Proposal and validation of a method to classify genetic subtypes of diffuse large B cell lymphoma

Lucía Pedrosa, Ismael Fernández-Miranda, David Pérez-Callejo, Cristina Quero, Marta Rodríguez, Paloma Martín-Acosta, Sagrario Gómez, Julia González-Rincón, Adrián Santos, Carlos Tarin, Juan F García, Francisco R García-Arroyo, Antonio Rueda, Francisca I Camacho, Mónica García-Cosío, Ana Heredero, Marta Llanos, Manuela Mollejo, Miguel Piris-Villaespesa, José Gómez-Codina, Natalia Yanguas-Casás, Antonio Sánchez, Miguel A Piris, Mariano Provencio, Margarita Sánchez-Beato, Lucía Pedrosa, Ismael Fernández-Miranda, David Pérez-Callejo, Cristina Quero, Marta Rodríguez, Paloma Martín-Acosta, Sagrario Gómez, Julia González-Rincón, Adrián Santos, Carlos Tarin, Juan F García, Francisco R García-Arroyo, Antonio Rueda, Francisca I Camacho, Mónica García-Cosío, Ana Heredero, Marta Llanos, Manuela Mollejo, Miguel Piris-Villaespesa, José Gómez-Codina, Natalia Yanguas-Casás, Antonio Sánchez, Miguel A Piris, Mariano Provencio, Margarita Sánchez-Beato

Abstract

Diffuse large B-cell lymphoma (DLBCL) is a heterogeneous disease whose prognosis is associated with clinical features, cell-of-origin and genetic aberrations. Recent integrative, multi-omic analyses had led to identifying overlapping genetic DLBCL subtypes. We used targeted massive sequencing to analyze 84 diagnostic samples from a multicenter cohort of patients with DLBCL treated with rituximab-containing therapies and a median follow-up of 6 years. The most frequently mutated genes were IGLL5 (43%), KMT2D (33.3%), CREBBP (28.6%), PIM1 (26.2%), and CARD11 (22.6%). Mutations in CD79B were associated with a higher risk of relapse after treatment, whereas patients with mutations in CD79B, ETS1, and CD58 had a significantly shorter survival. Based on the new genetic DLBCL classifications, we tested and validated a simplified method to classify samples in five genetic subtypes analyzing the mutational status of 26 genes and BCL2 and BCL6 translocations. We propose a two-step genetic DLBCL classifier (2-S), integrating the most significant features from previous algorithms, to classify the samples as N12-S, EZB2-S, MCD2-S, BN22-S, and ST22-S groups. We determined its sensitivity and specificity, compared with the other established algorithms, and evaluated its clinical impact. The results showed that ST22-S is the group with the best clinical outcome and N12-S, the more aggressive one. EZB2-S identified a subgroup with a worse prognosis among GCB-DLBLC cases.

Conflict of interest statement

The authors declare no competing interests.

Figures

Figure 1
Figure 1
Mutational prevalence in genes and pathways. (A) Most frequently mutated genes and pathways in the whole cohort. (B) Mutational prevalence for refractory/relapsed (R/R) and sensitive (S) cases for genes with more than four samples mutated.
Figure 2
Figure 2
Statistical analysis of genes and pathways in the PdH cohort. Mutated genes and pathways with significant p-values in the univariate Cox proportional-hazards model analysis for (A) progression-free (PFS) and (B) overall survival (OS). Error bars represent the 95% confidence intervals for the hazard ratios. FDR p-values for Benjamini–Hochberg correction are shown.
Figure 3
Figure 3
Genetic subtypes and association with overall survival (OS) and progression-free survival (PFS). Genetic classification in the five defined genetic subtypes of DLBCL. Clustering was performed using alterations in genes (rows) from 84 DLBCL samples (columns). The OS and event-free survival status, and the ABC/GCB classification based on Lymph2cx and Hans are represented at the top. The phi coefficient and Fisher's exact test significance are represented on the right of the figure (*p 

Figure 4

Comparison of the classifiers. Subtypes…

Figure 4

Comparison of the classifiers. Subtypes assigned to the HMRN cohort by LymphGen, AIC…

Figure 4
Comparison of the classifiers. Subtypes assigned to the HMRN cohort by LymphGen, AIC cluster and the two-step classifier.

Figure 5

Progression-free survival (PFS) and overall…

Figure 5

Progression-free survival (PFS) and overall survival (OS) according to the genetic subtypes of…

Figure 5
Progression-free survival (PFS) and overall survival (OS) according to the genetic subtypes of the two-step, AIC cluster and LymphGen classifiers in the HMRN cohort. Kaplan–Meier analysis of genetic subtypes from (A) two-step, (B) AIC cluster and (C) LymphGen methods. Tables on the right of the figure show the hazard ratio (HR) values from the Cox proportional-hazards model of genetic subtypes for PFS and OS status. Error bars represent the 95% confidence intervals. Significance: *p < 0.05; **p < 0.01.

Figure 6

Kaplan–Meier analysis of progression-free survival…

Figure 6

Kaplan–Meier analysis of progression-free survival (PFS) and overall survival (OS) for EZB 2-S…

Figure 6
Kaplan–Meier analysis of progression-free survival (PFS) and overall survival (OS) for EZB2-S-GCB and other GCB cases in the HMRN cohort when applying the two-step classifier.
Figure 4
Figure 4
Comparison of the classifiers. Subtypes assigned to the HMRN cohort by LymphGen, AIC cluster and the two-step classifier.
Figure 5
Figure 5
Progression-free survival (PFS) and overall survival (OS) according to the genetic subtypes of the two-step, AIC cluster and LymphGen classifiers in the HMRN cohort. Kaplan–Meier analysis of genetic subtypes from (A) two-step, (B) AIC cluster and (C) LymphGen methods. Tables on the right of the figure show the hazard ratio (HR) values from the Cox proportional-hazards model of genetic subtypes for PFS and OS status. Error bars represent the 95% confidence intervals. Significance: *p < 0.05; **p < 0.01.
Figure 6
Figure 6
Kaplan–Meier analysis of progression-free survival (PFS) and overall survival (OS) for EZB2-S-GCB and other GCB cases in the HMRN cohort when applying the two-step classifier.

References

    1. Sant M, et al. Incidence of hematologic malignancies in Europe by morphologic subtype: results of the HAEMACARE project. Blood. 2010;116:3724–3734. doi: 10.1182/blood-2010-05-282632.
    1. Howlader N, Noone AM, Krapcho M, Miller D, Brest A, Yu M, Ruhl J, Tatalovich Z, Mariotto A, Lewis DR, Chen HS, Feuer EJ, C. K. Diffuse Large B-Cell Lymphoma—Cancer Stat Facts. (2019).
    1. Coiffier, B. et al. Long-term outcome of patients in the LNH-98.5 trial, the first randomized study comparing rituximab-CHOP to standard CHOP chemotherapy in DLBCL patients: a study by the Groupe d’Etudes des Lymphomes de l’Adulte. Blood 116, 2040 (2010).
    1. Pérez-Callejo D, González-Rincón J, Sánchez A, Provencio M, Sánchez-Beato M. Action and resistance of monoclonal CD20 antibodies therapy in B-cell Non-Hodgkin Lymphomas. Cancer Treat. Rev. 2015;41:680–689. doi: 10.1016/j.ctrv.2015.05.007.
    1. International Non-Hodgkin’s Lymphoma Prognostic Factors Project A predictive model for aggressive non-Hodgkin’s lymphoma. N. Engl. J. Med. 1993;329:987–994. doi: 10.1056/NEJM199309303291402.
    1. Sehn LH, et al. The revised International Prognostic Index (R-IPI) is a better predictor of outcome than the standard IPI for patients with diffuse large B-cell lymphoma treated with R-CHOP. Blood. 2007;109:1857–1861. doi: 10.1182/blood-2006-08-038257.
    1. Zhou Z, et al. An enhanced International Prognostic Index (NCCN-IPI) for patients with diffuse large B-cell lymphoma treated in the rituximab era. Blood. 2014;123:837–842. doi: 10.1182/blood-2013-09-524108.
    1. Swerdlow SH, et al. The 2016 revision of the World Health Organization classification of lymphoid neoplasms. Blood. 2016;127:2375–2390. doi: 10.1182/blood-2016-01-643569.
    1. Alizadeh AA, et al. Distinct types of diffuse large B-cell lymphoma identified by gene expression profiling. Nature. 2000;403:503–511. doi: 10.1038/35000501.
    1. Rosenwald A, et al. The use of molecular profiling to predict survival after chemotherapy for diffuse large-B-cell lymphoma. N. Engl. J. Med. 2002;346:1937–1947. doi: 10.1056/NEJMoa012914.
    1. Wright G, et al. A gene expression-based method to diagnose clinically distinct subgroups of diffuse large B cell lymphoma. Proc. Natl. Acad. Sci. USA. 2003;100:9991–9996. doi: 10.1073/pnas.1732008100.
    1. Schmitz R, et al. Genetics and pathogenesis of diffuse large B-cell lymphoma. N. Engl. J. Med. 2018;378:1396–1407. doi: 10.1056/NEJMoa1801445.
    1. Wright GW, et al. A probabilistic classification tool for genetic subtypes of diffuse large B cell lymphoma with therapeutic implications. Cancer Cell. 2020;37:551–568.e14. doi: 10.1016/j.ccell.2020.03.015.
    1. Chapuy B, et al. Molecular subtypes of diffuse large B cell lymphoma are associated with distinct pathogenic mechanisms and outcomes. Nat. Med. 2018;24:679–690. doi: 10.1038/s41591-018-0016-8.
    1. Lacy SE, et al. Targeted sequencing in DLBCL, molecular subtypes, and outcomes: a Haematological Malignancy Research Network report. Blood. 2020;135:1759–1771. doi: 10.1182/blood.2019003535.
    1. Aggarwal M, et al. Functional signatures identified in B-cell non-Hodgkin lymphoma profiles. Leukemia Lymphoma. 2009;50:1699–1708. doi: 10.1080/10428190903189035.
    1. Pasqualucci L, et al. Analysis of the coding genome of diffuse large B-cell lymphoma. Nat. Genet. 2011;43:830. doi: 10.1038/ng.892.
    1. Morin RD, et al. Frequent mutation of histone-modifying genes in non-Hodgkin lymphoma. Nature. 2011;476:298–303. doi: 10.1038/nature10351.
    1. de Miranda NFCC, et al. Exome sequencing reveals novel mutation targets in diffuse large B-cell lymphomas derived from Chinese patients. Blood. 2014;124:2544–2553. doi: 10.1182/blood-2013-12-546309.
    1. Lohr JG, et al. Discovery and prioritization of somatic mutations in diffuse large B-cell lymphoma (DLBCL) by whole-exome sequencing. Proc. Natl. Acad. Sci. USA. 2012;109:3879. doi: 10.1073/pnas.1121343109.
    1. Pasqualucci L, et al. Inactivating mutations of acetyltransferase genes in B-cell lymphoma. Nature. 2011;471:189–195. doi: 10.1038/nature09730.
    1. Morin RD, et al. Genetic landscapes of relapsed and refractory diffuse large B-cell lymphomas. Clin. Cancer Res. 2016;22:2290–2300. doi: 10.1158/1078-0432.CCR-15-2123.
    1. Braggio E, et al. Genome-wide analysis uncovers novel recurrent alterations in primary central nervous system lymphomas. Clin. Cancer Res. 2015;21:3986–3994. doi: 10.1158/1078-0432.CCR-14-2116.
    1. Cao Y, et al. Mutations or copy number losses of CD58 and TP53 genes in diffuse large B cell lymphoma are independent unfavorable prognostic factors. Oncotarget. 2016;7:83294–83307. doi: 10.18632/oncotarget.13065.
    1. Mottok A, et al. Integrative genomic analysis identifies key pathogenic mechanisms in primary mediastinal large B-cell lymphoma. Blood. 2019;134:802–813. doi: 10.1182/blood.2019001126.
    1. Gonzalez-Farre B, et al. Burkitt-like lymphoma with 11q aberration: a germinal center-derived lymphoma genetically unrelated to Burkitt lymphoma. Haematologica. 2019;104:1822–1829. doi: 10.3324/haematol.2018.207928.
    1. Bonetti P, et al. Deregulation of ETS1 and FLI1 contributes to the pathogenesis of diffuse large B-cell lymphoma. Blood. 2013;122:2233–2241. doi: 10.1182/blood-2013-01-475772.
    1. Reddy A, et al. Genetic and functional drivers of diffuse large B cell lymphoma. Cell. 2017;171:481–494.e15. doi: 10.1016/j.cell.2017.09.027.
    1. Bolen CR, et al. Prognostic impact of somatic mutations in diffuse large B-cell lymphoma and relationship to cell-of-origin: data from the phase III GOYA study. Haematologica. 2019 doi: 10.3324/haematol.2019.227892.
    1. López-Guerra M, et al. The γ-secretase inhibitor PF-03084014 combined with fludarabine antagonizes migration, invasion and angiogenesis in NOTCH1-mutated CLL cells. Leukemia. 2015;29:96–106. doi: 10.1038/leu.2014.143.
    1. Messersmith WA, et al. A Phase I, dose-finding study in patients with advanced solid malignancies of the oral γ-secretase inhibitor PF-03084014. Clin. Cancer Res. 2015;21:60–67. doi: 10.1158/1078-0432.CCR-14-0607.
    1. Papayannidis C, et al. A Phase 1 study of the novel gamma-secretase inhibitor PF-03084014 in patients with T-cell acute lymphoblastic leukemia and T-cell lymphoblastic lymphoma. Blood Cancer J. 2015;5:e350. doi: 10.1038/bcj.2015.80.
    1. González-Rincón J, et al. Unraveling transformation of follicular lymphoma to diffuse large B-cell lymphoma. PLoS ONE. 2019;14:e0212813. doi: 10.1371/journal.pone.0212813.
    1. Zhang J, et al. Genetic heterogeneity of diffuse large B-cell lymphoma. Proc. Natl. Acad. Sci. 2013;110:1398–1403. doi: 10.1073/pnas.1205299110.
    1. Karube K, et al. Recurrent mutations of NOTCH genes in follicular lymphoma identify a distinctive subset of tumours. J. Pathol. 2014;234:423–430. doi: 10.1002/path.4428.
    1. Okosun J, et al. Integrated genomic analysis identifies recurrent mutations and evolution patterns driving the initiation and progression of follicular lymphoma. Nat. Genet. 2014;46:176–181. doi: 10.1038/ng.2856.
    1. Pasqualucci L, et al. Genetics of follicular lymphoma transformation. Cell Rep. 2014;6:130–140. doi: 10.1016/j.celrep.2013.12.027.
    1. Hans CP, et al. Confirmation of the molecular classification of diffuse large B-cell lymphoma by immunohistochemistry using a tissue microarray. Blood. 2004;103:275–282. doi: 10.1182/blood-2003-05-1545.
    1. Kulkarni, M. M. Digital multiplexed gene expression analysis using the NanoString nCounter system. Current protocols in molecular biology Chapter 25, Unit25B.10 (2011).
    1. Watatani Y, et al. Molecular heterogeneity in peripheral T-cell lymphoma, not otherwise specified revealed by comprehensive genetic profiling. Leukemia. 2019;33:2867–2883. doi: 10.1038/s41375-019-0473-1.
    1. Scott DW, 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;33:2848–2856. doi: 10.1200/JCO.2014.60.2383.
    1. Scott DW, et al. Determining cell-of-origin subtypes of diffuse large B-cell lymphoma using gene expression in formalin-fixed paraffin-embedded tissue. Blood. 2014;123:1214–1217. doi: 10.1182/blood-2013-11-536433.

Source: PubMed

3
Subscribe