Skip to main content Accessibility help
×
Hostname: page-component-848d4c4894-jbqgn Total loading time: 0 Render date: 2024-07-02T03:30:38.786Z Has data issue: false hasContentIssue false

68 - Molecular pathology of lymphoma

from Part 3.6 - Molecular pathology: lymphoma and leukemia

Published online by Cambridge University Press:  05 February 2015

Christof Schneider
Affiliation:
Institute for Cancer Genetics and Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY, USA
Laura Pasqualucci
Affiliation:
Institute for Cancer Genetics and Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY, USA
Riccardo Dalla-Favera
Affiliation:
Department of Genetics and Development, Department of Pathology and Cell Biology, and Department of Microbiology and Immunology, Institute for Cancer Genetics and Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY, USA
Edward P. Gelmann
Affiliation:
Columbia University, New York
Charles L. Sawyers
Affiliation:
Memorial Sloan-Kettering Cancer Center, New York
Frank J. Rauscher, III
Affiliation:
The Wistar Institute Cancer Centre, Philadelphia
Get access

Summary

Introduction

The most recent WHO Classification of Lymphoid Neoplasms describes more than 40 entities that originate from cells in the lymphoid organs at various stages of normal lymphocyte differentiation (1). Of these, ~90% derive from mature B cells, including B-cell non-Hodgkin lymphoma (B-NHL) and Hodgkin lymphoma (HL), while the remaining 10% derive from T-cells and natural killer cells (T-NHL). To date, most of the recurrent genetic alterations involved in the pathogenesis of B-NHL have been identified and, for many of them, the ability to promote lymphomagenesis has been documented in animal models. Conversely, little is known about the pathogenesis of T-NHLs, due to their uncommon occurrence and biological heterogeneity. This chapter will provide an overview of the most common B- and T-NHLs, with emphasis on their molecular pathogenesis.

B-cell development

The development of mature B lymphocytes from hematopoietic stem cells is regulated by the sequential rearrangement of the immunoglobulin (Ig) genes, together with the acquisition or loss of expression of specific proteins. B-cell precursors first rearrange their Ig heavy (IgH) and light chain (IgL) loci in the bone marrow, through the process of V(D)J recombination (2). If successful in completing this process, cells expressing a functional surface B-cell receptor (BCR) will migrate to secondary lymphoid organs as mature, naïve B-cells. Upon T-cell-dependent antigen stimulation, activated B cells can either differentiate directly into antibody-secreting cells, or start proliferating at high rates and form a characteristic structure known as germinal center (GC; Figure 68.1; 2,3). This structure can be schematically compartmentalized into a dark zone, where proliferating centroblasts diversify their Ig variable region (IgV) genes by the process of somatic hypermutation (SHM), to increase antibody affinity for the antigen, and a light zone, where non-dividing centrocytes undergo Ig class-switch recombination (CSR) and are selected based on their ability to produce high-affinity antibodies (Figure 68.1; 3–6). Cells with low affinity towards the antigen are eliminated by apoptosis. Both SHM and CSR require DNA strand breaks and depend on activation-induced cytidine deaminase (AID), which plays a central role in the generation of genetic alterations in most B-NHL (7–9).

Type
Chapter
Information
Molecular Oncology
Causes of Cancer and Targets for Treatment
, pp. 738 - 750
Publisher: Cambridge University Press
Print publication year: 2013

Access options

Get access to the full version of this content by using one of the access options below. (Log in options will check for institutional or personal access. Content may require purchase if you do not have access.)

References

Swerdlow, SH, Campo, E, Harris, NL, et al. WHO Classification of Tumours of Haematopoietic and Lymphoid Tissues. Lyon: International Agency for Research on Cancer (IARC);2008.
Willerford, DM, Swat, W, Alt, FW. Developmental regulation of V(D)J recombination and lymphocyte differentiation. Current Opinion in Genetic Development 1996;6:603–9.CrossRef
Klein, U, Dalla-Favera, R. Germinal centres: role in B-cell physiology and malignancy. Nature Reviews Immunology 2008;8:22–33.CrossRef
Berek, C, Berger, A, Apel, M. Maturation of the immune response in germinal centers. Cell 1991;67:1121–9.CrossRef
Rajewsky, K. Clonal selection and learning in the antibody system. Nature 1996;381:751–8.CrossRef
Jacob, J, Kelsoe, G, Rajewsky, K, Weiss, U. Intraclonal generation of antibody mutants in germinal centres. Nature 1991;354:389–92.CrossRef
Liu, YJ, Arpin, C, de Bouteiller, O, et al. Sequential triggering of apoptosis, somatic mutation and isotype switch during germinal center development. Seminars in Immunology 1996;8:169–77.CrossRef
Muramatsu, M, Kinoshita, K, Fagarasan, S, et al. Class switch recombination and hypermutation require activation-induced cytidine deaminase (AID), a potential RNA editing enzyme. Cell 2000;102:553–63.CrossRef
Revy, P, Muto, T, Levy, Y, et al. Activation-induced cytidine deaminase (AID) deficiency causes the autosomal recessive form of the Hyper-IgM syndrome (HIGM2). Cell 2000;102:565–75.CrossRef
Basso, K, Saito, M, Sumazin, P, et al. Integrated biochemical and computational approach identifies BCL6 direct target genes controlling multiple pathways in normal germinal center B cells. Blood 2010;115:975–84.CrossRef
Phan, RT, Dalla-Favera, R. The BCL6 proto-oncogene suppresses p53 expression in germinal-centre B cells. Nature 2004;432:635–9.CrossRef
Saito, M, Novak, U, Piovan, E, et al. BCL6 suppression of BCL2 via Miz1 and its disruption in diffuse large B cell lymphoma. Proceedings of the National Academy of Sciences USA 2009;106:11 294–9.
Shaffer, AL, Yu, X, He, Y, et al. BCL-6 represses genes that function in lymphocyte differentiation, inflammation, and cell cycle control. Immunity 2000;13:199–212.CrossRef
Tunyaplin, C, Shaffer, AL, AngeliinDuclos, CD, et al. Direct repression of prdm1 by Bcl-6 inhibits plasmacytic differentiation. Journal of Immunology 2004;173:1158–65.CrossRefGoogle ScholarPubMed
Phan, RT, Saito, M, Basso, K, Niu, H, Dalla-Favera, R. BCL6 interacts with the transcription factor Miz-1 to suppress the cyclin-dependent kinase inhibitor p21 and cell cycle arrest in germinal center B cells. Nature Immunology 2005;6:1054–60.CrossRef
Ci, W, Polo, JM, Cerchietti, L, et al. The BCL6 transcriptional program features repression of multiple oncogenes in primary B cells and is deregulated in DLBCL. Blood 2009;113:5536–48.CrossRef
Niu, H, Cattoretti, G, Dalla-Favera, R.BCL6 controls the expression of the B7–1/CD80 costimulatory receptor in germinal center B cells. Journal of Experimental Medicine 2003;198:211–21.CrossRefGoogle ScholarPubMed
Ranuncolo, SM, Polo, JM, Dierov, J, et al. Bcl-6 mediates the germinal center B cell phenotype and lymphomagenesis through transcriptional repression of the DNA-damage sensor ATR. Nature Immunology 2007;8:705–14.CrossRef
Turner, CA, Mack, DH, Davis, MM. Blimp-1, a novel zinc finger-containing protein that can drive the maturation of B lymphocytes into immunoglobulin-secreting cells. Cell 1994;77:297–306.CrossRef
Klein, U, Casola, S, Cattoretti, G, et al. Transcription factor IRF4 controls plasma cell differentiation and class-switch recombination. Nature Immunology 2006;7:773–82.CrossRef
Allman, D, Jain, A, Dent, A, et al. BCL-6 expression during B-cell activation. Blood 1996;87:5257–68.
Saito, M, Gao, J, Basso, K, et al. A signaling pathway mediating downregulation of BCL6 in germinal center B cells is blocked by BCL6 gene alterations in B cell lymphoma. Cancer Cell 2007;12:280–92.CrossRef
Niu, H, Ye, BH, Dalla-Favera, R. Antigen receptor signaling induces MAP kinase-mediated phosphorylation and degradation of the BCL-6 transcription factor. Genes and Development 1998;12:1953–61.CrossRef
von Boehmer, H, Aifantis, I, Gounari, F, et al. Thymic selection revisited: how essential is it? Immunological Reviews 2003;191:62–78.
Kuppers, R, Dalla-Favera, R. Mechanisms of chromosomal translocations in B cell lymphomas. Oncogene 2001;20:5580–94.CrossRef
Tsujimoto, Y, Gorha, J, Cossman, J, Jaffe, E, Croce, CM. The t(14;18) chromosome translocations involved in B-cell neoplasms result from mistakes in VDJ joining. Science 1985;229:1390–3.CrossRef
Tycko, B, Reynolds, TC, Smith, SD, Sklar, J.Consistent breakage between consensus recombinase heptamers of chromosome 9 DNA in a recurrent chromosomal translocation of human T cell leukemia. Journal of Experimental Medicine 1989;169:369–77.Google Scholar
Nussenzweig, MC. Immune receptor editing: revise and select. Cell 1998;95:875–8.CrossRef
Ramiro, AR, Jankovic, M, Eisenreich, T, et al. AID is required for c-myc/IgH chromosome translocations in vivo. Cell 2004;118:431–8.CrossRef
Robbiani, DF, Bothmer, A, Callen, E, et al. AID is required for the chromosomal breaks in c-myc that lead to c-myc/IgH translocations. Cell 2008;135:1028–38.CrossRef
Pasqualucci, L, Bhagat, G, Jankovic, M, et al. AID is required for germinal center-derived lymphomagenesis. Nature Genetics 2008;40:108–12.CrossRef
Jost, PJ, Ruland, J. Aberrant NF-kappaB signaling in lymphoma: mechanisms, consequences, and therapeutic implications. Blood 2007;109:2700–7.
Morris, SW, Kirstein, MN, Valentine, MB, et al. Fusion of a kinase gene, ALK, to a nucleolar protein gene, NPM, in non- Hodgkin's lymphoma. Science 1994;263:1281–4.CrossRef
Pasqualucci, L, Neumeister, P, Goossens, T, et al. Hypermutation of multiple proto-oncogenes in B-cell diffuse large-cell lymphomas. Nature 2001;412:341–6.CrossRef
Chaganti, RS, Nanjanqud, Schmidt H, Teruya-Feldstein, J. Recurring chromosomal abnormalities in non-Hodgkin's lymphoma: biologic and clinical significance. Seminars in Hematology 2000;37:396–411.CrossRef
Kieff, E, Leibowitz, D. Oncogenesis by herpesvirus. In R.A. Weinberg, Editor, Oncogenes and the Molecular Origin of Cancer. Cold Spring Harbor Laboratory Press: Cold Spring Harbor, NY;1989:259.
Tsujimoto, Y, Jaffe, E, Cossman, J, et al. Clustering of breakpoints on chromosome 11 in human B-cell neoplasms with the t(11;14) chromosome translocation. Nature 1985;315:340–3.CrossRef
Tsujimoto, Y, Finger, LR, Yunis, J, Nowwell, PC, Croce, CM. Cloning of the chromosome breakpoint of neoplastic B cells with the t(14;18) chromosome translocation. Science 1984;226:1097–9.CrossRef
Erikson, J, Finan, J, Tsujimoto, Y, Nowell, PC, Croce, CM. The chromosome 14 breakpoint in neoplastic B cells with the t(11;14) translocation involves the immunoglobulin heavy chain locus. Proceedings of the National Academy of Sciences USA 1984;81:4144–8.CrossRef
Wiestner, A, Tehrani, M, Chiorazzi, M, et al. Point mutations and genomic deletions in CCND1 create stable truncated cyclin D1 mRNAs that are associated with increased proliferation rate and shorter survival. Blood 2007;109:4599–606.CrossRef
Seto, M, Yamamoto, K, Iida, S, et al. Gene rearrangement and overexpression of PRAD1 in lymphoid malignancy with t(11;14)(q13;q32) translocation. Oncogene 1992;7:1401–6.
Jiang, W, Kahn, SM, Zhou, P, et al. Overexpression of cyclin D1 in rat fibroblasts causes abnormalities in growth control, cell cycle progression and gene expression. Oncogene 1993;8:3447–57.
Bodrug, SE, Warner, BJ, Bath, ML, et al. Cyclin D1 transgene impedes lymphocyte maturation and collaborates in lymphomagenesis with the myc gene. EMBO Journal 1994;13:2124–30.
Lovec, H, Grzeschiczek, A, Kowalski, MB, Möröy, T. Cyclin D1/bcl-1 cooperates with myc genes in the generation of B-cell lymphoma in transgenic mice. EMBO Journal 1994;13:3487–95.
Stilgenbauer, S, Winkler, D, Ott, G, et al. Molecular characterization of 11q deletions points to a pathogenic role of the ATM gene in mantle cell lymphoma. Blood 1999;94:3262–4.
Bentz, M, Plesch, A, Bullinger, L, et al. t(11;14)-positive mantle cell lymphomas exhibit complex karyotypes and share similarities with B-cell chronic lymphocytic leukemia. Genes, Chromosomes and Cancer 2000;27:285–94.3.0.CO;2-M>CrossRef
Camacho, E, Hernández, L, Hernández, S, et al. ATM gene inactivation in mantle cell lymphoma mainly occurs by truncating mutations and missense mutations involving the phosphatidylinositol-3 kinase domain and is associated with increasing numbers of chromosomal imbalances. Blood 2002;99:238–44.CrossRef
Schaffner, C, Idler, I, Stilgenbauer, S, Döhner, H, Lichter P. Mantle cell lymphoma is characterized by inactivation of the ATM gene. Proceedings of the National Academy of Sciences USA 2000;97:2773–8.CrossRef
Louie, DC, Offit, K, Jaslow, R, et al. p53 overexpression as a marker of poor prognosis in mantle cell lymphomas with t(11;14)(q13;q32). Blood 1995;86:2892–9.
Pinyol, M, Hernández, L, Cazorla, M, et al. Deletions and loss of expression of p16INK4a and p21Waf1 genes are associated with aggressive variants of mantle cell lymphomas. Blood 1997;89:272–80.
Beà, S, Salaverria, I, Armengol, L, et al. Uniparental disomies, homozygous deletions, amplifications, and target genes in mantle cell lymphoma revealed by integrative high-resolution whole-genome profiling. Blood 2009;113:3059–69.CrossRef
Beà, S, Tort, F, Pinyol, M, et al. BMI-1 gene amplification and overexpression in hematological malignancies occur mainly in mantle cell lymphomas. Cancer Research 2001;61:2409–12.
Kridel, R, Meissner, B, Rogic, S, et al. Whole transcriptome sequencing reveals recurrent NOTCH1 mutations in mantle cell lymphoma. Blood 2012;119:1963–71.CrossRef
Puente, XS, Pinyol, M, Quesada, V, et al. Whole-genome sequencing identifies recurrent mutations in chronic lymphocytic leukaemia. Nature 2011;475:101–5.CrossRef
Fabbri, G, Rasi, S, Rossi, D, et al. Analysis of the chronic lymphocytic leukemia coding genome: role of NOTCH1 mutational activation. Journal of Experimental Medicine 2011;208:1389–401.CrossRefGoogle ScholarPubMed
Chapman, CJ, Mockridge, CI, Rowe, M, Rickinson, AB, Stevenson, FK. Analysis of VH genes used by neoplastic B cells in endemic Burkitt's lymphoma shows somatic hypermutation and intraclonal heterogeneity. Blood 1995;85:2176–81.
Chapman, CJ, Zhou, JX, Gregory, C, Rickinson, AB, Stevenson, FK. VH and VL gene analysis in sporadic Burkitt's lymphoma shows somatic hypermutation, intraclonal heterogeneity, and a role for antigen selection. Blood 1996;88:3562–8.
Jain, R, Roncella, S, Hashimoto, S, et al. A potential role for antigen selection in the clonal evolution of Burkitt's lymphoma. Journal of Immunology 1994;153:45–52.Google ScholarPubMed
Tamaru, J, Hummel, M, Marafioti, T, et al. Burkitt's lymphomas express VH genes with a moderate number of antigen-selected somatic mutations. American Journal of Pathology 1995;147:1398–407.Google ScholarPubMed
Dave, SS, Fu, K, Wright, GW, et al. Molecular diagnosis of Burkitt's lymphoma. New England Journal of Medicine 2006;354:2431–42.CrossRefGoogle ScholarPubMed
Hummel, M, Bentink, S, Berger, H, et al. A biologic definition of Burkitt's lymphoma from transcriptional and genomic profiling. New England Journal of Medicine 2006;354:2419–30.CrossRefGoogle ScholarPubMed
Blum, KA, Lozanski, G, Byrd, JC. Adult Burkitt leukemia and lymphoma. Blood 2004;104:3009–20.CrossRef
Hayday, AC, Gillies, SD, Saito, H, et al. Activation of a translocated human c-myc gene by an enhancer in the immunoglobulin heavy-chain locus. Nature 1984;307:334–40.CrossRef
Gu, W, Bhatia, K, Magath, IT, Dang, CV, Dalla-Favera, R. Binding and suppression of the Myc transcriptional activation domain by p107. Science 1994;264:251–4.CrossRef
Meyer, N, Penn, LZ. Reflecting on 25 years with MYC. Nature Reviews Cancer 2008;8:976–90.CrossRef
Amati, B, Brooks, MW, Levy, N, et al. Oncogenic activity of the c-Myc protein requires dimerization with Max. Cell 1993;72:233–45.CrossRef
Dominguez-Sola, D, Ying, CY, Grandori, C, et al. Non-transcriptional control of DNA replication by c-Myc. Nature 2007;448:445–51.CrossRef
Adams, JM, Harris, AW, Pinkert, CA, et al. The c-myc oncogene driven by immunoglobulin enhancers induces lymphoid malignancy in transgenic mice. Nature 1985;318:533–8.CrossRef
Kovalchuk, AL, Qi, CF, Torrey, TA, et al. Burkitt lymphoma in the mouse. Journal of Experimental Medicine 2000;192:1183–90.CrossRefGoogle ScholarPubMed
Gaidano, G, Ballerini, P, Gong, JZ, et al. p53 mutations in human lymphoid malignancies: association with Burkitt lymphoma and chronic lymphocytic leukemia. Proceedings of the National Academy of Sciences USA 1991;88:5413–17.CrossRef
Schmitz, R, Young, RM, Ceribelli, M, et al. Burkitt lymphoma pathogenesis and therapeutic targets from structural and functional genomics. Nature 2012;490:116–20.CrossRef
Richter, J, Schlesner, M, Hoffmann, S, et al. Recurrent mutation of the ID3 gene in Burkitt lymphoma identified by integrated genome, exome and transcriptome sequencing. Nature Genetics 2012;44:1316–20.CrossRef
Love, C, Zun, S, Jima, D, et al. The genetic landscape of mutations in Burkitt lymphoma. Nature Genetics 2012;44:1321–5.CrossRef
Sander, S, Calado, DP, Srinivasan, L, et al. Synergy between PI3K signaling and MYC in Burkitt lymphomagenesis. Cancer Cell 2012;22167–79.
Neri, A, Barriga, F, Inghirami, G, et al. Epstein-Barr virus infection precedes clonal expansion in Burkitt's and acquired immunodeficiency syndrome-associated lymphoma. Blood 1991;77:1092–5.
zur Hausen, H, Schulte-Holthausen, H, Klein, G, et al. EBV DNA in biopsies of Burkitt tumours and anaplastic carcinomas of the nasopharynx. Nature 1970;228:1056–8.CrossRef
Hamilton-Dutoit, SJ, Pallesen, G.A survey of Epstein-Barr virus gene expression in sporadic non-Hodgkin's lymphomas: detection of Epstein-Barr virus in a subset of peripheral T-cell lymphomas. American Journal of Pathology 1992;140:1315–25.Google Scholar
Bende, RJ, Smit, LA, van Noesel, CJ. Molecular pathways in follicular lymphoma. Leukemia 2007;21:18–29.CrossRef
Bakhshi, A, Jensen, JP, Goldman, P, et al. Cloning the chromosomal breakpoint of t(14;18) human lymphomas: clustering around JH on chromosome 14 and near a transcriptional unit on 18. Cell 1985;41:899–906.CrossRef
Cleary, ML, Smith, SD, Sklar, J. Cloning and structural analysis of cDNAs for bcl-2 and a hybrid bcl-2/immunoglobulin transcript resulting from the t(14;18) translocation. Cell 1986;47:19–28.CrossRef
Cleary, ML, Galili, N, Sklar, J.Detection of a second t(14;18) breakpoint cluster region in human follicular lymphomas. Journal of Experimental Medicine 1986;164:315–20.CrossRefGoogle Scholar
Cleary, ML, Sklar, J. Nucleotide sequence of a t(14;18) chromosomal breakpoint in follicular lymphoma and demonstration of a breakpoint-cluster region near a transcriptionally active locus on chromosome 18. Proceedings of the National Academy of Sciences USA 1985;82:7439–43.CrossRef
Roulland, S, Navarro, JM, Grenot, P, et al. Follicular lymphoma-like B cells in healthy individuals: a novel intermediate step in early lymphomagenesis. Journal of Experimental Medicine 2006;203:2425–31.CrossRefGoogle ScholarPubMed
McDonnell, TJ, Korsmeyer, SJ. Progression from lymphoid hyperplasia to high-grade malignant lymphoma in mice transgenic for the t(14; 18). Nature 1991;349:254–6.CrossRef
Morin, RD, Mendez-Lago, M, Mungall, AJ, et al. Frequent mutation of histone-modifying genes in non-Hodgkin lymphoma. Nature 2011;476:298–303.CrossRef
Morin, RD, Johnson, NA, Severson, TM, et al. Somatic mutations altering EZH2 (Tyr641) in follicular and diffuse large B-cell lymphomas of germinal-center origin. Nature Genetics 2010;42:181–5.CrossRef
Pasqualucci, L, Trifonov, V, Fabbri, G, et al. Analysis of the coding genome of diffuse large B-cell lymphoma. Nature Genetics 2011;43:830–7.CrossRef
Davies, AJ, Rosenwald, A, Wright, A, et al. Transformation of follicular lymphoma to diffuse large B-cell lymphoma proceeds by distinct oncogenic mechanisms. British Journal of Haematology 2007;136:286–93.CrossRefGoogle ScholarPubMed
Savage, KJ, Monti, S, Kutok, JL, et al. The molecular signature of mediastinal large B-cell lymphoma differs from that of other diffuse large B-cell lymphomas and shares features with classical Hodgkin lymphoma. Blood 2003;102:3871–9.CrossRef
Monti, S, Savage, KJ, Kutok, JL, et al. Molecular profiling of diffuse large B-cell lymphoma identifies robust subtypes including one characterized by host inflammatory response. Blood 2005;105:1851–61.CrossRef
Alizadeh, AA, Eisen, MB, Davis, RE, et al. Distinct types of diffuse large B-cell lymphoma identified by gene expression profiling. Nature 2000;403:503–11.CrossRef
Basso, K, Dalla-Favera, R. BCL6: master regulator of the germinal center reaction and key oncogene in B cell lymphomagenesis. Advances in Immunology 2010;105:193–210.CrossRef
Cattoretti, G, Pasqualucci, L, Ballon, G, et al. Deregulated BCL6 expression recapitulates the pathogenesis of human diffuse large B cell lymphomas in mice. Cancer Cell 2005;7:445–55.CrossRef
Pasqualucci, L, Dominguez-Sola, D, Chiarenza, A, et al. Inactivating mutations of acetyltransferase genes in B-cell lymphoma. Nature 2011;471:189–95.CrossRef
Challa-Malladi, M, Lieu, YK, Califano, O, et al. Combined genetic inactivation of beta2-Microglobulin and CD58 reveals frequent escape from immune recognition in diffuse large B cell lymphoma. Cancer Cell 2011;20:728–40.CrossRef
Iqbal, J, Sanger, WG, Horsman, DE, et al. BCL2 translocation defines a unique tumor subset within the germinal center B-cell-like diffuse large B-cell lymphoma. American Journal of Pathology 2004;165:159–66.CrossRefGoogle ScholarPubMed
Staudt, LM, Wilson, WH. Focus on lymphomas. Cancer Cell 2002;2:363–6.CrossRef
Pasqualucci, L, Migliazza, A, Basso, K, et al. Mutations of the BCL6 proto-oncogene disrupt its negative autoregulation in diffuse large B-cell lymphoma. Blood 2003;101:2914–23.CrossRef
Iqbal, J, Greiner, TC, Patel, K, et al. Distinctive patterns of BCL6 molecular alterations and their functional consequences in different subgroups of diffuse large B-cell lymphoma. Leukemia 2007;21:2332–43.CrossRef
Pasqualucci, L, Migliazza, A, Fracchiolla, N, et al. BCL-6 mutations in normal germinal center B cells: evidence of somatic hypermutation acting outside Ig loci. Proceedings of the National Academy of Sciences USA 1998;95:11 816–21.
Shen, HM, Peters, A, Baron, B, Zhu, X, Storb, U. Mutation of BCL-6 gene in normal B cells by the process of somatic hypermutation of Ig genes. Science 1998;280:1750–2.CrossRef
Capello, D, Vitolo, U, Pasqualucci, L, et al. Distribution and pattern of BCL-6 mutations throughout the spectrum of B-cell neoplasia. Blood 2000;95:651–9.
Shaffer, AL, III, Young, RM, Staudt, LM. Pathogenesis of human B cell lymphomas. Annual Review of Immunology 2012;30:565–610.CrossRef
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;459:717–21.CrossRef
Kato, M, Sanada, M, Kato, I, et al. Frequent inactivation of A20 in B-cell lymphomas. Nature 2009;459:712–16.CrossRef
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. Journal of Experimental Medicine 2001;194:1861–74.CrossRefGoogle ScholarPubMed
Ngo, VN, Young, RM, Schmitz, R, et al. Oncogenically active MYD88 mutations in human lymphoma. Nature 2011;470:115–19.CrossRef
Lenz, G, Davivs, RE, Ngo, VN, et al. Oncogenic CARD11 mutations in human diffuse large B cell lymphoma. Science 2008;319:1676–9.CrossRef
Ye, BH, Lista, F, Lo Coco, F, et al. Alterations of a zinc finger-encoding gene, BCL-6, in diffuse large-cell lymphoma. Science 1993;262:747–50.CrossRef
Baron, BW, Nucifora, G, McCabe, N, et al. Identification of the gene associated with the recurring chromosomal translocations t(3;14)(q27;q32) and t(3;22)(q27;q11) in B-cell lymphomas. Proceedings of the National Academy of Sciences USA 1993;90:5262–6.CrossRef
Pasqualucci, L, Compagno, M, Houlsworth, J, et al. Inactivation of the PRDM1/BLIMP1 gene in diffuse large B cell lymphoma. Journal of Experimental Medicine 2006;203:311–17.CrossRefGoogle ScholarPubMed
Tam, W, Gomez, M, Chadburn, A, et al. Mutational analysis of PRDM1 indicates a tumor-suppressor role in diffuse large B-cell lymphomas. Blood 2006;107:4090–100.CrossRef
Mandelbaum, J, Bhagat, G, Tang, H, et al. BLIMP1 is a tumor suppressor gene frequently disrupted in activated B cell-like diffuse large B cell lymphoma. Cancer Cell 2010;18:568–79.CrossRef
Savage, KJ, Al-Rajhi, N, Voss, N, et al. Favorable outcome of primary mediastinal large B-cell lymphoma in a single institution: the British Columbia experience. Annals of Oncology 2006;17:123–30.CrossRef
Bentz, M, Barth, TF, Brüderlein, S, et al. Gain of chromosome arm 9p is characteristic of primary mediastinal B-cell lymphoma (MBL): comprehensive molecular cytogenetic analysis and presentation of a novel MBL cell line. Genes, Chromosomes and Cancer 2001;30:393–401.3.0.CO;2-I>CrossRef
Joos, S, Otaño-Joos, MI, Zeigler, S, et al. Primary mediastinal (thymic) B-cell lymphoma is characterized by gains of chromosomal material including 9p and amplification of the REL gene. Blood 1996;87:1571–8.
Feuerhake, F, Kutok, JL, Monti, S, et al. NFkappaB activity, function, and target-gene signatures in primary mediastinal large B-cell lymphoma and diffuse large B-cell lymphoma subtypes. Blood 2005;106:1392–9.CrossRef
Steidl, C, Shah, SP, Woolcock, BW, et al. MHC class II transactivator CIITA is a recurrent gene fusion partner in lymphoid cancers. Nature 2011;471:377–81.CrossRef
The Non-Hodgkin's Lymphoma Classification Project. A clinical evaluation of the International Lymphoma Study Group classification of non-Hodgkin's lymphoma. Blood 1997;89:3909–18.
Muller, AM, Ihorst, G, Mertelsmann, R, Engelhardt, M. Epidemiology of non-Hodgkin's lymphoma (NHL): trends, geographic distribution, and etiology. Annals of Hematology 2005;84:1–12.CrossRef
Bertoni, F, Cazzaniga, Bosshard G, et al. Immunoglobulin heavy chain diversity genes rearrangement pattern indicates that MALT-type gastric lymphoma B cells have undergone an antigen selection process. British Journal of Haematology 1997;97:830–6.CrossRefGoogle ScholarPubMed
Kuppers, R, Klein, U, Hansmann, ML, Rajewsky, K.Cellular origin of human B-cell lymphomas. New England Journal of Medicine 1999;341:1520–9.CrossRefGoogle ScholarPubMed
Parsonnet, J, Hansen, S, Rodriguez, L, et al. Helicobacter pylori infection and gastric lymphoma. New England Journal of Medicine 1994;330:1267–71.CrossRefGoogle ScholarPubMed
Bende, RJ, Aarts, WM, Riedl, RG, et al. Among B cell non-Hodgkin's lymphomas, MALT lymphomas express a unique antibody repertoire with frequent rheumatoid factor reactivity. Journal of Experimental Medicine 2005;201:1229–41.CrossRefGoogle ScholarPubMed
Streubel, B, Lamprecht, A, Dierlamm, J, et al. T(14;18)(q32;q21) involving IGH and MALT1 is a frequent chromosomal aberration in MALT lymphoma. Blood 2003;101:2335–9.CrossRef
Novak, U, Rinaldi, A, Kwee, I, et al. The NF-(kappa)B negative regulator TNFAIP3 (A20) is inactivated by somatic mutations and genomic deletions in marginal zone lymphomas. Blood 2009;113:4918–21.CrossRef
Hu, H, Wang, B, Borde, M, et al. Foxp1 is an essential transcriptional regulator of B cell development. Nature Immunology 2006;7:819–26.CrossRef
O’Leary, HM, Savage, KJ. Update on the World Health Organization classification of peripheral T-cell lymphomas. Current Hematologic Maligancy Reports 2009;4:227–35.CrossRef
Falini, B, Liso, A, Pasqualucci, L, et al. CD30+ anaplastic large-cell lymphoma, null type, with signet-ring appearance. Histopathology 1997;30:90–2.CrossRef
Shiota, M, Nakamura, S, Ichinohasama, R, et al. Anaplastic large cell lymphomas expressing the novel chimeric protein p80NPM/ALK: a distinct clinicopathologic entity. Blood 1995;86:1954–60.
Yee, HT, Ponzoni, M, Merson, A, et al. Molecular characterization of the t(2;5; p23; q35) translocation in anaplastic large cell lymphoma (Ki-1) and Hodgkin's disease. Blood 1996;87:1081–8.
Bai, RY, Dieter, P, Peschel, C, Morris, SW, Duyster, J. Nucleophosmin-anaplastic lymphoma kinase of large-cell anaplastic lymphoma is a constitutively active tyrosine kinase that utilizes phospholipase C-gamma to mediate its mitogenicity. Molecular and Cellular Biology 1998;18:6951–61.CrossRef
Chiarle, R, Gong, JZ, Guasparri, I, et al. NPM-ALK transgenic mice spontaneously develop T-cell lymphomas and plasma cell tumors. Blood 2003;101:1919–27.CrossRef
Kuefer, MU, Look, AT, Pulford, K, et al. Retrovirus-mediated gene transfer of NPM-ALK causes lymphoid malignancy in mice. Blood 1997;90:2901–10.
Lange, K, Uckhert, W, Blankenstein, T, et al. Overexpression of NPM-ALK induces different types of malignant lymphomas in IL-9 transgenic mice. Oncogene 2003;22:517–27.CrossRef
Taylor, GP.The epidemiology of HTLV-I in Europe. Journal of Acquired Immune Deficiency Syndrome and Human Retrovirology 1996;13:S8–14.CrossRefGoogle Scholar
Ferreira, OC, Planelles, V, Rosenblatt, JD. Human T-cell leukemia viruses: epidemiology, biology, and pathogenesis. Blood Reviews 1997;11:91–104.CrossRef
Uchiyama, T. Human T cell leukemia virus type I (HTLV-I) and human diseases. Annual Review of Immunology 1997;15:15–37.CrossRef
Yamada, Y, Kamihira, S. Inactivation of tumor suppressor genes and the progression of adult T-cell leukemia-lymphoma. Leukemia and Lymphoma 2005;46:1553–9.CrossRef
Gaulard, P, Bourguelot, P, Kanavaros, P, et al. Expression of the alpha/beta and gamma/delta T-cell receptors in 57 cases of peripheral T-cell lymphomas: identification of a subset of gamma/delta T-cell lymphomas. American Journal of Pathology 1990;137:617–28.Google ScholarPubMed
de Leval, L, Rickman, DS, Thielen, C, et al. The gene expression profile of nodal peripheral T-cell lymphoma demonstrates a molecular link between angioimmunoblastic T-cell lymphoma (AITL) and follicular helper T (TFH) cells. Blood 2007;109:4952–63.CrossRef
de Leval, L, Bisig, B, Thielen, C, Boniver, J, Gaulard, P. Molecular classification of T-cell lymphomas. Critical Reviews in Oncology/Hematology 2009;72:125–43.CrossRef
Kaneko, Y, Maseiki, N, Sakuri, M, et al. Characteristic karyotypic pattern in T-cell lymphoproliferative disorders with reactive “angioimmunoblastic lymphadenopathy with dysproteinemia-type” features. Blood 1988;72:413–21.
Dogan, A, Attygalle, AD, Kyriakou, C.Angioimmunoblastic T-cell lymphoma. British Journal of Haematology 2003;121:681–91.CrossRefGoogle ScholarPubMed
Küppers, R, Rajewshi, K, Zhao, M, et al. Hodgkin disease: Hodgkin and Reed-Sternberg cells picked from histological sections show clonal immunoglobulin gene rearrangements and appear to be derived from B cells at various stages of development. Proceedings of the National Academy of Sciences USA 1994;91:10 962–6.
Braeuninger, A, Küppers, R, Strickler, JG, et al. Hodgkin and Reed-Sternberg cells in lymphocyte predominant Hodgkin disease represent clonal populations of germinal center-derived tumor B cells. Proceedings of the National Academy of Sciences USA 1997;94:9337–42.CrossRef
Marafioti, T, Hummel, M, Anagnostopolous, I, et al. Origin of nodular lymphocyte-predominant Hodgkin's disease from a clonal expansion of highly mutated germinal-center B cells. New England Journal of Medicine 1997;337:453–8.CrossRefGoogle ScholarPubMed
Ohno, T, Stribley, JA, Wu, G, et al. Clonality in nodular lymphocyte-predominant Hodgkin's disease. New England Journal of Medicine 1997;337:459–65.CrossRefGoogle ScholarPubMed
Kanzler, H, Küppers, R, Hansmann, ML, Rajewsky, K.Hodgkin and Reed-Sternberg cells in Hodgkin's disease represent the outgrowth of a dominant tumor clone derived from (crippled) germinal center B cells. Journal of Experimental Medicine 1996;184:1495–505.CrossRefGoogle ScholarPubMed
Bechtel, D, Kurth, J, Unkel, C, Küppers, R. Transformation of BCR-deficient germinal-center B cells by EBV supports a major role of the virus in the pathogenesis of Hodgkin and posttransplantation lymphomas. Blood 2005;106:4345–50.CrossRef
Mancao, C, Hammerschmidt, W. Epstein-Barr virus latent membrane protein 2A is a B-cell receptor mimic and essential for B-cell survival. Blood 2007;110:3715–21.CrossRef
Joos, S, Manz, CK, Wrobel, G, et al. Classical Hodgkin lymphoma is characterized by recurrent copy number gains of the short arm of chromosome 2. Blood 2002;99:1381–7.CrossRef
Martín-Subero, JI, Gesk, S, Harder, L, et al. Recurrent involvement of the REL and BCL11A loci in classical Hodgkin lymphoma. Blood 2002;99:1474–7.CrossRef
Barth, TF, Martín-Subero, JI, Joos, S, et al. Gains of 2p involving the REL locus correlate with nuclear c-Rel protein accumulation in neoplastic cells of classical Hodgkin lymphoma. Blood 2003;101:3681–6.CrossRef
Otto, C, Giefing, M, Massow, A, et al. Genetic lesions of the TRAF3 and MAP3K14 genes in classical Hodgkin lymphoma. British Journal of Haematology 2012;157:702–8.CrossRefGoogle ScholarPubMed
Cabannes, E, Khan, G, Aillet, F, Jarrett, RF, Hay, RT. Mutations in the IkBa gene in Hodgkin's disease suggest a tumour suppressor role for IkappaBalpha. Oncogene 1999;18:3063–70.CrossRef
Emmerich, F, Meiser, M, Hummel, M, et al. Overexpression of I kappa B alpha without inhibition of NF-kappaB activity and mutations in the I kappa B alpha gene in Reed-Sternberg cells. Blood 1999;94:3129–34.
Emmerich, F, Theurich, S, Hummel, M, et al. Inactivating I kappa B epsilon mutations in Hodgkin/Reed-Sternberg cells. Journal of Pathology 2003;201:413–20.CrossRefGoogle ScholarPubMed
Jungnickel, B, Staratschek-Jox, A, Bräuninger, A, et al. Clonal deleterious mutations in the IkappaBalpha gene in the malignant cells in Hodgkin's lymphoma. Journal of Experimental Medicine 2000;191:395–402.CrossRefGoogle ScholarPubMed
Schmitz, R, Hansmann, ML, Bohle, V, et al. TNFAIP3 (A20) is a tumor suppressor gene in Hodgkin lymphoma and primary mediastinal B cell lymphoma. Journal of Experimental Medicine 2009;206:981–9.CrossRefGoogle ScholarPubMed
Green, MR, Monti, S, Rodig, SJ, et al. Integrative analysis reveals selective 9p24.1 amplification, increased PD-1 ligand expression, and further induction via JAK2 in nodular sclerosing Hodgkin lymphoma and primary mediastinal large B-cell lymphoma. Blood 2010;116:3268–77.CrossRef
Joos, S, Küpper, M, Ohl, S, et al. Genomic imbalances including amplification of the tyrosine kinase gene JAK2 in CD30+ Hodgkin cells. Cancer Research 2000;60:549–52.
Mottok, A, Renné, C, Willenbrock, K, Hansmann, ML, Bräuninger, A. Somatic hypermutation of SOCS1 in lymphocyte-predominant Hodgkin lymphoma is accompanied by high JAK2 expression and activation of STAT6. Blood 2007;110:3387–90.CrossRef
Mottok, A, Renné, C, Seifert, M, et al. Inactivating SOCS1 mutations are caused by aberrant somatic hypermutation and restricted to a subset of B-cell lymphoma entities. Blood 2009;114:4503–6.CrossRef
Weniger, MA, Melzner, I, Menz, CK, et al. Mutations of the tumor suppressor gene SOCS-1 in classical Hodgkin lymphoma are frequent and associated with nuclear phospho-STAT5 accumulation. Oncogene 2006;25:2679–84.CrossRef
Weber-Matthiesen, K, Deerberg, J, Poetsch, M, Grote, W, Schlegelberger, B. Numerical chromosome aberrations are present within the CD30+ Hodgkin and Reed-Sternberg cells in 100% of analyzed cases of Hodgkin's disease. Blood 1995;86:1464–8.
Renne, C, Martín-Subero, JI, Hansmann, ML, Siebert, R.Molecular cytogenetic analyses of immunoglobulin loci in nodular lymphocyte predominant Hodgkin's lymphoma reveal a recurrent IGH-BCL6 juxtaposition. Journal of Molecular Diagnostics 2005;7:352–6.CrossRefGoogle ScholarPubMed
Martín-Subero, JI, Klapper, W, Sotnikova, A, et al. Chromosomal breakpoints affecting immunoglobulin loci are recurrent in Hodgkin and Reed-Sternberg cells of classical Hodgkin lymphoma. Cancer Research 2006;66:10 332–8.

Save book to Kindle

To save this book to your Kindle, first ensure coreplatform@cambridge.org is added to your Approved Personal Document E-mail List under your Personal Document Settings on the Manage Your Content and Devices page of your Amazon account. Then enter the ‘name’ part of your Kindle email address below. Find out more about saving to your Kindle.

Note you can select to save to either the @free.kindle.com or @kindle.com variations. ‘@free.kindle.com’ emails are free but can only be saved to your device when it is connected to wi-fi. ‘@kindle.com’ emails can be delivered even when you are not connected to wi-fi, but note that service fees apply.

Find out more about the Kindle Personal Document Service.

  • Molecular pathology of lymphoma
    • By Christof Schneider, Institute for Cancer Genetics and Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY, USA, Laura Pasqualucci, Institute for Cancer Genetics and Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY, USA, Riccardo Dalla-Favera, Department of Genetics and Development, Department of Pathology and Cell Biology, and Department of Microbiology and Immunology, Institute for Cancer Genetics and Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY, USA
  • Edited by Edward P. Gelmann, Columbia University, New York, Charles L. Sawyers, Memorial Sloan-Kettering Cancer Center, New York, Frank J. Rauscher, III
  • Book: Molecular Oncology
  • Online publication: 05 February 2015
  • Chapter DOI: https://doi.org/10.1017/CBO9781139046947.069
Available formats
×

Save book to Dropbox

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Dropbox.

  • Molecular pathology of lymphoma
    • By Christof Schneider, Institute for Cancer Genetics and Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY, USA, Laura Pasqualucci, Institute for Cancer Genetics and Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY, USA, Riccardo Dalla-Favera, Department of Genetics and Development, Department of Pathology and Cell Biology, and Department of Microbiology and Immunology, Institute for Cancer Genetics and Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY, USA
  • Edited by Edward P. Gelmann, Columbia University, New York, Charles L. Sawyers, Memorial Sloan-Kettering Cancer Center, New York, Frank J. Rauscher, III
  • Book: Molecular Oncology
  • Online publication: 05 February 2015
  • Chapter DOI: https://doi.org/10.1017/CBO9781139046947.069
Available formats
×

Save book to Google Drive

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Google Drive.

  • Molecular pathology of lymphoma
    • By Christof Schneider, Institute for Cancer Genetics and Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY, USA, Laura Pasqualucci, Institute for Cancer Genetics and Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY, USA, Riccardo Dalla-Favera, Department of Genetics and Development, Department of Pathology and Cell Biology, and Department of Microbiology and Immunology, Institute for Cancer Genetics and Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY, USA
  • Edited by Edward P. Gelmann, Columbia University, New York, Charles L. Sawyers, Memorial Sloan-Kettering Cancer Center, New York, Frank J. Rauscher, III
  • Book: Molecular Oncology
  • Online publication: 05 February 2015
  • Chapter DOI: https://doi.org/10.1017/CBO9781139046947.069
Available formats
×