SnapShot: Diffuse Large B Cell Lymphoma Laura Pasqualucci1,2,5 and Riccardo Dalla-Favera1,2,3,4,5 Institute for Cancer Genetics, 2Department of Pathology and Cell Biology, 3Department of Genetics, 4 Department of Microbiology and Immunology, 5Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY 10032, USA

Shared GCB- & ABC-DLBCL

Altered histone/ chromatin modifications CREBBP/EP300 MLL2/MLL3

32 32-38

Immune escape B2M CD58

29 21

Deregulated BCL6 activity BCL6 MEF2B Other FOXO1 TP53

CELL OF ORIGIN CN gains

Amplifications

Translocations

% of DLBCL

Deletions

Loss of function Gain of function

Mutations

1

Naive B cell

GCB-DLBCL ABC-DLBCL

Constitutive NF-κB/BCR activity TNFAIP3 MYD88 CD79A/B CARD11 Terminal differentiation block PRDM1/BLIMP1 Apoptosis BCL2 Cell cycle checkpoint CDKN2A/B Other SPIB

PMBCL

JAK/STAT activation JAK2/JMJD2C SOCS1 STAT6 Constitutive NF-κB activity TNFAIP3 Immune escape PDL1/PDL2 CIITA Deregulated BCL6 activity BCL6

Centroblast

Memory B cell

T cell

Centrocyte Thymic B cell

Plasmablast Plasma cell

GCB-DLBCL ABC-DLBCL

20-40 11 8 20

Proliferation/ Apoptosis BCL2 34-45 10 MYC miR-17~92 6-12.5

Other EZH2 BCL6 BSE1 sites 2p16.1

Thymus

FDC

IMMUNE ESCAPE

% of subtype

Signaling TNFRSF14 GNA13 SGK1 PTEN

Germinal Center LIGHT ZONE DARK ZONE T cell

Ag

30 30-37 21 10

DEREGULATED BCL6 ACTIVITY

20-40%

CTL

HLA-I loss relieves inhibition on NK

Non-self antigen activates CTL

21%

4%

32%

FBXO11

CBP/EP300

BCL6 protein

29%

DLBCL

NK

CTL

ABERRANT HISTONE/ CHROMATIN MODIFICATIONS

DNA damage response (p53, ATR)

Cell cycle arrest (p21)

B cell Programmed Plasma cell activation cell death differentiation (CD80) (BCL2) (BLIMP1)

CONSTITUTIVE BCR AND NF-κB SIGNALING BTK-i

CD79A/B

K27

SYK LYN

H3 24-30% GCB-DLBCL

22%

MAPK/ERK PI3K/AKT

32% CREBBP

EZH2

MLL2 EP300 MLL3 11%

SUZ12

30-45 45 36

BTK

MALT1-i

10%

CARD11 BCL10 30-37% MALT1 MyD88

10%

Transcriptional repressors K27

36

30% A20

NF-κ B

NF-κB-i

JAK/STAT Interferon p38/MAPK

HDAC-i Lenalidomide

IRF4 25% BCL6 translocation

H3

25-32% BCL6-i

33*

TLR/IL-1R

PKCβ

24-30

EZH2-i

CD40L CD40

BCR

21%

K4

ABC-DLBCL

Ag

Transcriptional activators

13

30-45 38

BCL6 translocation; more frequent in ABC-DLBCL

BCL6 gene

25-32

26

IRF4

MEF2B

Normal B cell HLA-I TCR

NK

NF-κB 11%

Non-self Ag

CD2 CD58

13 11 13 6-11 22 15 16

PMBCL

K4 methylation

K27 methylation

K27 acetylation

*Based on analysis of 12 cases only

132 Cancer Cell 25, January 13, 2014 ©2014 Elsevier Inc.

DOI 10.1016/j.ccr.2013.12.012

BCL6

Loss of function genetic lesions

PRDM1 Gain of function genetic lesions

See online version for legend and references.

SnapShot: Diffuse Large B Cell Lymphoma Laura Pasqualucci1,2,5 and Riccardo Dalla-Favera1,2,3,4,5 Institute for Cancer Genetics, 2Department of Pathology and Cell Biology, 3Department of Genetics, 4 Department of Microbiology and Immunology, 5Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY 10032, USA

1

Diffuse large B cell lymphoma (DLBCL) is the most prevalent non-Hodgkin lymphoma (NHL) in adulthood, comprising 30%–40% of all new diagnoses. This aggressive disease can arise de novo or, less frequently, from the clinical evolution of various indolent B cell malignancies. While durable remissions can be achieved in a substantial proportion of cases by combined chemoimmunotherapy, over 30% of patients will not respond to currently available regimens or will relapse with resistant disease. One explanation for such incomplete therapeutic success is the considerable heterogeneity of this tumor. DLBCL comprises multiple molecular subgroups, which can be recognized by their gene expression profiles and reflect either the stage in B cell development from which the disease originates or the activity of different biological programs, including metabolic dysregulation. DLBCL subgroups differ in the oncogenic pathways that drive them and in their response to treatment. Thus, the recognition of dysregulated genes/programs that are critical to the survival of the lymphoma cells is central to the development of rationally targeted therapeutic approaches for DLBCL. Cell of Origin Analogous to most B-NHL, DLBCL derives from a mature B cell that has experienced the germinal center (GC) reaction. DLBCLs have been divided into three subgroups based on putative cells of origin. GC B cell-like (GCB)-DLBCL exhibits a transcriptional profile that resembles that of GC B cells, including having elevated expression of BCL6 and the presence of hypermutated immunoglobulin genes with ongoing somatic hypermutation (SHM). Activated B cell-like (ABC)-DLBCL shows several features of BCR-activated B cells entering plasmablastic differentiation; these tumors downregulate the GC-specific program, concomitant with activation of NF-kB and BCR signaling pathways, and upregulate genes required for plasma cell differentiation (e.g., IRF4). Consistent with their late GC origin, ABC-DLBCLs do not show evidence of ongoing SHM. Primary mediastinal B cell lymphoma (PMBCL) is postulated to arise from a post-GC thymic B cell in the mediastinum and shares histological, molecular, and clinical features with nodular sclerosis Hodgkin lymphoma, including a robust immune/inflammatory cell infiltrate, a distinctive cytokine profile, and constitutive NF-kB activation. An additional 15%–30% of DLBCL cases remain unclassified. Stratification of DLBCL patients according to the cell-of-origin classification was shown to have prognostic significance, with GCB-DLBCL displaying a better overall survival compared to ABC-DLBCL. Genetic Alterations Compared to other B cell malignancies, DLBCL shows a significantly higher degree of genomic complexity, typically harboring between 50 and >100 lesions per case, with high variability across patients. These figures, which include point mutations, copy number aberrations, and chromosomal translocations, likely represent an underestimate because sequencing studies performed so far have not interrogated noncoding portions of the genome. Recent genomic analysis of DLBCL revealed a number of previously unrecognized genes/pathways that are dysregulated by genetic lesions and presumably play central roles in tumor initiation and maintenance. Some of these lesions can be observed in both GCB and ABC subtypes of DLBCL, while others are preferentially associated with individual DLBCL subtypes, suggesting their potential for diagnostic and therapeutic stratification. The following paragraphs focus on the most frequent and well-characterized alterations (see the table for a comprehensive list). Alterations Shared Across Subtypes Inactivating mutations and deletions of the histone acetyltransferases CREBBP/EP300 and the histone methyltransferase MLL2 emerged as the most common genetic lesions in DLBCL, where they may favor tumor development by reprogramming the cancer epigenome. The prevalence of these lesions has therapeutic implications given the potential of histone deacetylase inhibitors to restore physiologic acetylation levels. A multitude of genetic lesions result in deregulation of BCL6 activity, either directly (chromosomal translocations or mutations abrogating its promoter regulatory sequences) or indirectly, by enhancing the activity of its positive regulator MEF2B, preventing acetylation-mediated inactivation of its function (CREBBP/EP300 mutations/ deletions), or abrogating mechanisms controlling protein degradation (FBXO11 mutations/deletions). Notably, pharmacologic inhibition of BCL6 is lethal to most DLBCL. DLBCL cells acquire the ability to escape immune surveillance, including CTL-mediated cytotoxicity (through genetic loss of B2M or HLA-I genes) and natural killer cellmediated death (through loss of the gene encoding the CD58 ligand). In PMBCL, reduced tumor cell immunogenicity is achieved by disruption of the MHC-II transactivator CIITA and amplification of PDL1 and/or PDL2, genes encoding for immunomodulatory proteins. Most DLBCLs harbor mutations in multiple genes as the result of an aberrant function of the physiologic SHM mechanism. While the causes of this phenomenon are unknown, its widespread activity may have powerful consequences by promoting genomic instability, favoring DNA breaks/chromosomal translocations, and deregulating oncogenes or tumor suppressor genes. Subtype-Associated Alterations Translocations resulting in deregulated MYC and BCL2 and gain-of-function mutations in the H3K27 methyltransferase EZH2 are exquisitely restricted to GCB-DLBCL. Also frequent for this subtype, but of unclear functional role, are truncating mutations in TNFRSF14, encoding a TNF-receptor superfamily member, and mutations of GNA13, encoding a G protein involved in Rho GTPase signaling. Importantly, EZH2 inhibitors showed specific activity against EZH2 mutated tumors in preclinical studies. Constitutive activation of the NF-kB transcription factor represents a hallmark of ABC-DLBCL. The underlying causes are heterogeneous and include gain-of-function mutations in several signal transduction components of the BCR (CD79B and CARD11) and Toll-like receptor (MyD88) signaling pathway or loss-of-function mutations in the NF-kB negative regulator TNFAIP3/A20. While specific NF-kB inhibitors are under development, kinase inhibitors that interfere with BCR signaling (e.g., BTK inhibitors) are emerging as a new treatment paradigm for ABC-DLBCL. Impaired plasma cell differentiation is another major transformation mechanism in this subtype, caused by mutually exclusive lesions deregulating BCL6 and inactivating PRDM1/BLIMP1. In addition to abnormalities leading to constitutive NF-kB responses, several lesions converge on the JAK-STAT signaling pathway preferentially in PMBCL, suggesting a pathogenic role. Moreover, amplification of JAK2 and JMJD2C, encoding for a H3K9 demethylase, may favor epigenetic dysregulation and alter the transcription of multiple genes, including MYC. References Caro, P., Kishan, A.U., Norberg, E., Stanley, I.A., Chapuy, B., Ficarro, S.B., Polak, K., Tondera, D., Gounarides, J., Yin, H., et al. (2012). Cancer Cell 22, 547–560. Cerchietti, L.C., Ghetu, A.F., Zhu, X., Da Silva, G.F., Zhong, S., Matthews, M., Bunting, K.L., Polo, J.M., Farès, C., Arrowsmith, C.H., et al. (2010). Cancer Cell 17, 400–411. Challa-Malladi, M., Lieu, Y.K., Califano, O., Holmes, A.B., Bhagat, G., Murty, V.V., Dominguez-Sola, D., Pasqualucci, L., and Dalla-Favera, R. (2011). Cancer Cell 20, 728–740. Monti, S., Savage, K.J., Kutok, J.L., Feuerhake, F., Kurtin, P., Mihm, M., Wu, B., Pasqualucci, L., Neuberg, D., Aguiar, R.C., et al. (2005). Blood 105, 1851–1861. Morin, R.D., Johnson, N.A., Severson, T.M., Mungall, A.J., An, J., Goya, R., Paul, J.E., Boyle, M., Woolcock, B.W., Kuchenbauer, F., et al. (2010). Nat. Genet. 42, 181–185. Pasqualucci, L., Neumeister, P., Goossens, T., Nanjangud, G., Chaganti, R.S., Küppers, R., and Dalla-Favera, R. (2001). Nature 412, 341–346. Pasqualucci, L., Dominguez-Sola, D., Chiarenza, A., Fabbri, G., Grunn, A., Trifonov, V., Kasper, L.H., Lerach, S., Tang, H., Ma, J., et al. (2011a). Nature 471, 189–195. Shaffer, A.L., 3rd, Young, R.M., and Staudt, L.M. (2012). Annu. Rev. Immunol. 30, 565–610. Steidl, C., and Gascoyne, R.D. (2011). Blood 118, 2659–2669.

132.e1  Cancer Cell 25, January 13, 2014 ©2014 Elsevier Inc.  DOI 10.1016/j.ccr.2013.12.012

SnapShot: diffuse large B cell lymphoma.

SnapShot: diffuse large B cell lymphoma. - PDF Download Free
535KB Sizes 0 Downloads 0 Views