Skip to main content Accessibility help
×
Hostname: page-component-84b7d79bbc-g78kv Total loading time: 0 Render date: 2024-07-26T00:41:43.991Z Has data issue: false hasContentIssue false

13 - WHO classification of lymphomas

Published online by Cambridge University Press:  10 January 2011

William W. L. Choi
Affiliation:
Department of Pathology, University of Hong Kong, Queen Mary Hospital, Hong Kong
Wing C. Chan
Affiliation:
Center for Lymphoma and Leukemia Research, Department of Pathology, Nebraska Medical Center, Omaha, NE, USA
Susan O'Brien
Affiliation:
University of Texas/MD Anderson Cancer Center, Houston
Julie M. Vose
Affiliation:
University of Nebraska Medical Center, Omaha
Hagop M. Kantarjian
Affiliation:
University of Texas/MD Anderson Cancer Center, Houston
Get access

Summary

Introduction

The latest fourth edition of the World Health Organization (WHO) classification of tumors of hematopoietic and lymphoid tissues (WHO classification) was published in 2008. The current and the previous (2001) editions of the WHO classification adopt the guiding principles of the widely accepted Revised European–American Classification of Lymphoid Neoplasms (REAL) classification published in 1994. Both the REAL and the WHO classifications aim at defining distinct disease entities that are both recognizable by pathologists and are meaningful to the clinicians. All available information from morphologic, immunophenotypic, genetic, and clinical assessments was utilized to define non-overlapping disease entities. Additionally, the complexity of the field necessitated the recruitment of a larger number of expert pathologists from around the world, and the inclusion of input from the clinicians in the development of the WHO classification. Broad agreement among the pathologists was required, even at the expense of compromise, which was considered vital for wide acceptance of the WHO classification.

Essential elements of the WHO classification

The WHO classification takes into account a combination of clinical, morphologic, immunophenotypic, and genetic information in recognizing distinct lymphoma entities, although the weight of each of these four parameters varies. Indeed, there is no ‘gold standard’ among these four features when defining individual entities.

Morphology

Morphologic features remain the foundation in the WHO classification and in day-to-day lymphoma diagnosis. Some disease entities, like the various subtypes of Hodgkin lymphomas (HLs), are still defined and diagnosed on morphologic grounds, with support from immunophenotypic results.

Type
Chapter
Information
Publisher: Cambridge University Press
Print publication year: 2010

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. (eds.). WHO Classification of Tumours of the Haematopoietic and Lymphoid Tissues. Lyon, IARC Press, 2008.
Harris, NL, Jaffe, ES, Stein, H, et al. A revised European-American classification of lymphoid neoplasms: a proposal from the International Lymphoma Study Group. Blood 1994;84:1361–92.Google ScholarPubMed
Chan, JKC. Tumors of the lymphoreticular system. In: Fletcher, CDM, ed. Diagnostic Histopathology of Tumors, 3rd edn. Edinburgh, Churchill Livingstone. 2007; 1139–288.Google Scholar
Matutes, E, Catovsky, D. The value of scoring systems for the diagnosis of biphenotypic leukemia and mature B-cell disorders. Leuk Lymphoma 1994;13 Suppl 1:11–14.CrossRefGoogle Scholar
Han, X, Bueso-Ramos, CE. Precursor T-cell acute lymphoblastic leukemia/lymphoblastic lymphoma and acute biphenotypic leukemias. Am J Clin Pathol 2007;127:528–44.CrossRefGoogle ScholarPubMed
Pui, CH, Robison, LL, Look, AT. Acute lymphoblastic leukemia. Lancet 2008;371:1030–43.CrossRefGoogle Scholar
Weng, AP, Ferrando, AA, Lee, W, et al. Activating mutations of NOTCH1 in human T cell acute lymphoblastic leukemia. Science 2004;306:269–71.CrossRefGoogle ScholarPubMed
Wolfe, MS. Gamma-Secretase inhibition and modulation for Alzheimer's disease. Curr Alzheimer Res 2008;5:158–64.CrossRefGoogle ScholarPubMed
,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.
Shimodaira, S, Hidaka, E, Katsuyama, T. Clonal identity of nodular lymphocyte-predominant Hodgkin's disease and T-cell-rich B-cell lymphoma. N Engl J Med 2000;343:1124–5.CrossRefGoogle ScholarPubMed
Chan, WC. Cellular origin of nodular lymphocyte-predominant Hodgkin's lymphoma: immunophenotypic and molecular studies. Semin Hematol 1999;36:242–52.Google ScholarPubMed
Campbell, J, Seymour, JF, Matthews, J, et al. The prognostic impact of bone marrow involvement in patients with diffuse large cell lymphoma varies according to the degree of infiltration and presence of discordant marrow involvement. Eur J Haematol 2006;76:473–80.CrossRefGoogle ScholarPubMed
Chung, R, Lai, R, Wei, P, et al. Concordant but not discordant bone marrow involvement in diffuse large B-cell lymphoma predicts a poor clinical outcome independent of the International Prognostic Index. Blood 2007;110:1278–82.CrossRefGoogle Scholar
Hodges, GF, Lenhardt, TM, Cotelingam, JD. Bone marrow involvement in large-cell lymphoma. Prognostic implications of discordant disease. Am J Clin Pathol 1994;101:305–11.CrossRefGoogle ScholarPubMed
Robertson, , Redman, JR, Butler, JJ, et al. Discordant bone marrow involvement in diffuse large-cell lymphoma: a distinct clinical-pathologic entity associated with a continuous risk of relapse. J Clin Oncol 1991;9:236–42.CrossRefGoogle ScholarPubMed
Fisher, , Jacobson, JO, Ault, KA, et al. Diffuse large cell lymphoma with discordant bone marrow histology. Clinical features and biological implications. Cancer 1989;64:1879–87.3.0.CO;2-D>CrossRefGoogle ScholarPubMed
Yan, Y, Chan, WC, Weisenburger, DD, et al. Clinical and prognostic significance of bone marrow involvement in patients with diffuse aggressive B-cell lymphoma. J Clin Oncol 1995;13:1336–42.CrossRefGoogle ScholarPubMed
Rosenwald, A, Wright, G, Chan, WC, et al. The use of molecular profiling to predict survival after chemotherapy for diffuse large-B-cell lymphoma. N Engl J Med 2002;346:1937–47.CrossRefGoogle ScholarPubMed
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;8:68–74.CrossRefGoogle ScholarPubMed
Iqbal, J, Sanger, WG, Horsman, , et al. BCL2 translocation defines a unique tumor subset within the germinal center B-cell-like diffuse large B-cell lymphoma. Am J Pathol 2004;165:159–66.CrossRefGoogle ScholarPubMed
Bea, S, Zettl, A, Wright, G, et al. Diffuse large B-cell lymphoma subgroups have distinct genetic profiles that influence tumor biology and improve gene-expression-based survival prediction. Blood 2005;106:3183–90.CrossRefGoogle ScholarPubMed
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.CrossRefGoogle ScholarPubMed
Hans, CP, Weisenburger, DD, Vose, JM, et al. A significant diffuse component predicts for inferior survival in grade 3 follicular lymphoma, but cytologic subtypes do not predict survival. Blood 2003;101:2363–7.CrossRefGoogle Scholar
Choi, WW, Weisenburger, DD, Greiner, TC, et al. A new immunostain algorithm classifies diffuse large B-cell lymphoma into molecular subtypes with high accuracy. Clin Cancer Res 2009;15:5494–502.CrossRefGoogle ScholarPubMed
Kobayashi, T, Yamaguchi, M, Kim, S, et al. Microarray reveals differences in both tumors and vascular specific gene expression in de novo CD5+ and CD5- diffuse large B-cell lymphomas. Cancer Res 2003;63:60–6.Google ScholarPubMed
Colomo, L, Loong, F, Rives, S, et al. Diffuse large B-cell lymphomas with plasmablastic differentiation represent a heterogeneous group of disease entities. Am J Surg Pathol 2004;28:736–47.CrossRefGoogle ScholarPubMed
Delecluse, HJ, Anagnostopoulos, I, Dallenbach, F, et al. Plasmablastic lymphomas of the oral cavity: a new entity associated with the human immunodeficiency virus infection. Blood 1997;89:1413–20.Google ScholarPubMed
Dong, HY, Scadden, DT, Leval, L, et al. Plasmablastic lymphoma in HIV-positive patients: an aggressive Epstein-Barr virus-associated extramedullary plasmacytic neoplasm. Am J Surg Pathol 2005;29:1633–41.CrossRefGoogle ScholarPubMed
Oksenhendler, E, Boulanger, E, Galicier, L, et al. High incidence of Kaposi sarcoma-associated herpesvirus-related non-Hodgkin lymphoma in patients with HIV infection and multicentric Castleman disease. Blood 2002;99:2331–6.CrossRefGoogle ScholarPubMed
Nador, RG, Cesarman, E, Chadburn, A, et al. Primary effusion lymphoma: a distinct clinicopathologic entity associated with the Kaposi's sarcoma-associated herpes virus. Blood 1996;88:645–56.Google ScholarPubMed
Carbone, A, Gloghini, A, Cozzi, MR, et al. Expression of MUM-1/IRF4 selectively clusters with primary effusion lymphoma among lymphomatous effusions: implications for disease histogenesis and pathogenesis. Br J Haematol 2000;111:247–57.CrossRefGoogle Scholar
Klein, U, Gloghini, A, Gaidano, G, et al. Gene expression profile analysis of AIDS-related primary effusion lymphoma (PEL) suggests a plasmablastic derivation and identifies PEL-specific transcripts. Blood 2003;101:4115–21.CrossRefGoogle ScholarPubMed
Oyama, T, Yamamoto, K, Asano, N, et al. Age-related EBV-associated B-cell lymphoproliferative disorders constitute a distinct clinicopathologic group: a study of 96 patients. Clin Cancer Res 2007;13:5124–32.CrossRefGoogle ScholarPubMed
Park, S, Lee, J, Ko, YH, et al. The impact of Epstein-Barr virus status on clinical outcome in diffuse large B-cell lymphoma. Blood 2007;110:972–8.CrossRefGoogle ScholarPubMed
Guinee, DG, Perkins, SL, Travis, WD, et al. Proliferation and cellular phenotype in lymphomatoid granulomatosis: implications of a higher proliferation index in B cells. Am J Surg Pathol 1998;22:1093–100.CrossRefGoogle ScholarPubMed
Koss, MN, Hochholzer, L, Langloss, JM, et al. Lymphomatoid granulomatosis: a clinicopathologic study of 42 patients. Pathology 1986;18:283–8.CrossRefGoogle ScholarPubMed
Nishiu, M, Tomita, Y, Nakatsuka, S, et al. Distinct pattern of gene expression in pyothorax-associated lymphoma (PAL), a lymphoma developing in long-standing inflammation. Cancer Sci 2004;95:828–34.CrossRefGoogle Scholar
Cobbers, JM, Wolter, M, Reifenberger, J, et al. Frequent inactivation of CDKN2A and rare mutation of TP53 in PCNSL. Brain Pathol 1998;8:263–76.CrossRefGoogle ScholarPubMed
Boleti, E, Johnson, PW. Primary mediastinal B-cell lymphoma. Hematol Oncol 2007;25:157–63.CrossRefGoogle ScholarPubMed
Rosenwald, A, Wright, G, Leroy, K, et al. Molecular diagnosis of primary mediastinal B cell lymphoma identifies a clinically favorable subgroup of diffuse large B cell lymphoma related to Hodgkin lymphoma. J Exp Med 2003;198:851–62.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.CrossRefGoogle ScholarPubMed
Joos, S, Kupper, M, Ohl, S, et al. Genomic imbalances including amplification of the tyrosine kinase gene JAK2 in CD30+ Hodgkin cells. Cancer Res 2000; 60:549–52.Google ScholarPubMed
Ferreri, AJ, Dognini, GP, Campo, E, et al. Variations in clinical presentation, frequency of hemophagocytosis and clinical behavior of intravascular lymphoma diagnosed in different geographical regions. Haematologica 2007;92:486–92.CrossRefGoogle ScholarPubMed
Matsue, K, Asada, N, Takeuchi, M, et al. A clinicopathological study of 13 cases of intravascular lymphoma: experience in a single institution over a 9-yr period. Eur J Haematol 2008;80:236–44.CrossRefGoogle Scholar
Achten, R, Verhoef, G, Vanuytsel, L, et al. T-cell/histiocyte-rich large B-cell lymphoma: a distinct clinicopathologic entity. J Clin Oncol 2002;20:1269–77.Google ScholarPubMed
Bouabdallah, R, Mounier, N, Guettier, C, et al. T-cell/histiocyte-rich large B-cell lymphomas and classical diffuse large B-cell lymphomas have similar outcome after chemotherapy: a matched-control analysis. J Clin Oncol 2003;21:1271–7.CrossRefGoogle ScholarPubMed
Delsol, G, Lamant, L, Mariame, B, et al. A new subtype of large B-cell lymphoma expressing the ALK kinase and lacking the 2; 5 translocation. Blood 1997;89:1483–90.Google ScholarPubMed
Chikatsu, N, Kojima, H, Suzukawa, K, et al. ALK+, CD30-, CD20- large B-cell lymphoma containing anaplastic lymphoma kinase (ALK) fused to clathrin heavy chain gene (CLTC). Mod Pathol 2003;16:828–32.CrossRefGoogle Scholar
Blum, KA, Lozanski, G, Byrd, JC. Adult Burkitt leukemia and lymphoma. Blood 2004;104:3009–20.CrossRefGoogle ScholarPubMed
Patte, C, Auperin, A, Michon, J, et al. The Societe Francaise d'Oncologie Pediatrique LMB89 protocol: highly effective multiagent chemotherapy tailored to the tumor burden and initial response in 561 unselected children with B-cell lymphomas and L3 leukemia. Blood 2001;97:3370–9.CrossRefGoogle ScholarPubMed
Nyman, H, Adde, M, Karjalainen-Lindsberg, ML, et al. Prognostic impact of immunohistochemically defined germinal center phenotype in diffuse large B-cell lymphoma patients treated with immunochemotherapy. Blood 2007;109:4930–5.CrossRefGoogle ScholarPubMed
Dave, SS, Fu, K, Wright, GW, et al. Molecular diagnosis of Burkitt's lymphoma. N Engl J Med 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. N Engl J Med 2006;354:2419–30.CrossRefGoogle ScholarPubMed
Gouill, S, Talmant, P, Touzeau, C, et al. The clinical presentation and prognosis of diffuse large B-cell lymphoma with t(14;18) and 8q24/c-MYC rearrangement. Haematologica 2007;92:1335–42.CrossRefGoogle Scholar
Chuang, SS, Ye, H, Du, MQ, et al. Histopathology and immunohistochemistry in distinguishing Burkitt lymphoma from diffuse large B-cell lymphoma with very high proliferation index and with or without a starry-sky pattern: a comparative study with EBER and FISH. Am J Clin Pathol 2007;128:558–64.CrossRefGoogle ScholarPubMed
Ott, G, Katzenberger, T, Lohr, A, et al. Cytomorphologic, immunohistochemical, and cytogenetic profiles of follicular lymphoma: 2 types of follicular lymphoma grade 3. Blood 2002;99:3806–12.CrossRefGoogle ScholarPubMed
Bosga-Bouwer, AG, Imhoff, GW, Boonstra, R, et al. Follicular lymphoma grade 3B includes 3 cytogenetically defined subgroups with primary t(14;18), 3q27, or other translocations: t(14;18) and 3q27 are mutually exclusive. Blood 2003;101:1149–54.CrossRefGoogle Scholar
Bosga-Bouwer, AG, Berg, A, Haralambieva, E, et al. Molecular, cytogenetic, and immunophenotypic characterization of follicular lymphoma grade 3B; a separate entity or part of the spectrum of diffuse large B-cell lymphoma or follicular lymphoma?Hum Pathol 2006;37:528–33.CrossRefGoogle ScholarPubMed
Katzenberger, T, Ott, G, Klein, T, et al. Cytogenetic alterations affecting BCL6 are predominantly found in follicular lymphomas grade 3B with a diffuse large B-cell component. Am J Pathol 2004;165:481–90.CrossRefGoogle ScholarPubMed
Swerdlow, SH. Pediatric follicular lymphomas, marginal zone lymphomas, and marginal zone hyperplasia. Am J Clin Pathol 2004;122 Suppl:S98–109.Google ScholarPubMed
Salar, A, Juanpere, N, Bellosillo, B, et al. Gastrointestinal involvement in mantle cell lymphoma: a prospective clinic, endoscopic, and pathologic study. Am J Surg Pathol 2006;30:1274–80.CrossRefGoogle ScholarPubMed
Romaguera, JE, Medeiros, LJ, Hagemeister, FB, et al. Frequency of gastrointestinal involvement and its clinical significance in mantle cell lymphoma. Cancer 2003;97:586–91.CrossRefGoogle ScholarPubMed
Suzuki, R, Takemura, K, Tsutsumi, M, et al. Detection of cyclin D1 overexpression by real-time reverse-transcriptase-mediated quantitative polymerase chain reaction for the diagnosis of mantle cell lymphoma. Am J Pathol 2001;159:425–9.CrossRefGoogle Scholar
Hui, P, Howe, JG, Crouch, J, et al. Real-time quantitative RT-PCR of cyclin D1 mRNA in mantle cell lymphoma: comparison with FISH and immunohistochemistry. Leuk Lymphoma 2003;44:1385–94.CrossRefGoogle ScholarPubMed
Belaud-Rotureau, MA, Parrens, M, Dubus, P, et al. A comparative analysis of FISH, RT-PCR, PCR, and immunohistochemistry for the diagnosis of mantle cell lymphomas. Mod Pathol 2002;15:517–25.CrossRefGoogle Scholar
Specht, K, Kremer, M, Muller, U, et al. Identification of cyclin D1 mRNA overexpression in B-cell neoplasias by real-time reverse transcription-PCR of microdissected paraffin sections. Clin Cancer Res 2002;8:2902–11.Google ScholarPubMed
Howe, JG, Crouch, J, Cooper, D, et al. Real-time quantitative reverse transcription-PCR for cyclin D1 mRNA in blood, marrow, and tissue specimens for diagnosis of mantle cell lymphoma. Clin Chem 2004;50:80–7.CrossRefGoogle ScholarPubMed
Thomazy, VA, Luthra, R, Uthman, MO, et al. Determination of cyclin D1 and CD20 mRNA levels by real-time quantitative RT-PCR from archival tissue sections of mantle cell lymphoma and other non-Hodgkin's lymphomas. J Mol Diagn 2002;4:201–8.CrossRefGoogle ScholarPubMed
Medeiros, LJ, Hai, S, Thomazy, VA, et al. Real-time RT-PCR assay for quantifying cyclin D1 mRNA in B-cell non-Hodgkin's lymphomas. Mod Pathol 2002;15:556–64.CrossRefGoogle ScholarPubMed
Bijwaard, KE, Aguilera, NS, Monczak, Y, et al. Quantitative real-time reverse transcription-PCR assay for cyclin D1 expression: utility in the diagnosis of mantle cell lymphoma. Clin Chem 2001;47:195–201.Google Scholar
Peghini, PE, Fehr, J. Analysis of cyclin D1 expression by quantitative real-time reverse transcription-polymerase chain reaction in the diagnosis of mantle cell lymphoma. Am J Clin Pathol 2002;117:237–45.CrossRefGoogle Scholar
Brizova, H, Kalinova, M, Krskova, L, et al. Quantitative measurement of cyclin D1 mRNA, a potent diagnostic tool to separate mantle cell lymphoma from other B-cell lymphoproliferative disorders. Diagn Mol Pathol 2008;17:39–50.Google ScholarPubMed
O'Malley, DP, Vance, GH, Orazi, A. Chronic lymphocytic leukemia/small lymphocytic lymphoma with trisomy 12 and focal cyclin d1 expression: a potential diagnostic pitfall. Arch Pathol Lab Med 2005;129:92–5.Google ScholarPubMed
Zukerberg, LR, Yang, WI, Arnold, A, et al. Cyclin D1 expression in non-Hodgkin's lymphomas. Detection by immunohistochemistry. Am J Clin Pathol 1995;103:756–60.CrossRefGoogle ScholarPubMed
Miranda, RN, Briggs, RC, Kinney, MC, et al. Immunohistochemical detection of cyclin D1 using optimized conditions is highly specific for mantle cell lymphoma and hairy cell leukemia. Mod Pathol 2000;13:1308–14.CrossRefGoogle ScholarPubMed
Fonseca, R, Blood, EA, Oken, MM, et al. Myeloma and the t(11;14)(q13;q32); evidence for a biologically defined unique subset of patients. Blood 2002;99:3735–41.CrossRefGoogle Scholar
Fu, K, Weisenburger, DD, Greiner, TC, et al. Cyclin D1-negative mantle cell lymphoma: a clinicopathologic study based on gene expression profiling. Blood 2005;106:4315–21.CrossRefGoogle ScholarPubMed
Gesk, S, Klapper, W, Martin-Subero, JI, et al. A chromosomal translocation in cyclin D1-negative/cyclin D2-positive mantle cell lymphoma fuses the CCND2 gene to the IGK locus. Blood 2006;108:1109–10.CrossRefGoogle ScholarPubMed
Wlodarska, I, Dierickx, D, Vanhentenrijk, V, et al. Translocations targeting CCND2, CCND3, and MYCN do occur in t(11;14)-negative mantle cell lymphomas. Blood 2008;111:5683–90.CrossRefGoogle Scholar
Ek, S, Dictor, M, Jerkeman, M, et al. Nuclear expression of the non B-cell lineage Sox11 transcription factor identifies mantle cell lymphoma. Blood 2008;111:800–5.CrossRefGoogle ScholarPubMed
Mozos, A, Royo, C, Hartmann, E, et al. SOX11 expression is highly specific for mantle cell lymphoma and identifies the cyclin D1-negative subtype. Haematologica 2009;94:1555–62.CrossRefGoogle ScholarPubMed
Suarez, F, Lortholary, O, Hermine, O, et al. Infection-associated lymphomas derived from marginal zone B cells: a model of antigen-driven lymphoproliferation. Blood 2006;107:3034–44.CrossRefGoogle Scholar
Dierlamm, J. Genetic abnormalities in marginal zone B-cell lymphoma. Haematologica 2003;88:8–12.Google ScholarPubMed
Streubel, B, Vinatzer, U, Lamprecht, A, et al. T(3;14)(p14.1;q32) involving IGH and FOXP1 is a novel recurrent chromosomal aberration in MALT lymphoma. Leukemia 2005;19:652–8.CrossRefGoogle Scholar
Remstein, ED, Dogan, A, Einerson, RR, et al. The incidence and anatomic site specificity of chromosomal translocations in primary extranodal marginal zone B-cell lymphoma of mucosa-associated lymphoid tissue (MALT lymphoma) in North America. Am J Surg Pathol 2006;30:1546–53.CrossRefGoogle Scholar
Nakamura, S, Ye, H, Bacon, CM, et al. Clinical impact of genetic aberrations in gastric MALT lymphoma: a comprehensive analysis using interphase fluorescence in situ hybridisation. Gut 2007;56:1358–63.CrossRefGoogle ScholarPubMed
Baro, C, Salido, M, Espinet, B, et al. New chromosomal alterations in a series of 23 splenic marginal zone lymphoma patients revealed by Spectral Karyotyping (SKY). Leuk Res 2008;32:727–36.CrossRefGoogle Scholar
Remstein, ED, James, CD, Kurtin, PJ. Incidence and subtype specificity of API2-MALT1 fusion translocations in extranodal, nodal, and splenic marginal zone lymphomas. Am J Pathol 2000;156:1183–8.CrossRefGoogle ScholarPubMed
Sagaert, X, Wolf-Peeters, C, Noels, H, et al. The pathogenesis of MALT lymphomas: where do we stand?Leukemia 2007;21:389–96.CrossRefGoogle ScholarPubMed
Owen, RG, Treon, SP, Al-Katib, A, et al. Clinicopathological definition of Waldenstrom's macroglobulinemia: consensus panel recommendations from the Second International Workshop on Waldenstrom's Macroglobulinemia. Semin Oncol 2003;30:110–15.CrossRefGoogle ScholarPubMed
Offit, K, Parsa, NZ, Filippa D, et al. t(9;14)(p13;q32) denotes a subset of low-grade non-Hodgkin's lymphoma with plasmacytoid differentiation. Blood 1992;80:2594–9.Google Scholar
Iida, S, Rao, PH, Nallasivam, P, et al. The t(9;14)(p13;q32) chromosomal translocation associated with lymphoplasmacytoid lymphoma involves the PAX-5 gene. Blood 1996;88:4110–17.Google Scholar
Schop, RF, Kuehl, WM, Wier, SA, et al. Waldenstrom macroglobulinemia neoplastic cells lack immunoglobulin heavy chain locus translocations but have frequent 6q deletions. Blood 2002;100:2996–3001.CrossRefGoogle ScholarPubMed
Jankovic, GM, Colovic, MD, Wiernik, PH, et al. Multiple karyotypic aberrations in a polymorphous variant of Waldenstrom macroglobulinemia. Cancer Genet Cytogenet 1999;111:77–80.CrossRefGoogle Scholar
Mansoor, A, Medeiros, LJ, Weber, DM, et al. Cytogenetic findings in lymphoplasmacytic lymphoma/Waldenstrom macroglobulinemia. Chromosomal abnormalities are associated with the polymorphous subtype and an aggressive clinical course. Am J Clin Pathol 2001;116:543–9.CrossRefGoogle ScholarPubMed
Wong, KF, So, CC. Waldenstrom macroglobulinemia with karyotypic aberrations involving both homologous 6q. Cancer Genet Cytogenet 2001;124:137–9.CrossRefGoogle ScholarPubMed
Terre, C, Nguyen-Khac, F, Barin, C, et al. Trisomy 4, a new chromosomal abnormality in Waldenstrom's macroglobulinemia: a study of 39 cases. Leukemia 2006;20:1634–6.CrossRefGoogle ScholarPubMed
Chng, WJ, Schop, RF, Price-Troska, T, et al. Gene-expression profiling of Waldenstrom macroglobulinemia reveals a phenotype more similar to chronic lymphocytic leukemia than multiple myeloma. Blood 2006;108:2755–63.CrossRefGoogle ScholarPubMed
Gutierrez, NC, Ocio, EM, Las Rivas, J, et al. Gene expression profiling of B lymphocytes and plasma cells from Waldenstrom's macroglobulinemia: comparison with expression patterns of the same cell counterparts from chronic lymphocytic leukemia, multiple myeloma and normal individuals. Leukemia 2007;21:541–9.CrossRefGoogle ScholarPubMed
Barosi, G, Boccadoro, M, Cavo, M, et al. Management of multiple myeloma and related-disorders: guidelines from the Italian Society of Hematology (SIE), Italian Society of Experimental Hematology (SIES) and Italian Group for Bone Marrow Transplantation (GITMO). Haematologica 2004;89:717–41.Google Scholar
Dimopoulos, MA, Hamilos, G. Solitary bone plasmacytoma and extramedullary plasmacytoma. Curr Treat Options Oncol 2002;3:255–9.CrossRefGoogle ScholarPubMed
Holland, J, Trenkner, DA, Wasserman, TH, et al. Plasmacytoma. Treatment results and conversion to myeloma. Cancer 1992;69:1513–17.3.0.CO;2-X>CrossRefGoogle ScholarPubMed
Debes-Marun, CS, Dewald, GW, Bryant, S, et al. Chromosome abnormalities clustering and its implications for pathogenesis and prognosis in myeloma. Leukemia 2003;17:427–36.CrossRefGoogle Scholar
Fonseca, R, Oken, MM, Harrington, D, et al. Deletions of chromosome 13 in multiple myeloma identified by interphase FISH usually denote large deletions of the q arm or monosomy. Leukemia 2001;15:981–6.CrossRefGoogle ScholarPubMed
Fonseca, R, Barlogie, B, Bataille, R, et al. Genetics and cytogenetics of multiple myeloma: a workshop report. Cancer Res 2004;64:1546–58.CrossRefGoogle ScholarPubMed
Chng, WJ, Price-Troska, T, Gonzalez-Paz, N, et al. Clinical significance of TP53 mutation in myeloma. Leukemia 2007;21:582–4.CrossRefGoogle Scholar
Romagnoli, M, Desplanques, G, Maiga, S, et al. Canonical nuclear factor kappaB pathway inhibition blocks myeloma cell growth and induces apoptosis in strong synergy with TRAIL. Clin Cancer Res 2007;13:6010–18.CrossRefGoogle ScholarPubMed
Zhan, F, Huang, Y, Colla, S, et al. The molecular classification of multiple myeloma. Blood 2006;108:2020–8.CrossRefGoogle ScholarPubMed
Rawstron, AC, Green, MJ, Kuzmicki, A, et al. Monoclonal B lymphocytes with the characteristics of “indolent” chronic lymphocytic leukemia are present in 3.5% of adults with normal blood counts. Blood 2002;100:635–9.CrossRefGoogle ScholarPubMed
Marti, GE, Rawstron, AC, Ghia, P, et al. Diagnostic criteria for monoclonal B-cell lymphocytosis. Br J Haematol 2005;130:325–32.CrossRefGoogle ScholarPubMed
Fung, SS, Hillier, KL, Leger, CS, et al. Clinical progression and outcome of patients with monoclonal B-cell lymphocytosis. Leuk Lymphoma 2007;48:1087–91.CrossRefGoogle ScholarPubMed
Shim, YK, Vogt, RF, Middleton, D, et al. Prevalence and natural history of monoclonal and polyclonal B-cell lymphocytosis in a residential adult population. Cytometry B Clin Cytom 2007;72:344–53.CrossRefGoogle Scholar
Rawstron, AC. Monoclonal B-cell lymphocytosis: good news for patients and CLL investigators. Leuk Lymphoma 2007;48:1057–8.CrossRefGoogle ScholarPubMed
Cessna, MH, Hartung, L, Tripp, S, et al. Hairy cell leukemia variant: fact or fiction? Am J Clin Pathol 2005;123:132–8.CrossRefGoogle ScholarPubMed
Traverse-Glehen, A, Baseggio, L, Bauchu, EC, et al. Splenic red pulp lymphoma with numerous basophilic villous lymphocytes: a distinct clinicopathologic and molecular entity?Blood 2008;111:2253–60.CrossRefGoogle ScholarPubMed
Went, PT, Zimpfer, A, Pehrs, AC, et al. High specificity of combined TRAP and DBA.44 expression for hairy cell leukemia. Am J Surg Pathol 2005;29:474–8.CrossRefGoogle ScholarPubMed
Dunphy, CH. Reaction patterns of TRAP and DBA.44 in hairy cell leukemia, hairy cell variant, and nodal and extranodal marginal zone B-cell lymphomas. Appl Immunohistochem Mol Morphol 2008;16:135–9.CrossRefGoogle ScholarPubMed
Matutes, E. Immunophenotyping and differential diagnosis of hairy cell leukemia. Hematol Oncol Clin North Am 2006;20:1051–63.CrossRefGoogle ScholarPubMed
Matutes, E, Morilla, R, Owusu-Ankomah, K, et al. The immunophenotype of hairy cell leukemia (HCL). Proposal for a scoring system to distinguish HCL from B-cell disorders with hairy or villous lymphocytes. Leuk Lymphoma 1994;14 Suppl 1:57–61.Google ScholarPubMed
Del Giudice, I, Matutes, E, Morilla, R, et al. The diagnostic value of CD123 in B-cell disorders with hairy or villous lymphocytes. Haematologica 2004;89:303–8.Google ScholarPubMed
Munoz, L, Nomdedeu, JF, Lopez, O, et al. Interleukin-3 receptor alpha chain (CD123) is widely expressed in hematologic malignancies. Haematologica 2001;86:1261–9.Google ScholarPubMed
Salomon-Nguyen, F, Valensi, F, Troussard, X, et al. The value of the monoclonal antibody, DBA44, in the diagnosis of B-lymphoid disorders. Leuk Res 1996;20:909–13.CrossRefGoogle Scholar
Papadaki, T, Stamatopoulos, K, Belessi, C, et al. Splenic marginal-zone lymphoma: one or more entities? A histologic, immunohistochemical, and molecular study of 42 cases. Am J Surg Pathol 2007;31:438–46.CrossRefGoogle ScholarPubMed
Hammer, RD, Glick, AD, Greer, JP, et al. Splenic marginal zone lymphoma. A distinct B-cell neoplasm. Am J Surg Pathol 1996;20:613–26.CrossRefGoogle ScholarPubMed
Kluin-Nelemans, HC, Krouwels, MM, Jansen, JH, et al. Hairy cell leukemia preferentially expresses the IgG3-subclass. Blood 1990;75:972–5.Google ScholarPubMed
Forconi, F, Sahota, SS, Raspadori, D, et al. Hairy cell leukemia: at the crossroad of somatic mutation and isotype switch. Blood 2004;104:3312–17.CrossRefGoogle ScholarPubMed
Lamy, T, Loughran, TPCurrent concepts: large granular lymphocyte leukemia. Blood Rev 1999;13:230–40.CrossRefGoogle ScholarPubMed
Lima, M, Almeida, J, Santos, AH, et al. Immunophenotypic analysis of the TCR-Vbeta repertoire in 98 persistent expansions of CD3(+)/TCR-alphabeta(+) large granular lymphocytes: utility in assessing clonality and insights into the pathogenesis of the disease. Am J Pathol 2001;159:1861–8.CrossRefGoogle ScholarPubMed
Langerak, AW, Beemd, R, Wolvers-Tettero, IL, et al. Molecular and flow cytometric analysis of the Vbeta repertoire for clonality assessment in mature TCRalphabeta T-cell proliferations. Blood 2001;98:165–73.CrossRefGoogle ScholarPubMed
O'Malley, DP. T-cell large granular leukemia and related proliferations. Am J Clin Pathol 2007;127:850–9.CrossRefGoogle ScholarPubMed
Hoffmann, T, Libero, G, Colonna, M, et al. Natural killer-type receptors for HLA class I antigens are clonally expressed in lymphoproliferative disorders of natural killer and T-cell type. Br J Haematol 2000;110:525–36.CrossRefGoogle ScholarPubMed
Morice, WG, Kurtin, PJ, Leibson, PJ, et al. Demonstration of aberrant T-cell and natural killer-cell antigen expression in all cases of granular lymphocytic leukemia. Br J Haematol 2003;120:1026–36.CrossRefGoogle Scholar
Lundell, R, Hartung, L, Hill, S, et al. T-cell large granular lymphocyte leukemias have multiple phenotypic abnormalities involving pan-T-cell antigens and receptors for MHC molecules. Am J Clin Pathol 2005;124:937–46.CrossRefGoogle ScholarPubMed
Gorczyca, W, Weisberger, J, Liu, Z, et al. An approach to diagnosis of T-cell lymphoproliferative disorders by flow cytometry. Cytometry 2002;50:177–90.CrossRefGoogle ScholarPubMed
Melenhorst, JJ, Sorbara, L, Kirby, M, et al. Large granular lymphocyte leukemia is characterized by a clonal T-cell receptor rearrangement in both memory and effector CD8(+) lymphocyte populations. Br J Haematol 2001;112:189–94.CrossRefGoogle ScholarPubMed
Melenhorst, JJ, Eniafe, R, Follmann, D, et al. T-cell large granular lymphocyte leukemia is characterized by massive TCRBV-restricted clonal CD8 expansion and a generalized overexpression of the effector cell marker CD57. Hematol J 2003;4:18–25.CrossRefGoogle Scholar
Wong, KF, Chan, JC, Liu, HS, et al. Chromosomal abnormalities in T-cell large granular lymphocyte leukemia: report of two cases and review of the literature. Br J Haematol 2002;116:598–600.CrossRefGoogle Scholar
Man, C, Au, WY, Pang, A, et al. Deletion 6q as a recurrent chromosomal aberration in T-cell large granular lymphocyte leukemia. Cancer Genet Cytogenet 2002;139:71–4.CrossRefGoogle ScholarPubMed
Epling-Burnette, PK, Painter, JS, Chaurasia, P, et al. Dysregulated NK receptor expression in patients with lymphoproliferative disease of granular lymphocytes. Blood 2004;103:3431–9.CrossRefGoogle ScholarPubMed
Pascal, V, Schleinitz, N, Brunet, C, et al. Comparative analysis of NK cell subset distribution in normal and lymphoproliferative disease of granular lymphocyte conditions. Eur J Immunol 2004;34:2930–40.CrossRefGoogle ScholarPubMed
Zambello, R, Falco, M, Della Chiesa, M, et al. Expression and function of KIR and natural cytotoxicity receptors in NK-type lymphoproliferative diseases of granular lymphocytes. Blood 2003;102:1797–805.CrossRefGoogle ScholarPubMed
Dearden, CE, Matutes, E, Cazin, B, et al. High remission rate in T-cell prolymphocytic leukemia with CAMPATH-1H. Blood 2001;98:1721–6.CrossRefGoogle ScholarPubMed
Foucar, K. Mature T-cell leukemias including T-prolymphocytic leukemia, adult T-cell leukemia/lymphoma, and Sezary syndrome. Am J Clin Pathol 2007;127:496–510.CrossRefGoogle ScholarPubMed
Herling, M, Khoury, JD, Washington, LT, et al. A systematic approach to diagnosis of mature T-cell leukemias reveals heterogeneity among WHO categories. Blood 2004;104:328–35.CrossRefGoogle ScholarPubMed
Moid, F, Day, E, Schneider, MA, et al. An indolent case of T-prolymphocytic leukemia with t(3;22)(q21;q11.2) and elevated serum beta2-microglobulin. Arch Pathol Lab Med 2005;129:1164–7.Google Scholar
Soma, L, Cornfield, DB, Prager, D, et al. Unusually indolent T-cell prolymphocytic leukemia associated with a complex karyotype: is this T-cell chronic lymphocytic leukemia?Am J Hematol 2002;71:224–6.CrossRefGoogle ScholarPubMed
Chan, WC, Hans, CP. Genetic and molecular genetic studies in the diagnosis of T and NK cell neoplasia. Hum Pathol 2003;34:314–21.CrossRefGoogle Scholar
Garand, R, Goasguen, J, Brizard, A, et al. Indolent course as a relatively frequent presentation in T-prolymphocytic leukemia. Groupe Francais d'Hematologie Cellulaire. Br J Haematol 1998;103:488–94.CrossRefGoogle ScholarPubMed
Hasserjian, RP, Harris, NL. NK-cell lymphomas and leukemias: a spectrum of tumors with variable manifestations and immunophenotype. Am J Clin Pathol 2007;127:860–8.CrossRefGoogle ScholarPubMed
Chan, JK. Natural killer cell neoplasms. Anat Pathol 1998;3:77–145.Google ScholarPubMed
Nakashima, Y, Tagawa, H, Suzuki, R, et al. Genome-wide array-based comparative genomic hybridization of natural killer cell lymphoma/leukemia: different genomic alteration patterns of aggressive NK-cell leukemia and extranodal Nk/T-cell lymphoma, nasal type. Genes Chromosomes Cancer 2005;44:247–55.CrossRefGoogle ScholarPubMed
Uchiyama, T, Yodoi, J, Sagawa, K, et al. Adult T-cell leukemia: clinical and hematologic features of 16 cases. Blood 1977;50:481–92.Google ScholarPubMed
Hinuma, Y, Nagata, K, Hanaoka, M, et al. Adult T-cell leukemia: antigen in an ATL cell line and detection of antibodies to the antigen in human sera. Proc Natl Acad Sci U S A 1981;78:6476–80.CrossRefGoogle Scholar
Shimoyama, M. Diagnostic criteria and classification of clinical subtypes of adult T-cell leukemia-lymphoma. A report from the Lymphoma Study Group (1984–87). Br J Haematol 1991;79:428–37.CrossRefGoogle Scholar
Matutes, E. Adult T-cell leukaemia/lymphoma. J Clin Pathol 2007;60:1373–7.CrossRefGoogle ScholarPubMed
Karube, K, Ohshima, K, Tsuchiya, T, et al. Expression of FoxP3, a key molecule in CD4CD25 regulatory T cells, in adult T-cell leukemia/lymphoma cells. Br J Haematol 2004;126:81–4.CrossRefGoogle Scholar
Matsubara, Y, Hori, T, Morita, R, et al. Phenotypic and functional relationship between adult T-cell leukemia cells and regulatory T cells. Leukemia 2005;19:482–3.CrossRefGoogle ScholarPubMed
Roncador, G, Garcia, JF, Garcia, JF, et al. FOXP3, a selective marker for a subset of adult T-cell leukemia/lymphoma. Leukemia 2005;19:2247–53.CrossRefGoogle Scholar
Karube, K, Aoki, R, Sugita, Y, et al. The relationship of FOXP3 expression and clinicopathological characteristics in adult T-cell leukemia/lymphoma. Mod Pathol 2008;21:617–25.CrossRefGoogle ScholarPubMed
,Correlation of chromosome abnormalities with histologic and immunologic characteristics in non-Hodgkin's lymphoma and adult T cell leukemia-lymphoma. Fifth International Workshop on Chromosomes in Leukemia-Lymphoma. Blood 1987;70:1554–64.Google Scholar
Datta, A, Bellon, M, Sinha-Datta, U, et al. Persistent inhibition of telomerase reprograms adult T-cell leukemia to p53-dependent senescence. Blood 2006;108:1021–9.CrossRefGoogle ScholarPubMed
Matutes, E, Taylor, GP, Cavenagh, J, et al. Interferon alpha and zidovudine therapy in adult T-cell leukemia lymphoma: response and outcome in 15 patients. Br J Haematol 2001;113:779–84.CrossRefGoogle ScholarPubMed
Hermine, O, Allard, I, Levy, V, et al. A prospective phase II clinical trial with the use of zidovudine and interferon-alpha in the acute and lymphoma forms of adult T-cell leukemia/lymphoma. Hematol J 2002;3:276–82.CrossRefGoogle ScholarPubMed
Tsukasaki, K, Tsushima, H, Yamamura, M, et al. Integration patterns of HTLV-I provirus in relation to the clinical course of ATL: frequent clonal change at crisis from indolent disease. Blood 1997;89:948–56.Google ScholarPubMed
Medeiros, LJ, Elenitoba-Johnson, KS. Anaplastic large cell lymphoma. Am J Clin Pathol 2007;127:707–22.CrossRefGoogle ScholarPubMed
Pulford, K, Lamant, L, Morris, SW, et al. Detection of anaplastic lymphoma kinase (ALK) and nucleolar protein nucleophosmin (NPM)-ALK proteins in normal and neoplastic cells with the monoclonal antibody ALK1. Blood 1997;89:1394–404.Google ScholarPubMed
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.CrossRefGoogle ScholarPubMed
Wlodarska, I, Wolf-Peeters, C, Falini, B, et al. The cryptic inv(2)(p23q35) defines a new molecular genetic subtype of ALK-positive anaplastic large-cell lymphoma. Blood 1998;92:2688–95.Google ScholarPubMed
Ma, Z, Cools, J, Marynen, P, et al. Inv(2)(p23q35) in anaplastic large-cell lymphoma induces constitutive anaplastic lymphoma kinase (ALK) tyrosine kinase activation by fusion to ATIC, an enzyme involved in purine nucleotide biosynthesis. Blood 2000;95:2144–9.Google ScholarPubMed
Colleoni, GW, Bridge, JA, Garicochea, B, et al. ATIC-ALK: a novel variant ALK gene fusion in anaplastic large cell lymphoma resulting from the recurrent cryptic chromosomal inversion, inv(2)(p23q35). Am J Pathol 2000;156:781–9.CrossRefGoogle Scholar
Trinei, M, Lanfrancone, L, Campo, E, et al. A new variant anaplastic lymphoma kinase (ALK)-fusion protein (ATIC-ALK) in a case of ALK-positive anaplastic large cell lymphoma. Cancer Res 2000;60:793–8.Google Scholar
Hernandez, L, Pinyol, M, Hernandez, S, et al. TRK-fused gene (TFG) is a new partner of ALK in anaplastic large cell lymphoma producing two structurally different TFG-ALK translocations. Blood 1999;94:3265–8.Google ScholarPubMed
Rosenwald, A, Ott, G, Pulford, K, et al. t(1;2)(q21;p23) and t(2;3)(p23;q21): two novel variant translocations of the t(2;5)(p23;q35) in anaplastic large cell lymphoma. Blood 1999;94:362–4.Google Scholar
Meech, SJ, McGavran, L, Odom, LF, et al. Unusual childhood extramedullary hematologic malignancy with natural killer cell properties that contains tropomyosin 4–anaplastic lymphoma kinase gene fusion. Blood 2001;98:1209–16.CrossRefGoogle ScholarPubMed
Tort, F, Pinyol, M, Pulford, K, et al. Molecular characterization of a new ALK translocation involving moesin (MSN-ALK) in anaplastic large cell lymphoma. Lab Invest 2001;81:419–26.CrossRefGoogle Scholar
Cools, J, Wlodarska, I, Somers, R, et al. Identification of novel fusion partners of ALK, the anaplastic lymphoma kinase, in anaplastic large-cell lymphoma and inflammatory myofibroblastic tumor. Genes Chromosomes Cancer 2002;34:354–62.CrossRefGoogle ScholarPubMed
Lamant, L, Gascoyne, RD, Duplantier, MM, et al. Non-muscle myosin heavy chain (MYH9): a new partner fused to ALK in anaplastic large cell lymphoma. Genes Chromosomes Cancer 2003;37:427–32.CrossRefGoogle ScholarPubMed
Lamant, L, Meggetto, F, al Saati, T, et al. High incidence of the t(2;5)(p23;q35) translocation in anaplastic large cell lymphoma and its lack of detection in Hodgkin's disease. Comparison of cytogenetic analysis, reverse transcriptase-polymerase chain reaction, and P-80 immunostaining. Blood 1996;87:284–91.Google Scholar
Lamant, L, Dastugue, N, Pulford, K, et al. A new fusion gene TPM3-ALK in anaplastic large cell lymphoma created by a (1;2)(q25;p23) translocation. Blood 1999;93:3088–95.Google ScholarPubMed
Wilton, SD, Eyre, H, Akkari, PA, et al. Assignment of the human a-tropomyosin gene TPM3 to 1q22–>q23 by fluorescence in situ hybridisation. Cytogenet Cell Genet 1995;68:122–4.CrossRefGoogle ScholarPubMed
Mencinger, M, Panagopoulos, I, Andreasson, P, et al. Characterization and chromosomal mapping of the human TFG gene involved in thyroid carcinoma. Genomics 1997;41:327–31.CrossRefGoogle ScholarPubMed
Touriol, C, Greenland, C, Lamant, L, et al. Further demonstration of the diversity of chromosomal changes involving 2p23 in ALK-positive lymphoma: 2 cases expressing ALK kinase fused to CLTCL (clathrin chain polypeptide-like). Blood 2000;95:3204–7.Google Scholar
Gascoyne, RD, Lamant, L, Martin-Subero, JI, et al. ALK-positive diffuse large B-cell lymphoma is associated with Clathrin-ALK rearrangements: report of 6 cases. Blood 2003;102:2568–73.CrossRefGoogle ScholarPubMed
Foss, HD, Anagnostopoulos, I, Araujo, I, et al. Anaplastic large-cell lymphomas of T-cell and null-cell phenotype express cytotoxic molecules. Blood 1996;88:4005–11.Google ScholarPubMed
Salaverria, I, Bea, S, Lopez-Guillermo, A, et al. Genomic profiling reveals different genetic aberrations in systemic ALK-positive and ALK-negative anaplastic large cell lymphomas. Br J Haematol 2008;140:516–26.CrossRefGoogle ScholarPubMed
Zettl, A, Rudiger, T, Konrad, MA, et al. Genomic profiling of peripheral T-cell lymphoma, unspecified, and anaplastic large T-cell lymphoma delineates novel recurrent chromosomal alterations. Am J Pathol 2004;164:1837–48.CrossRefGoogle ScholarPubMed
Savage, KJ, Harris, NL, Vose, JM, et al. ALK-negative anaplastic large-cell lymphoma (ALCL) is clinically and immunophenotypically different from both ALK-positive ALCL and peripheral T-cell lymphoma, not otherwise specified: report from the International Peripheral T-Cell Lymphoma Project. Blood 2008;111:5496–504.CrossRefGoogle Scholar
Falini, B, Pileri, S, Zinzani, PL, et al. ALK+ lymphoma: clinico-pathological findings and outcome. Blood 1999;93:2697–706.Google ScholarPubMed
Gascoyne, RD, Aoun, P, Wu, D, et al. Prognostic significance of anaplastic lymphoma kinase (ALK) protein expression in adults with anaplastic large cell lymphoma. Blood 1999;93:3913–21.Google ScholarPubMed
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.Google ScholarPubMed
ten Berge, RL, Oudejans, JJ, Ossenkoppele, GJ, et al. ALK expression in extranodal anaplastic large cell lymphoma favours systemic disease with (primary) nodal involvement and a good prognosis and occurs before dissemination. J Clin Pathol 2000;53:445–50.CrossRefGoogle Scholar
Sasaki, K, Sugaya, M, Fujita, H, et al. A case of primary cutaneous anaplastic large cell lymphoma with variant anaplastic lymphoma kinase translocation. Br J Dermatol 2004;150:1202–7.CrossRefGoogle ScholarPubMed
Willemze, R, Kerl, H, Sterry, W, et al. EORTC classification for primary cutaneous lymphomas: a proposal from the Cutaneous Lymphoma Study Group of the European Organization for Research and Treatment of Cancer. Blood 1997;90:354–71.Google Scholar
Bonzheim, I, Geissinger, E, Roth, S, et al. Anaplastic large cell lymphomas lack the expression of T-cell receptor molecules or molecules of proximal T-cell receptor signaling. Blood 2004;104:3358–60.CrossRefGoogle ScholarPubMed
Admirand, JH, Rassidakis, GZ, Abruzzo, LV, et al. Immunohistochemical detection of ZAP-70 in 341 cases of non-Hodgkin and Hodgkin lymphoma. Mod Pathol 2004;17:954–61.CrossRefGoogle ScholarPubMed
Kinney, MC, Jones, D. Cutaneous T-cell and NK-cell lymphomas: the WHO-EORTC classification and the increasing recognition of specialized tumor types. Am J Clin Pathol 2007;127:670–86.CrossRefGoogle ScholarPubMed
El Shabrawi-Caelen, L, Kerl, H, Cerroni, L. Lymphomatoid papulosis: reappraisal of clinicopathologic presentation and classification into subtypes A, B, and C. Arch Dermatol 2004;140:441–7.CrossRefGoogle ScholarPubMed
Willemze, R, Beljaards, RC. Spectrum of primary cutaneous CD30 (Ki-1)-positive lymphoproliferative disorders. A proposal for classification and guidelines for management and treatment. J Am Acad Dermatol 1993;28:973–80.CrossRefGoogle ScholarPubMed
Willemze, R, Jaffe, ES, Burg, G, et al. WHO-EORTC classification for cutaneous lymphomas. Blood 2005;105:3768–85.CrossRefGoogle ScholarPubMed
Weiss, LM, Wood, GS, Trela, M, et al. Clonal T-cell populations in lymphomatoid papulosis. Evidence of a lymphoproliferative origin for a clinically benign disease. N Engl J Med 1986;315:475–9.CrossRefGoogle ScholarPubMed
Whittaker, S, Smith, N, Jones, RR, et al. Analysis of beta, gamma, and delta T-cell receptor genes in lymphomatoid papulosis: cellular basis of two distinct histologic subsets. J Invest Dermatol 1991;96:786–91.CrossRefGoogle ScholarPubMed
DeCoteau, JF, Butmarc, JR, Kinney, MC, et al. The t(2;5) chromosomal translocation is not a common feature of primary cutaneous CD30+ lymphoproliferative disorders: comparison with anaplastic large-cell lymphoma of nodal origin. Blood 1996;87:3437–41.Google Scholar
Attygalle, A, Al-Jehani, R, Diss, TC, et al. Neoplastic T cells in angioimmunoblastic T-cell lymphoma express CD10. Blood 2002;99:627–33.CrossRefGoogle ScholarPubMed
Grogg, KL, Attygalle, AD, Macon, WR, et al. Angioimmunoblastic T-cell lymphoma: a neoplasm of germinal-center T-helper cells?Blood 2005;106:1501–2.CrossRefGoogle ScholarPubMed
Grogg, KL, Attygalle, AD, Macon, WR, et al. Expression of CXCL13, a chemokine highly upregulated in germinal center T-helper cells, distinguishes angioimmunoblastic T-cell lymphoma from peripheral T-cell lymphoma, unspecified. Mod Pathol 2006;19:1101–7.CrossRefGoogle ScholarPubMed
Dupuis, J, Boye, K, Martin, N, et al. Expression of CXCL13 by neoplastic cells in angioimmunoblastic T-cell lymphoma (AITL): a new diagnostic marker providing evidence that AITL derives from follicular helper T cells. Am J Surg Pathol 2006;30:490–4.CrossRefGoogle ScholarPubMed
Piccaluga, PP, Agostinelli, C, Califano, A, et al. Gene expression analysis of angioimmunoblastic lymphoma indicates derivation from T follicular helper cells and vascular endothelial growth factor deregulation. Cancer Res 2007;67:10 703–10.CrossRefGoogle Scholar
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.CrossRefGoogle Scholar
Leval, L, Savilo, E, Longtine, J, et al. Peripheral T-cell lymphoma with follicular involvement and a CD4+/BCL6+ phenotype. Am J Surg Pathol 2001;25:395–400.CrossRefGoogle Scholar
Iqbal, J, Weisenburger, DD, Greiner, TC, et al. Molecular signatures to improve diagnosis in peripheral T-cell lymphoma and prognostication in angioimmunoblastic T-cell lymphoma. Blood 2010;115:1026–36.CrossRefGoogle ScholarPubMed
Zettl, A, deLeeuw, R, Haralambieva, E, et al. Enteropathy-type T-cell lymphoma. Am J Clin Pathol 2007;127:701–6.CrossRefGoogle ScholarPubMed
Deleeuw, RJ, Zettl, A, Klinker, E, et al. Whole-genome analysis and HLA genotyping of enteropathy-type T-cell lymphoma reveals 2 distinct lymphoma subtypes. Gastroenterology 2007;132:1902–11.CrossRefGoogle ScholarPubMed
Zettl, A, Ott, G, Makulik, A, et al. Chromosomal gains at 9q characterize enteropathy-type T-cell lymphoma. Am J Pathol 2002;161:1635–45.CrossRefGoogle ScholarPubMed
Tsukasaki, K, Krebs, J, Nagai, K, et al. Comparative genomic hybridization analysis in adult T-cell leukemia/lymphoma: correlation with clinical course. Blood 2001;97:3875–81.CrossRefGoogle ScholarPubMed
Soulier, J, Pierron, G, Vecchione, D, et al. A complex pattern of recurrent chromosomal losses and gains in T-cell prolymphocytic leukemia. Genes Chromosomes Cancer 2001;31:248–54.CrossRefGoogle ScholarPubMed
Siu, LL, Wong, KF, Chan, JK, et al. Comparative genomic hybridization analysis of natural killer cell lymphoma/leukemia. Recognition of consistent patterns of genetic alterations. Am J Pathol 1999;155:1419–25.CrossRefGoogle ScholarPubMed
Arnulf, B, Copie-Bergman, C, Delfau-Larue, MH, et al. Nonhepatosplenic gammadelta T-cell lymphoma: a subset of cytotoxic lymphomas with mucosal or skin localization. Blood 1998;91:1723–31.Google ScholarPubMed
Katoh, A, Ohshima, K, Kanda, M, et al. Gastrointestinal T cell lymphoma: predominant cytotoxic phenotypes, including alpha/beta, gamma/delta T cell and natural killer cells. Leuk Lymphoma 2000;39:97–111.CrossRefGoogle ScholarPubMed
Macon, WR, Levy, NB, Kurtin, PJ, et al. Hepatosplenic alphabeta T-cell lymphomas: a report of 14 cases and comparison with hepatosplenic gammadelta T-cell lymphomas. Am J Surg Pathol 2001;25:285–96.CrossRefGoogle ScholarPubMed
Weidmann, E. Hepatosplenic T cell lymphoma. A review on 45 cases since the first report describing the disease as a distinct lymphoma entity in 1990. Leukemia 2000;14:991–7.CrossRefGoogle ScholarPubMed
Belhadj, K, Reyes, F, Farcet, JP, et al. Hepatosplenic gammadelta T-cell lymphoma is a rare clinicopathologic entity with poor outcome: report on a series of 21 patients. Blood 2003;102:4261–9.CrossRefGoogle ScholarPubMed
Salhany, KE, Macon, WR, Choi, JK, et al. Subcutaneous panniculitis-like T-cell lymphoma: clinicopathologic, immunophenotypic, and genotypic analysis of alpha/beta and gamma/delta subtypes. Am J Surg Pathol 1998;22:881–93.CrossRefGoogle ScholarPubMed
Santucci, M, Pimpinelli, N, Massi, D, et al. Cytotoxic/natural killer cell cutaneous lymphomas. Report of EORTC Cutaneous Lymphoma Task Force Workshop. Cancer 2003;97:610–27.CrossRefGoogle ScholarPubMed
Hoque, SR, Child, FJ, Whittaker, SJ, et al. Subcutaneous panniculitis-like T-cell lymphoma: a clinicopathological, immunophenotypic and molecular analysis of six patients. Br J Dermatol 2003;148:516–25.CrossRefGoogle ScholarPubMed
Takeshita, M, Imayama, S, Oshiro, Y, et al. Clinicopathologic analysis of 22 cases of subcutaneous panniculitis-like CD56- or CD56+ lymphoma and review of 44 other reported cases. Am J Clin Pathol 2004;121:408–16.CrossRefGoogle ScholarPubMed
Sibaud, V, Beylot-Barry, M, Thiebaut, R, et al. Bone marrow histopathologic and molecular staging in epidermotropic T-cell lymphomas. Am J Clin Pathol 2003;119:414–23.CrossRefGoogle ScholarPubMed
Ponti, R, Quaglino, P, Novelli, M, et al. T-cell receptor gamma gene rearrangement by multiplex polymerase chain reaction/heteroduplex analysis in patients with cutaneous T-cell lymphoma (mycosis fungoides/Sezary syndrome) and benign inflammatory disease: correlation with clinical, histological and immunophenotypical findings. Br J Dermatol 2005;153:565–73.CrossRefGoogle ScholarPubMed
Wieselthier, JS, Koh, HK. Sezary syndrome: diagnosis, prognosis, and critical review of treatment options. J Am Acad Dermatol 1990;22:381–401.CrossRefGoogle ScholarPubMed
Vonderheid, EC, Bernengo, MG, Burg, G, et al. Update on erythrodermic cutaneous T-cell lymphoma: report of the International Society for Cutaneous Lymphomas. J Am Acad Dermatol 2002;46:95–106.CrossRefGoogle ScholarPubMed
Yamanaka, K, Clark, R, Dowgiert, R, et al. Expression of interleukin-18 and caspase-1 in cutaneous T-cell lymphoma. Clin Cancer Res 2006;12:376–82.CrossRefGoogle ScholarPubMed
Vowels, BR, Cassin, M, Vonderheid, EC, et al. Aberrant cytokine production by Sezary syndrome patients: cytokine secretion pattern resembles murine Th-2 cells. J Invest Dermatol 1992;99:90–4.CrossRefGoogle Scholar
Vowels, BR, Lessin, SR, Cassin, M, et al. Th-2 cytokine mRNA expression in skin in cutaneous T-cell lymphoma. J Invest Dermatol 1994;103:669–73.CrossRefGoogle Scholar
Tracey, L, Villuendas, R, Dotor, AM, et al. Mycosis fungoides shows concurrent deregulation of multiple genes involved in the TNF signaling pathway: an expression profile study. Blood 2003;102:1042–50.CrossRefGoogle ScholarPubMed
Kari, L, Loboda, A, Nebozhyn, M, et al. Classification and prediction of survival in patients with the leukemic phase of cutaneous T cell lymphoma. J Exp Med 2003;197:1477–88.CrossRefGoogle ScholarPubMed
Jones, JF, Shurin, S, Abramowsky, C, et al. T-cell lymphomas containing Epstein-Barr viral DNA in patients with chronic Epstein-Barr virus infections. N Engl J Med 1988;318:733–41.CrossRefGoogle ScholarPubMed
Kanegane, H, Bhatia, K, Gutierrez, M, et al. A syndrome of peripheral blood T-cell infection with Epstein-Barr virus (EBV) followed by EBV-positive T-cell lymphoma. Blood 1998;91:2085–91.Google ScholarPubMed
Quintanilla-Martinez, L, Kumar, S, Fend, F, et al. Fulminant EBV(+) T-cell lymphoproliferative disorder following acute/chronic EBV infection: a distinct clinicopathologic syndrome. Blood 2000;96:443–51.Google ScholarPubMed
Braeuninger, A, Kuppers, 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. Proc Natl Acad Sci U S A 1997;94:9337–42.CrossRefGoogle ScholarPubMed
Kanzler, H, Kuppers, R, Hansmann, ML, et al. Hodgkin and Reed-Sternberg cells in Hodgkin's disease represent the outgrowth of a dominant tumor clone derived from (crippled) germinal center B cells. J Exp Med 1996;184:1495–505.CrossRefGoogle ScholarPubMed
Marafioti, T, Hummel, M, Foss, HD, et al. Hodgkin and reed-sternberg cells represent an expansion of a single clone originating from a germinal center B-cell with functional immunoglobulin gene rearrangements but defective immunoglobulin transcription. Blood 2000;95:1443–50.Google ScholarPubMed
Muschen, M, Rajewsky, K, Brauninger, A, et al. Rare occurrence of classical Hodgkin's disease as a T cell lymphoma. J Exp Med 2000;191:387–94.CrossRefGoogle ScholarPubMed
Seitz, V, Hummel, M, Marafioti, T, et al. Detection of clonal T-cell receptor gamma-chain gene rearrangements in Reed-Sternberg cells of classic Hodgkin disease. Blood 2000;95:3020–4.Google ScholarPubMed
Skinnider, BF, Mak, TW. The role of cytokines in classical Hodgkin lymphoma. Blood 2002;99:4283–97.CrossRefGoogle ScholarPubMed
Stein, H, Marafioti, T, Foss, HD, et al. Down-regulation of BOB.1/OBF.1 and Oct2 in classical Hodgkin disease but not in lymphocyte predominant Hodgkin disease correlates with immunoglobulin transcription. Blood 2001;97:496–501.CrossRefGoogle Scholar
Jundt, F, Kley, K, Anagnostopoulos, I, et al. Loss of PU.1 expression is associated with defective immunoglobulin transcription in Hodgkin and Reed-Sternberg cells of classical Hodgkin disease. Blood 2002;99:3060–2.CrossRefGoogle ScholarPubMed
Theil, J, Laumen, H, Marafioti, T, et al. Defective octamer-dependent transcription is responsible for silenced immunoglobulin transcription in Reed-Sternberg cells. Blood 2001;97:3191–6.CrossRefGoogle ScholarPubMed
Re, D, Muschen, M, Ahmadi, T, et al. Oct-2 and Bob-1 deficiency in Hodgkin and Reed Sternberg cells. Cancer Res 2001;61:2080–4.Google ScholarPubMed
Schwering, I, Brauninger, A, Klein, U, et al. Loss of the B-lineage-specific gene expression program in Hodgkin and Reed-Sternberg cells of Hodgkin lymphoma. Blood 2003;101:1505–12.CrossRefGoogle ScholarPubMed
Torlakovic, E, Torlakovic, G, Nguyen, PL, et al. The value of anti-pax-5 immunostaining in routinely fixed and paraffin-embedded sections: a novel pan pre-B and B-cell marker. Am J Surg Pathol 2002;26:1343–50.CrossRefGoogle ScholarPubMed
Bellas, C, Santon, A, Manzanal, A, et al. Pathological, immunological, and molecular features of Hodgkin's disease associated with HIV infection. Comparison with ordinary Hodgkin's disease. Am J Surg Pathol 1996;20:1520–4.CrossRefGoogle ScholarPubMed
,International collaboration on HIV and cancer. Highly active antiretroviral therapy and incidence of cancer in human immunodeficiency virus-infected adults. J Natl Cancer Inst 2000;92:1823–30.CrossRefGoogle Scholar
Engels, EA, Pfeiffer, RM, Goedert, JJ, et al. Trends in cancer risk among people with AIDS in the United States 1980–2002. Aids 2006;20:1645–54.CrossRefGoogle Scholar
Clifford, GM, Polesel, J, Rickenbach, M, et al. Cancer risk in the Swiss HIV Cohort Study: associations with immunodeficiency, smoking, and highly active antiretroviral therapy. J Natl Cancer Inst 2005;97:425–32.CrossRefGoogle ScholarPubMed
Engels, EA, Biggar, RJ, Hall, HI, et al. Cancer risk in people infected with human immunodeficiency virus in the United States. Int J Cancer 2008;123:187–94.CrossRefGoogle ScholarPubMed
Kraus, MD, Haley, J. Lymphocyte predominance Hodgkin's disease: the use of BCL6 and CD57 in diagnosis and differential diagnosis. Am J Surg Pathol 2000;24:1068–78.CrossRefGoogle ScholarPubMed
Joos, S, Menz, 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.CrossRefGoogle ScholarPubMed
Chui, DT, Hammond, D, Baird, M, et al. Classical Hodgkin lymphoma is associated with frequent gains of 17q. Genes Chromosomes Cancer 2003;38:126–36.CrossRefGoogle ScholarPubMed
Devilard, E, Bertucci, F, Trempat, P, et al. Gene expression profiling defines molecular subtypes of classical Hodgkin's disease. Oncogene 2002;21:3095–102.CrossRefGoogle ScholarPubMed
Chaperot, L, Bendriss, N, Manches, O, et al. Identification of a leukemic counterpart of the plasmacytoid dendritic cells. Blood 2001;97:3210–17.CrossRefGoogle ScholarPubMed
Petrella, T, Comeau, MR, Maynadie, M, et al. ‘Agranular CD4+ CD56+ hematodermic neoplasm’ (blastic NK-cell lymphoma) originates from a population of CD56+ precursor cells related to plasmacytoid monocytes. Am J Surg Pathol 2002;26:852–62.CrossRefGoogle ScholarPubMed
Urosevic, M, Conrad, C, Kamarashev, J, et al. CD4+CD56+ hematodermic neoplasms bear a plasmacytoid dendritic cell phenotype. Hum Pathol 2005;36:1020–4.CrossRefGoogle Scholar
Jacob, MC, Chaperot, L, Mossuz, P, et al. CD4+ CD56+ lineage negative malignancies: a new entity developed from malignant early plasmacytoid dendritic cells. Haematologica 2003;88:941–55.Google ScholarPubMed
Grouard, G, Rissoan, MC, Filgueira, L, et al. The enigmatic plasmacytoid T cells develop into dendritic cells with interleukin (IL)-3 and CD40-ligand. J Exp Med 1997;185:1101–11.CrossRefGoogle ScholarPubMed
Petrella, T, Bagot, M, Willemze, R, et al. Blastic NK-cell lymphomas (agranular CD4+CD56+ hematodermic neoplasms): a review. Am J Clin Pathol 2005;123:662–75.CrossRefGoogle ScholarPubMed
Herling, M, Jones, D. CD4+/CD56+ hematodermic tumor: the features of an evolving entity and its relationship to dendritic cells. Am J Clin Pathol 2007;127:687–700.CrossRefGoogle ScholarPubMed
Dijkman, R, Doorn, R, Szuhai, K, et al. Gene-expression profiling and array-based CGH classify CD4+CD56+ hematodermic neoplasm and cutaneous myelomonocytic leukemia as distinct disease entities. Blood 2007;109:1720–7.CrossRefGoogle ScholarPubMed
Leroux, D, Mugneret, F, Callanan, M, et al. CD4(+), CD56(+) DC2 acute leukemia is characterized by recurrent clonal chromosomal changes affecting 6 major targets: a study of 21 cases by the Groupe Francais de Cytogenetique Hematologique. Blood 2002;99:4154–9.CrossRefGoogle ScholarPubMed

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.

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.

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.

Available formats
×