruneri, Umberto Vitolo, Alessandro Massimo Gianni, Alessandro Rambaldi, Paolo Corradini, Pier Luigi Zinzani, Corrado Tarella, Stefano Pileri

Abstract

Recent randomized trials focused on gene expression-based determination of the cell of origin in diffuse large B-cell lymphoma could not show significant improvements by adding novel agents to standard chemoimmunotherapy. The aim of this study was the identification of a gene signature able to refine current prognostication algorithms and applicable to clinical practice. Here we used a targeted gene expression profiling panel combining the Lymph2Cx signature for cell of origin classification with additional targets including MYC, BCL-2 and NFKBIA, in 186 patients from 2 randomized trials (discovery cohort) (NCT00355199 and NCT00499018). Data were validated in 3 independent series (2 large public datasets and a real-life cohort). By integrating the cell of origin, MYC/BCL-2 double expressor status and NFKBIA expression, we defined a 3-gene signature combining MYC, BCL-2 and NFKBIA (MBN-signature), which outperformed the MYC/BCL-2 double expressor status in multivariate analysis, and allowed further risk stratification within the germinal center B-cell/unclassified subset. The high-risk (MBN Sig-high) subgroup identified the vast majority of double hit cases and a significant fraction of Activated B-Cell-derived diffuse large B-cell lymphomas. These results were validated in 3 independent series including a cohort from the REMoDL-B trial, where, in an exploratory ad hoc analysis, the addition of bortezomib in the MBN Sig-high subgroup provided a progression free survival advantage compared with standard chemoimmunotherapy. These data indicate that a simple 3-gene signature based on MYC, BCL-2 and NFKBIA could refine the prognostic stratification in diffuse large B-cell lymphoma, and might be the basis for future precision-therapy approaches.

Figures

Figure 1.
Figure 1.
Study algorithm. On the left, the discovery cohort is represented; 224 diffuse large B-cell lymphoma (DLBCL) patients enrolled in the DLCL04 (n= 130) and R-HDS0305 (n= 94) trials with available formalin-fixed, paraffin embedded (FFPE) tissue were initially considered in this analysis. Targeted gene expression profiling (TGEP) success rate was 92.4% (n=207), with 17 cases not yelding enough highquality mRNA to undergo successful GEP assessment. Only cases originally diagnosed as DLBCL non-otherwise specified (NOS) were considered. Therefore 21 cases classified in different DLBCL categories were excluded; 99 NOS-DLBCL FFPE patient samples from the DLCL04 trial and 87 samples from the R-HDS0305 trial were finally included in this study. On the right, the three validation cohorts: a cohort of 928 patients from Sha and coworkers (469 treated with R-CHOP; 459 with RBCHOP), a public gene expression dataset (Affymetrix Human Genome U133 Plus 2.0 Array), GSE10846, (https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE10846), including 233 patients treated with R-CHOP regimen (Lenz et al. 2008); an additional validation cohort including 102 consecutive DLBCL NOS cases with available FFPE tissue, treated with R-CHOP/R-CHOP-like regimens. RB-CHOP: R-CHOP plus bortezomib.
Figure 2.
Figure 2.
Integrating cell of origin with MYC/BCL-2 DEXP_mRNA status for prognostication in diffuse large Bcell lymphoma. (A) Recursive partitioning analysis integrating cell of origin (COO) classification and DEXP_mRNA status, allowing segregation of patients in three main prognostic subgroups (a low risk non-DEXP-mRNA GCB/U subset, and two high risk groups: MYC/BCL-2-DEXP-mRNA GCB/U and ABC). (B) Box plot graphs indicating the expression levels of the additional targets included in the panel (MYC, BCL-2, NFKBIA, STAT3, PIK3CA, PTEN) in the three main patients subgroups identified by the recursive partitioning analysis (non-DEXPmRNA GCB/U, MYC/BCL-2 DEXP_mRNA GCB/U, ABCderived diffuse large B-cell lymphoma [DLBCL]. P-value was calculated with Student ttest by comparing non DEXP_mRNA GCB/U group, selected as a reference, versus other groups. ABC: activated B-cells; GCB: germinal center B cells; GCB/U: GCB unclassified; DEXP_mRNA: double expressor GCB/U DLBCL. COO Nano: COO as determined by T-GEP with NanoString profiling
Figure 3.
Figure 3.
Survival curves according to MBN signature and multivariate analyses for overall survival. (A) Overall survival (OS) of the discovery cohort (RHDS0305+ DLCL04; n=186 patients) according to the MBN signature (MBN-Sig) showing significant differences in outcome between MBN-Sig low versus MBN-Sig high patient subsets. P-values were calculated with the log rank test. Frequencies of MBN-Sig high versus low cases in activated B-cells (ABC) and germinal center B cells/ unclassified (GCB/U) subsets in the discovery cohort are represented in the pie chart. (B) Forest plot depicting multivariate analyses for OS (discovery cohort). Only factors significantly asociated with OS in univariate analyses were considered. According to this analysis only the cell of origin (COO) as determined by NanoString-based targeted gene expression profiling (T-GEP) (COO_Nano), the MBN-Sig and the age adjusted international prognostic index (aaIPI) score retained statistical significance for OS, whereas MYC/BCL-2 DEXP_mRNA status, STAT3 and NFKBIA levels determined by T-GEP were not significantly associated with OS. HR: hazard ratio. (C) OS of the 469 patients treated with R-CHOP in the Sha’s cohort according to the MBN signature showing significant differences in outcome between MBN-Sig low versus MBN-Sig high patient subsets. P-values were calculated with the log rank test. Frequencies of MBN-Sig high versus low cases in ABC and GCB/U subsets in the Sha’s cohort are represented in the pie chart. (C) Forest plot depicting multivariate analyses for OS (Sha’s dataset), confirming the significant independent association with OS of the MBN-Sig in this large validation cohort.
Figure 4.
Figure 4.
Real-life applicability of the MBN signature. (A) Heatmap representing the three informative genes of the MBN signature (MBN-Sig) shown as rows and diffuse large B-cell lymphoma (DLBCL) tissue samples shown as columns in the real-life cohort of 102 patients, with the actual MBN-Sig and the predicted MBN-Sig class based on the application of a random forest (RF) model built on the discovery cohort on the top of the heatmap. (B) Violin plot showing the fractions of false predictions (false positive [FP], and false negative [FN]) as well as true predictions (true positive [TP], and true negative [TN]) in the real-life cohort by applying a three-gene RF model. (C) ROC curve of the real-life cohort using RF classifier. (D) Overall survival (OS) curve of the real-life cohort (n=102) based on the predicted MBN-Sig class. P-value was calculated with the log rank test.
Figure 5.
Figure 5.
The MBN signature identifies prognostically distinct subgroups including activated B cells and a fraction of germinal center B cells/ unclassified diffuse large B-cell lymphoma (DLBCL) enriched in double hit DLBCL cases, providing opportunities for precision therapies. (A) Overall survival (OS) of the germinal center B cells/ unclassified (GCB/U) subset in the discovery cohort (n=146 patients) according to the integration of the targeted gene expression profiling (T-GEP) panel (Lymph2Cx) with the MBN signature (MBN-Sig), distinguishing two risk categories according to the MBN-Sig. (B) Heatmap representing the three informative genes of the MBN-Sig shown as rows and diffuse large B-cell lymphoma (DLBCL) tissue samples shown as columns, in the discovery cohort (n=186 patients). (C) OS of the GCB/U subset in the Sha’s validation cohort (n=340 patients) according to the MBN-Sig, showing superimposable results compared to Figure 4C. (D) Heatmap representing the three informative genes of the MBN-Sig shown as rows and DLBCL tissue samples shown as columns, in the Sha’s cohort (n=469 patients treated with R-CHOP). (E) Progression-free survival (PFS) of patients treated with R-CHOP versus RB-CHOP in the MBN-Sig high subgroup (Full Sha’s cohort, n=928 patients; MBNSig high n=464 patients). (F) OS of patients treated with R-CHOP versus RB-CHOP in the MBN-Sig high subgroup (Full Sha’s cohort, n=928 patients; MBN-Sig high n=464 patients). In all panels the P-value was calculated with the log rank test. NA: not available; DH: double hit; DE: double expressor (based on DEXP_mRNA status); aaIPI: age adjusted international prognostic index; COO: cell of origin; IPI: international prognostic index); MHG: molecular high grade. RB-CHOP: R-CHOP plus Bortezomib.

References

    1. Alizadeh AA, Eisen MB, Davis RE, et al. . Distinct types of diffuse large B-cell lymphoma identified by gene expression profiling. Nature. 2000;403(6769):503-511.
    1. Shipp MA, Ross KN, Tamayo P, et al. . Diffuse large B-cell lymphoma outcome prediction by gene-expression profiling and supervised machine learning. Nat Med. 2002;8(1):68-74.
    1. Davis RE, Brown KD, Siebenlist U, Staudt LM. Constitutive nuclear factor kappaB activity is required for survival of activated B cell-like diffuse large B cell lymphoma cells. J Exp Med. 2001;194(12):1861-1874.
    1. Hans CP, Weisenburger DD, Greiner TC, et al. . Confirmation of the molecular classification of diffuse large B-cell lymphoma by immunohistochemistry using a tissue microarray. Blood. 2004;103(1):275-282.
    1. Meyer PN, Fu K, Greiner TC, et al. . Immunohistochemical methods for predicting cell of origin and survival in patients with diffuse large B-cell lymphoma treated with rituximab. J Clin Oncol. 2011; 29(2):200-207.
    1. Lawrie CH, Ballabio E, Soilleux E, et al. . Interand intra-observational variability in immunohistochemistry: a multicentre analysis of diffuse large B-cell lymphoma staining. Histopathology. 2012;61(1):18-25.
    1. Castillo JJ, Beltran BE, Song MK, et al. . The Hans algorithm is not prognostic in patients with diffuse large B-cell lymphoma treated with R-CHOP. Leuk Res. 2012;36(4):413-417.
    1. Coutinho R, Clear AJ, Owen A, et al. . Poor concordance among nine immunohistochemistry classifiers of cell-of-origin for diffuse large B-cell lymphoma: implications for therapeutic strategies. Clin Cancer Res. 2013;19(24):6686-6695.
    1. Hu S, Xu-Monette ZY, Tzankov A, et al. . MYC/BCL2 protein coexpression contributes to the inferior survival of activated Bcell subtype of diffuse large B-cell lymphoma and demonstrates high-risk gene expression signatures: a report from The International DLBCL Rituximab-CHOP Consortium Program. Blood. 2013; 121(20): 4021-4031.
    1. Reinke S, Richter J, Fend F, et al. . Round-robin test for the cell-of-origin classification of diffuse large B-cell lymphoma-a feasibility study using full slide staining. Virchows Arch. 2018;473(3):341-349.
    1. Scott DW, Wright GW, Williams PM, et al. . Determining cell-of-origin subtypes of diffuse large B-cell lymphoma using gene expression in formalin-fixed paraffin-embedded tissue. Blood. 2014;123(8):1214-1217.
    1. Scott DW, Mottok A, Ennishi D, et al. . Prognostic significance of diffuse large B-cell lymphoma cell of origin determined by digital gene expression in formalin-Fixed paraffin- embedded tissue biopsies. J Clin Oncol. 2015;33(26):2848-2856.
    1. Painter D, Barrans S, Lacy S, et al. . Cell-of-origin in diffuse large B-cell lymphomafindings from the UK’s population-based Haematological Malignancy Research Network. Br J Haematol. 2019;185(4):752–806.
    1. Veldman-Jones MH, Lai Z, Wappett M, et al. . Reproducible, quantitative, and flexible molecular subtyping of clinical DLBCL samples using the NanoString nCounter System. Clin Cancer Res. 2015; 21(10):2367-2378.
    1. Rimsza LM, Wright G, Schwartz M, et al. . Accurate classification of diffuse large B-cell lymphoma into germinal center and activated B-cell subtypes using a nuclease protection assay on formalin-fixed, paraffinembedded tissues. Clin Cancer Res. 2011;17(11):3727-3732.
    1. Younes A, Sehn LH, Johnson P, et al. . Randomized phase III trial of ibrutinib and rituximab plus cyclophosphamide, doxorubicin, vincristine, and prednisone in non-germinal center B-cell diffuse large B-cell lymphoma. J Clin Oncol. 2019;37(15):1285-1295.
    1. Vitolo U, Witzig T, Gascoyne R, et al. . ROBUST: first report of phase III randomized study of lenalidomide/R CHOP (R2 CHOP) vs placebo/R CHOP in previously untreated ABC type diffuse large B cell lymphoma. Hematol Oncol. 2019; 37(S2):36-37.
    1. Davies A, Cummin TE, Barrans S, et al. . Gene-expression profiling of bortezomib added to standard chemoimmunotherapy for diffuse large B-cell lymphoma (REMoDLB): an open-label, randomised, phase 3 trial. Lancet Oncol. 2019;20(5):649-662.
    1. Green TM, Young KH, Visco C, et al. . Immunohistochemical double-hit score is a strong predictor of outcome in patients with diffuse large B-cell lymphoma treated with rituximab plus cyclophosphamide, doxorubicin, vincristine, and prednisone. J Clin Oncol. 2012;30(28):3460-3467.
    1. Johnson NA, Slack GW, Savage KJ, et al. . Concurrent expression of MYC and BCL2 in diffuse large B-cell lymphoma treated with rituximab plus cyclophosphamide, doxorubicin, vincristine, and prednisone. J Clin Oncol. 2012;30(28):3452-3459.
    1. taiger AM, Ziepert M, Horn H, et al. . Clinical impact of the Cell-of-Origin Classification and the MYC/ BCL2 dual expresser status in diffuse large B-cell lymphoma treated within prospective clinical trials of the German High-Grade Non-Hodgkin's Lymphoma Study Group. J Clin Oncol. 2017;35(22):2515-2526.
    1. Swerdlow SH, Campo E, Harris NL, et al. . WHO Classification of Tumour of Haematopoietic and Lymphoid Tissues, Revised 4th edition. 2017. IARC Press, Lyon.
    1. Reddy A, Zhang J, Davis NS, et al. . Genetic and functional drivers of diffuse large B cell lymphoma. Cell. 2017;171(2):481-494.
    1. Schmitz R, Wright GW, Huang DW, et al. . Genetics and pathogenesis of diffuse large Bcell lymphoma. N Engl J Med. 2018; 378(15):1396-1407.
    1. Chapuy B, Stewart C, Dunford AJ, et al. . Molecular subtypes of diffuse large B cell lymphoma are associated with distinct pathogenic mechanisms and outcomes. Nat Med. 2018;24(8):679-690.
    1. Ennishi D, Jiang A, Boyle M, et al. . Double-hit gene expression signature defines a distinct subgroup of germinal center B-cell-like diffuse large B-cell lymphoma. J Clin Oncol. 2019;37(3):190-201.
    1. Sha C, Barrans S, Cucco F, et al. . Molecular high-grade B-cell lymphoma: defining a poor-risk group that requires different approaches to therapy. J Clin Oncol. 2019;37(3):202-212.
    1. Pasqualucci L, Dalla-Favera R.Genetics of diffuse large B-cell lymphoma. Blood. 2018; 131(21):2307-2319.
    1. Roschewski M, Staudt LM, Wilson WH. Diffuse large B-cell lymphoma-treatment approaches in the molecular era. Nat Rev Clin Oncol. 2014;11(1):12-23.
    1. Paul J, Soujon M, Wengner AM, et al. . Simultaneous inhibition of PI3Kδ and PI3Kαinduces ABC-DLBCL regression by blocking BCR-dependent and -independent activation of NF-κB and AKT. Cancer Cell. 2017;31(1):64-78.
    1. Pfeifer M, Grau M, Lenze D, et al. . PTEN loss defines a PI3K/AKT pathway-dependent germinal center subtype of diffuse large B-cell lymphoma. Proc Natl Acad Sci U S A. 2013;110(30):12420-12425.
    1. Compagno M, Lim WK, Grunn A, et al. . Mutations of multiple genes cause deregulation of NF-kappaB in diffuse large B-cell lymphoma. Nature. 2009;459(7247):717-721.
    1. Pan YR, Chen CC, Chan YT, et al. . STAT3- coordinated migration facilitates the dissemination of diffuse large B-cell lymphomas. Nat Commun. 2018;9(1):3696.
    1. Cortelazzo S, Tarella C, Gianni AM, et al. . Randomized trial comparing R-CHOP versus high-dose sequential chemotherapy in highrisk patients with diffuse large B-cell lymphomas. J Clin Oncol. 2016; 34(33):4015-4022.
    1. Chiappella A, Martelli M, Angelucci E, et al. . Rituximab-dose-dense chemotherapy with or without high-dose chemotherapy plus autologous stem-cell transplantation in highrisk diffuse large B-cell lymphoma (DLCL04): final results of a multicentre, open-label, randomised, controlled, phase 3 study. Lancet Oncol. 2017;18(8):1076-1088.
    1. Lenz G, Wright G, Dave SS, et al. . Stromal gene signatures in large-B-cell lymphomas. N Engl J Med 2008;359(22):2313-2323.
    1. Kaplan EL, Meier P.Nonparametric estimations from incomplete observations. J Am Stat Assoc. 1958;53(282):457-481.
    1. Curran WJ, Scott CB, Horton J, et al. . Recursive partitioning analysis of prognostic factors in three Radiation Therapy Oncology Group malignant glioma trials. J Natl Cancer Inst. 1993;85(9):704-710.
    1. R Core Team. R: a language and environment for statistical computing. R Foundation for Statistical Computing, Vienna, Austria. 2014. URL .
    1. Oeckinghaus A, Ghosh S.The NF-κB family of transcription factors and its regulation. Cold Spring Harbor Perspectives in Biology. 2009;1(4):a000034.
    1. Jost PJ, Ruland J.Aberrant NF-κB signaling in lymphoma: mechanisms, consequences, and therapeutic implications. Blood. 2007; 109(7):2700-2707.
    1. Packham G. The role of NF-kappaB in lymphoid malignancies. Br J Haematol. 2008;143(1):3-15.
    1. McConkey DJ, Zhu K.Mechanisms of proteasome inhibitor action and resistance in cancer. Drug Resist Updat. 2008;11(4-5):164-179.
    1. Mujtaba T, Dou QP. Advances in the understanding of mechanisms and therapeutic use of bortezomib. Discov Med. 2011;12(67): 471-480.
    1. Bu R, Hussain AR, Al-Obaisi KA, Ahmed M, Uddin S, Al-Kuraya KS. Bortezomib inhibits proteasomal degradation of IκBα and induces mitochondrial dependent apoptosis in activated B-cell diffuse large B-cell lymphoma. Leuk Lymphoma. 2014; 55(2):415-424.
    1. Delmore JE, Issa GC, Lemieux ME, et al. . BET bromodomain inhibition as a therapeutic strategy to target c-Myc. Cell. 2011;146(6): 904-917.
    1. Derenzini E, Mondello P, Erazo T, et al. . BET inhibition-induced GSK3β feedback enhances lymphoma vulnerability to PI3K inhibitors. Cell Rep. 2018;24(8):2155-2166.
    1. Ceribelli M, Kelly PN, Shaffer AL, et al. . Blockade of oncogenic IκB kinase activity in diffuse large B-cell lymphoma by bromodomain and extraterminal domain protein inhibitors. Proc Natl Acad Sci U S A. 2014;111(31):11365-11370.

Source: PubMed

3
購読する