In the past 50 years, we have witnessed explosive growth in the understanding of normal and neoplastic lymphoid cells. B-cell, T-cell, and natural killer (NK)–cell neoplasms in many respects recapitulate normal stages of lymphoid cell differentiation and function, so that they can be to some extent classified according to the corresponding normal stage. Likewise, the molecular mechanisms involved the pathogenesis of lymphomas and lymphoid leukemias are often based on the physiology of the lymphoid cells, capitalizing on deregulated normal physiology by harnessing the promoters of genes essential for lymphocyte function. The clinical manifestations of lymphomas likewise reflect the normal function of lymphoid cells in vivo. The multiparameter approach to classification adopted by the World Health Organization (WHO) classification has been validated in international studies as being highly reproducible, and enhancing the interpretation of clinical and translational studies. In addition, accurate and precise classification of disease entities facilitates the discovery of the molecular basis of lymphoid neoplasms in the basic science laboratory.

Mature B-cell and T/natural killer (NK)–cell neoplasms are clonal tumors of B cells, T cells, or NK cells that in many respects recapitulate stages of normal B-cell or T-cell differentiation. Although classifications of lymphoid malignancies have historically treated lymphomas and leukemias separately, this distinction is now appreciated as artificial. However, as the precursor neoplasms—lymphoblastic lymphomas/leukemias and acute myeloid leukemias—are closely related from a clinical and biological perspective, they will not be covered in this review. In addition, plasma cell neoplasms often have been treated separately from other lymphoid neoplasms, but this segregation is equally artificial, and the World Health Organization (WHO) classification of lymphoid neoplasms1  includes plasma disorders. A recent review by Kyle and Rajkumar in this series comprehensively discussed plasma cell myeloma,2  and thus we will not include it further in this review.

As immunologists and pathologists have come to understand the ontogeny of lymphoid cells, it has been tempting to base the classification of lymphoid malignancies strictly on the corresponding normal stage. However, some neoplasms—for example, hairy cell leukemia—do not clearly correspond to a normal B-cell differentiation stage, and some clinically distinctive neoplasms may exhibit immunophenotypic heterogeneity, such as hepatosplenic T-cell lymphoma, which is derived from either α/β or γ/δ T cells. Thus, the normal counterpart of the neoplastic cell cannot at this time be the sole basis for classification.

Cancers are increasingly recognized as genetic diseases, with precise molecular alterations often defining entities. However, progress in identifying the molecular pathogenesis of lymphoid malignancies has in most instances followed an accurate description of the disease by pathologists, based on morphologic, immunophenotypic and clinical parameters (Table 1). This paradigm is exemplified by both mantle cell lymphoma (MCL)3,4  and anaplastic large cell lymphoma (ALCL),5  in which careful pathologic descriptions preceded the recognition of CCND1 and ALK, respectively, as genes critical to their pathogenesis. However, the process is iterative, and once the underlying molecular cause is known, we gain access to new diagnostic tools that help better define the borderlines of the disease (Figure 1). Thus, the use of monoclonal antibodies reactive in routine sections to cyclin D1 and the ALK tyrosine kinase helped define the morphologic spectrum of MCL and ALCL. This partnership between the pathologist and the molecular biologist is crucial in the classification of disease, and should be equally applicable to other organ systems.

Table 1

Pathogenetic insights based on a disease-oriented approach to lymphoma classification

Lymphomas associated with infectious agents  
    Nasal, cutaneous and systemic NK/T-cell lymphomas EBV 
    Adult T-cell leukemia/lymphoma HTLV1 
    Marginal zone lymphomas H pylori, B burgdorferi, C jejuni, Hepatitis C, and others 
    Primary effusion lymphoma, LBCL associated with multicentric CD HHV-8/ KSHV 
    Plasmablastic, Burkitt, DLBCL, CHL EBV (subset of cases) 
Lymphomas with deregulation of apoptosis and survival pathways  
    Follicular lymphoma BCL2/IGH@ 
    MALT lymphomas API2/MALT1 and variants 
Lymphomas with deregulation of the cell cycle  
    Mantle cell lymphoma CCND1/IGH@ 
    Burkitt's lymphoma MYC/IGH@ and variants 
Lymphomas with deregulation of cell signaling or transcriptional regulation  
    Anaplastic large cell lymphoma NPM/ALK and variants 
    Diffuse large B-cell lymphomas BCL6, NFκB, Stat6 
Lymphomas associated with host susceptibility factors, congenital or acquired  
    Enteropathy-associated T-cell lymphoma Genetics, gliadin allergy 
    Extranodal and systemic EBV + T/NK cell lymphomas Genetics, host response to EBV 
    Hepatosplenic T-cell lymphoma Immunosuppression combined with chronic antigenic stimulation 
    Lymphomatoid granulomatosis Partial immune dysfunction and EBV 
    Burkitt lymphoma Polyclonal B-cell activation with or without immunosuppression (malaria, HIV) 
    Posttransplantation and other iatrogenic lymphoproliferative disorders Iatrogenic immunosuppression 
Lymphomas associated with infectious agents  
    Nasal, cutaneous and systemic NK/T-cell lymphomas EBV 
    Adult T-cell leukemia/lymphoma HTLV1 
    Marginal zone lymphomas H pylori, B burgdorferi, C jejuni, Hepatitis C, and others 
    Primary effusion lymphoma, LBCL associated with multicentric CD HHV-8/ KSHV 
    Plasmablastic, Burkitt, DLBCL, CHL EBV (subset of cases) 
Lymphomas with deregulation of apoptosis and survival pathways  
    Follicular lymphoma BCL2/IGH@ 
    MALT lymphomas API2/MALT1 and variants 
Lymphomas with deregulation of the cell cycle  
    Mantle cell lymphoma CCND1/IGH@ 
    Burkitt's lymphoma MYC/IGH@ and variants 
Lymphomas with deregulation of cell signaling or transcriptional regulation  
    Anaplastic large cell lymphoma NPM/ALK and variants 
    Diffuse large B-cell lymphomas BCL6, NFκB, Stat6 
Lymphomas associated with host susceptibility factors, congenital or acquired  
    Enteropathy-associated T-cell lymphoma Genetics, gliadin allergy 
    Extranodal and systemic EBV + T/NK cell lymphomas Genetics, host response to EBV 
    Hepatosplenic T-cell lymphoma Immunosuppression combined with chronic antigenic stimulation 
    Lymphomatoid granulomatosis Partial immune dysfunction and EBV 
    Burkitt lymphoma Polyclonal B-cell activation with or without immunosuppression (malaria, HIV) 
    Posttransplantation and other iatrogenic lymphoproliferative disorders Iatrogenic immunosuppression 

LBCL indicates large B-cell lymphoma.

Figure 1

Schematic diagram illustrating evolution of the entity MCL. MCL was recognized in Kiel classification and modified Rappaport classification as centrocytic lymphoma and lymphocytic lymphoma of intermediate differentiation (IDL), respectively.6  Precise criteria for the distinction from other morphologically similar lymphomas were lacking. The recognition of a characteristic immunophenotype (CD5+, CD23, CD10, monoclonal B cell) helped better define the entity. The identification of the t(11;14) resulting in CCNDI/IGH translocation in virtually all cases of MCL led to the use of cyclin D1 detection by immunohistochemistry for diagnosis. In addition, secondary genetic events such as p53 and p16 deletion/mutation were identified in high-grade variants of MCL, which had been recognized histologically as the “blastoid” subtype. The data derived from immunophenotypic and genetic studies are integrated, culminating in our current definition of the disease. PDL indicates poorly differentiated lymphocytic; WDL, well-differentiated lymphocytic; DHL, diffuse histiocytic lymphoma; and MZL, marginal zone lymphoma.

Figure 1

Schematic diagram illustrating evolution of the entity MCL. MCL was recognized in Kiel classification and modified Rappaport classification as centrocytic lymphoma and lymphocytic lymphoma of intermediate differentiation (IDL), respectively.6  Precise criteria for the distinction from other morphologically similar lymphomas were lacking. The recognition of a characteristic immunophenotype (CD5+, CD23, CD10, monoclonal B cell) helped better define the entity. The identification of the t(11;14) resulting in CCNDI/IGH translocation in virtually all cases of MCL led to the use of cyclin D1 detection by immunohistochemistry for diagnosis. In addition, secondary genetic events such as p53 and p16 deletion/mutation were identified in high-grade variants of MCL, which had been recognized histologically as the “blastoid” subtype. The data derived from immunophenotypic and genetic studies are integrated, culminating in our current definition of the disease. PDL indicates poorly differentiated lymphocytic; WDL, well-differentiated lymphocytic; DHL, diffuse histiocytic lymphoma; and MZL, marginal zone lymphoma.

Close modal

The first description of what we now recognize as a lymphoma is generally attributed to Thomas Hodgkin in 1832 (Table 2).46  The first use of the term lymphosarcoma is attributed to Rudolf Virchow in 1863 (reviewed in Trumper et al8 ). In 1898 and 1902, Carl Sternberg and Dorothy Reed independently described the characteristic binucleate and multinucleate giant cells that came to be called the Reed-Sternberg or Sternberg-Reed cell.7  Sternberg felt that Hodgkin disease was an inflammatory process, related to tuberculosis, but Reed disagreed.47  She made several other key observations: the general health of the patient before the onset of the disease is usually excellent; there was an early peak among children and young adults; and Hodgkin disease usually presents as painless progressive cervical adenopathy without leukemia. She proposed that the pathologic picture is sufficient for diagnosis. Having considerable success as a fellow in pathology, she sought a teaching post at Johns Hopkins Medical School. Her chairman, William Welch, answered, “No women had ever held a teaching position in the school, and there would be great opposition to it.”47  In her diary she noted, “Suddenly, as I saw what I had to face in acceptance of injustice—I knew that I couldn't take it.”47  She left pathology and took up a career in public health.

Table 2

Milestones in the evolution of the classification of lymphoid neoplasms

YearReferencePrincipal contributorsEvent
1806 * J. Alibert Clinical description of mycosis fungoides 
1828 * R. Carswell “Cancer cerebriformis of the lymphatic glands and spleen”; the first case of what was later recognized as “Hodgkin's disease” 
1832 46  T. Hodgkin “On some morbid appearances of the absorbent glands and spleen”; clinical report of what would come to be known as Hodgkin disease 
1865 * S. Wilks Proposes the eponym “Hodgkin's disease” 
1845, 1863 * R. Virchow Describes both leukemia and lymphosarcoma 
1898, 1902 * C. Sternberg, D. Reed Define the microscopic features of the neoplastic cell of Hodgkin disease, establishing an accurate micropscopic description of the disease, the first lymphoma to be defined histologically 
1914 * J. Ewing Describe “reticulosarcomas” (reticulum cell sarcomas) of bone and lymphoid organs 
1928 * C. Oberling Describe “reticulosarcomas” (reticulum cell sarcomas) of bone and lymphoid organs 
1930 * F. Roulet Describe “reticulosarcomas” (reticulum cell sarcomas) of bone and lymphoid organs 
1916 * C. Sternberg Describes “leukosarkomatose,” a process with characteristic features of precursor T-lymphoblastic lymphoma 
1925 9,11  N. Brill Describe “giant follicle hyperplasia” and “follicular lymphadenopathy”—processes with features of follicular lymphoma and florid follicular hyperplasia 
1927  D. Symmers  
1941, 1942 12,13  E. Gall, T. Mallory Accurate description of follicular lymphoma, and propose the first modern classification system of lymphoma 
1947 * H. Jackson, F. Parker Propose a classification of Hodgkin disease 
1958 14  D. Burkitt Describes clinical syndrome of Burkitt lymphoma in African children 
1960 15  P. Nowell Phytohemaggutinin used to “transform” lymphocytes in vitro 
1961 16  G. O'Conor Provides histopathologic description of Burkitt lymphoma 
1964 17  M. A. Epstein Description of viral particles, the Epstein-Barr virus, in cultured cells from Burkitt lymphoma 
1956, 1966 18,19  H. Rappaport Proposes an alternative classification for “non-Hodgkin's” lymphoma 
1966 20  R. Lukes, J. Butler Propose the modern classification of Hodgkin lymphoma 
1972 21  H. Stein Identifies high levels of IgM in “histiocytic” lymphomas 
1973  K. Lennert Lennert and colleagues (R. Gérard-Marchant, I. Hamlin, K.L., F. Rilke, A. G. Stansfeld, and J. A. M. van Unnik) meet to form the European Lymphoma Club, the predecessor of the European Association for Haematopathology 
1974 22,60,63  K. Lennert Proposes the Kiel classification of lymphomas 
1974 23  C. Taylor Immunohistochemical detection of immunoglobulin within cells within FFPE sections 
  D. Mason  
1974 24  E. Jaffe Identification of complement receptors on cells of “nodular lymphoma” linking them to the lymphoid follicle 
1975 25   Failed consensus meeting of proponents of lymphoma classification systems; Lennert (Kiel), Lukes and Collins, Dorfman, Bennett (BNLI); Mathe (WHO), and Rappaport, leading to Working Formulation study by NCI 
1975 26  E. Southern Development of Southern blot technique to separate and analyze DNA fragments 
1976 27  G. Klein Identification of t(8;14)(q24;q32) as a recurrent translocation in Burkitt lymphoma 
1979 28  S. Fukuhara, J. Rowley Identification of t(14;18)(q32;q21) as a recurrent translocation in “lymphocytic lymphoma” (follicular lymphoma) 
1979 29  A. McMichael First monoclonal antibody to a human leukocyte differentiation antigen, later defined as CD1a 
1980-1982 30,,33  H. Stein, S. Poppema, R. Warnke, D. Mason Characterization of lymphoid cells by immunohistochemistry on frozen and paraffin sections 
1982 34  A. Bernard, L. Boumsell First international workshop on Human Leucocyte Differentiation Antigens (HLDA) 
1982 35,36  P. Leder, R. Dalla-Favera, C. Croce Cloning of MYC gene, and idenfication of MYC and IGH@ as reciprocal partners in t(8;14)(q24;q32) 
1982 37  J. Yunis Recurrent translocations identified in follicular lymphoma, Burkitt lymphoma, and chronic lymphocytic leukemia 
1982 25  C. Berard, R. Dorfman, V. DeVitaS. Rosenberg Publication of “Non-Hodgkin's lymphoma pathologic classification project. National Cancer Institute sponsored study of classifications of non-Hodgkin's lymphomas: summary and description of a Working Formulation for clinical usage” 
1985 38  K. Mullis Development of the polymerase chain reaction (PCR) technique for amplification of specific DNA sequences 
1986 39  T. Cremer Development of in situ hybridization techniques for analysis of chromosome aberrations in interphase nuclei 
1991-1992 40  P. Isaacson, H. Stein Founding of the International Lymphoma Study Group (ILSG) and publication of consensus report on mantle cell lymphoma 
1994 41  R. Kuppers, K. Rajewsky Identification of IgH gene rearrangements in Reed-Sternberg cells picked from tissue sections of classical Hodgkin lymphoma 
1994 42  N. Harris, ILSG Publication of the Revised European-American Classification of Lymphoid Neoplasms (REAL) 
1997 43  J. Armitage and colleagues Validation of the REAL classification by the International Lymphoma Classification Project study 
2000 44  L. Staudt and colleagues Application of gene expression profiling to human lymphomas 
2001 45  E.A.H.P., S.H. Publication of WHO Monograph: Pathology and Genetics: Tumours of Hematopoietic and Lymphoid Tissues (3rd edition) 
2008 1  E.A.H.P., S.H. WHO classification of Tumours of Haematopoietic and Lymphoid Tissues (4th Edition) 
YearReferencePrincipal contributorsEvent
1806 * J. Alibert Clinical description of mycosis fungoides 
1828 * R. Carswell “Cancer cerebriformis of the lymphatic glands and spleen”; the first case of what was later recognized as “Hodgkin's disease” 
1832 46  T. Hodgkin “On some morbid appearances of the absorbent glands and spleen”; clinical report of what would come to be known as Hodgkin disease 
1865 * S. Wilks Proposes the eponym “Hodgkin's disease” 
1845, 1863 * R. Virchow Describes both leukemia and lymphosarcoma 
1898, 1902 * C. Sternberg, D. Reed Define the microscopic features of the neoplastic cell of Hodgkin disease, establishing an accurate micropscopic description of the disease, the first lymphoma to be defined histologically 
1914 * J. Ewing Describe “reticulosarcomas” (reticulum cell sarcomas) of bone and lymphoid organs 
1928 * C. Oberling Describe “reticulosarcomas” (reticulum cell sarcomas) of bone and lymphoid organs 
1930 * F. Roulet Describe “reticulosarcomas” (reticulum cell sarcomas) of bone and lymphoid organs 
1916 * C. Sternberg Describes “leukosarkomatose,” a process with characteristic features of precursor T-lymphoblastic lymphoma 
1925 9,11  N. Brill Describe “giant follicle hyperplasia” and “follicular lymphadenopathy”—processes with features of follicular lymphoma and florid follicular hyperplasia 
1927  D. Symmers  
1941, 1942 12,13  E. Gall, T. Mallory Accurate description of follicular lymphoma, and propose the first modern classification system of lymphoma 
1947 * H. Jackson, F. Parker Propose a classification of Hodgkin disease 
1958 14  D. Burkitt Describes clinical syndrome of Burkitt lymphoma in African children 
1960 15  P. Nowell Phytohemaggutinin used to “transform” lymphocytes in vitro 
1961 16  G. O'Conor Provides histopathologic description of Burkitt lymphoma 
1964 17  M. A. Epstein Description of viral particles, the Epstein-Barr virus, in cultured cells from Burkitt lymphoma 
1956, 1966 18,19  H. Rappaport Proposes an alternative classification for “non-Hodgkin's” lymphoma 
1966 20  R. Lukes, J. Butler Propose the modern classification of Hodgkin lymphoma 
1972 21  H. Stein Identifies high levels of IgM in “histiocytic” lymphomas 
1973  K. Lennert Lennert and colleagues (R. Gérard-Marchant, I. Hamlin, K.L., F. Rilke, A. G. Stansfeld, and J. A. M. van Unnik) meet to form the European Lymphoma Club, the predecessor of the European Association for Haematopathology 
1974 22,60,63  K. Lennert Proposes the Kiel classification of lymphomas 
1974 23  C. Taylor Immunohistochemical detection of immunoglobulin within cells within FFPE sections 
  D. Mason  
1974 24  E. Jaffe Identification of complement receptors on cells of “nodular lymphoma” linking them to the lymphoid follicle 
1975 25   Failed consensus meeting of proponents of lymphoma classification systems; Lennert (Kiel), Lukes and Collins, Dorfman, Bennett (BNLI); Mathe (WHO), and Rappaport, leading to Working Formulation study by NCI 
1975 26  E. Southern Development of Southern blot technique to separate and analyze DNA fragments 
1976 27  G. Klein Identification of t(8;14)(q24;q32) as a recurrent translocation in Burkitt lymphoma 
1979 28  S. Fukuhara, J. Rowley Identification of t(14;18)(q32;q21) as a recurrent translocation in “lymphocytic lymphoma” (follicular lymphoma) 
1979 29  A. McMichael First monoclonal antibody to a human leukocyte differentiation antigen, later defined as CD1a 
1980-1982 30,,33  H. Stein, S. Poppema, R. Warnke, D. Mason Characterization of lymphoid cells by immunohistochemistry on frozen and paraffin sections 
1982 34  A. Bernard, L. Boumsell First international workshop on Human Leucocyte Differentiation Antigens (HLDA) 
1982 35,36  P. Leder, R. Dalla-Favera, C. Croce Cloning of MYC gene, and idenfication of MYC and IGH@ as reciprocal partners in t(8;14)(q24;q32) 
1982 37  J. Yunis Recurrent translocations identified in follicular lymphoma, Burkitt lymphoma, and chronic lymphocytic leukemia 
1982 25  C. Berard, R. Dorfman, V. DeVitaS. Rosenberg Publication of “Non-Hodgkin's lymphoma pathologic classification project. National Cancer Institute sponsored study of classifications of non-Hodgkin's lymphomas: summary and description of a Working Formulation for clinical usage” 
1985 38  K. Mullis Development of the polymerase chain reaction (PCR) technique for amplification of specific DNA sequences 
1986 39  T. Cremer Development of in situ hybridization techniques for analysis of chromosome aberrations in interphase nuclei 
1991-1992 40  P. Isaacson, H. Stein Founding of the International Lymphoma Study Group (ILSG) and publication of consensus report on mantle cell lymphoma 
1994 41  R. Kuppers, K. Rajewsky Identification of IgH gene rearrangements in Reed-Sternberg cells picked from tissue sections of classical Hodgkin lymphoma 
1994 42  N. Harris, ILSG Publication of the Revised European-American Classification of Lymphoid Neoplasms (REAL) 
1997 43  J. Armitage and colleagues Validation of the REAL classification by the International Lymphoma Classification Project study 
2000 44  L. Staudt and colleagues Application of gene expression profiling to human lymphomas 
2001 45  E.A.H.P., S.H. Publication of WHO Monograph: Pathology and Genetics: Tumours of Hematopoietic and Lymphoid Tissues (3rd edition) 
2008 1  E.A.H.P., S.H. WHO classification of Tumours of Haematopoietic and Lymphoid Tissues (4th Edition) 
*

Reviewed in reference 8.

The term “reticulum cell sarcoma” is attributed to Ewing, Oberling, and Roulet, who each described tumors of large cells, thought to be related to the supporting fibrous reticulum of lymphoid tissues.8  While the origin and function of these cells were unknown, the term reticulum cell sarcoma came to be used for “large cell” neoplasms—as distinguished from “lymphosarcoma,” which was applied to those composed of smaller cells more recognizable as lymphocytes.

In 1925, Brill and colleagues described patients with enlargement of lymph nodes and spleen, characterized pathologically by a proliferation of lymphoid follicles9 ; additional cases were reported by Symmers,10  who also described progression to a large-cell neoplasm.11  In 1941, Edward A. Gall and Tracy B. Mallory, pathologists at Massachusetts General Hospital, proposed a morphologic classification, based on review of their own collection of 618 patients for whom clinical data were available.12  They recognized follicular lymphoma (FL) as a distinctive morphologic and clinical entity, and described instances of histologic progression over time.13  This was the first widely used classification of lymphomas in the United States; this classification included Hodgkin disease as a separate type of lymphoma, but further subdivisions of Hodgkin disease were proposed by Jackson and Parker.48 

The Rappaport classification was initially published in 1956 in a report primarily focused on FL,18  and fully developed in the Armed Forces Institute of Pathology (AFIP) fascicle of 1966. Rappaport questioned the concept of the time that FL could arise from reactive lymphoid follicles, and to avoid this confusion, he proposed the term “nodular” to replace “follicular” when describing the pattern of the lymphoma. He further observed that most cytologic types of lymphoma could have either a nodular or diffuse pattern, including Hodgkin lymphoma. Thus, the Rappaport classification took as its primary stratification the pattern of the lymphoma—nodular or diffuse.19  The previously well-defined “FL” of Gall and Mallory was lost amid 4 categories of “nodular lymphoma”: well differentiated lymphocytic, poorly differentiated lymphocytic, mixed lymphocytic and histiocytic, and histiocytic. Within each tumor type, those with a nodular pattern in general had a better prognosis than those with a diffuse pattern. Despite the fact that by 1966, the phenomenon of lymphocyte transformation had been recognized,15  the Rappaport classification continued to regard neoplasms of large cells as derived from nonlymphoid stromal or other cell types. Thus, the term “histiocytic” replaced the terms “reticulum cell or clasmatocyte,” and “undifferentiated” replaced the “stem cell” of Gall and Mallory. This classification was widely accepted in the United States, where the high frequency of FL meant that simply recognition of a nodular pattern was highly clinically useful in predicting prognosis.

In the same era, in 1966, Lukes and Butler published a new classification of Hodgkin disease, recognizing the new categories of nodular sclerosis and mixed cellularity types, formerly lumped together as “Hodgkin granuloma.”20  They also defined what came to be known as nodular lymphocyte predominant Hodgkin lymphoma (NLPHL), describing 2 forms rich in normal lymphocytes and histiocytes that could have either a nodular or predominantly diffuse growth pattern. This subtype contained a variant of the classical Reed-Sternberg cell, which they termed the L&H cell, often informally referred to as a “popcorn cell.” The subsequent Rye conference reduced the 6 subtypes of Lukes and Butler to 4, combining the nodular and diffuse forms of lymphocytic and histiocytic into lymphocytic predominance (LP), and the reticular and diffuse fibrosis forms of Hodgkin disease into lymphocyte depleted.49  The lymphocyte depleted group was the most problematic of the Lukes-Butler scheme, and probably included cases of classical Hodgkin lymphoma as well as pleomorphic B-cell and T-cell lymphomas.50  Nevertheless, the subsequent 50 years have seen remarkably few changes in the Lukes-Butler classification of Hodgkin lymphoma, with most of its basic principles affirmed by subsequent biological studies.51,52 

In the 1960s, 2 discoveries revolutionized the understanding of the immune system and its neoplasms. These were (1) the potential of lymphocytes—which had been thought to be end-stage, terminally differentiated cells—to transform into large, proliferating cells in response to mitogens or antigens;15  and (2) the existence of several distinct lymphocyte lineages (T, B, and NK) that could not be reliably predicted by morphology, but that had different functions and physiology (Figures 2, 3).53,54  In the early 1970s, lymphoid cells were found to have surface antigens or receptors that could be exploited to identify the lineage of both normal and neoplastic cells.55,56  These observations led to the recognition that lymphomas were tumors of the immune system. One of the first lymphomas to be subjected to this analysis was Rappaport's nodular lymphoma, which was shown to be of follicular B-cell origin using these new techniques.24  Similarly, tissue lysates created from biopsy specimens of “reticulum cell sarcomas” showed high levels of IgM, providing evidence for a B-cell derivation.21  The “E rosette” phenomenon used sheep erythrocytes to identify the as-yet-undiscovered CD2 antigen on T cells; this method identified the cells of Sezary syndrome and most lymphoblastic lymphomas to be of T-cell derivation.57,58 

Figure 2

Diagrammatic representation of B-cell differentiation and relationship to major B-cell neoplasms. B-cell neoplasms correspond to stages of B-cell maturation, even though the precise cell counterparts are not known in all instances. Precursor B cells that mature in the bone marrow may undergo apoptosis or develop into mature naive B cells that, following exposure to antigen and blast transformation, may develop into short-lived plasma cells or enter the germinal center (GC), where somatic hypermutation and heavy chain class-switching occur. Centroblasts, the transformed cells of the GC, either undergo apoptosis or develop into centrocytes. Post-GC cells include both long-lived plasma cells and memory/marginal zone B cells. Most B cells are activated within the GC, but T cell–independent activation can take place outside of the GC and also probably leads to memory-type B cells. Monocytoid B cells, many of which lack somatic hypermutation, are not illustrated. AG indicates antigen; and FDC, folllicular dendritic cell. Red bar represents immunoglobulin heavy chain gene (IGH@) rearrangement; blue bar, immunoglobulin light chain gene (IGL) rearrangement; and black insertions in the red and blue bars indicate somatic hypermutation.

Figure 2

Diagrammatic representation of B-cell differentiation and relationship to major B-cell neoplasms. B-cell neoplasms correspond to stages of B-cell maturation, even though the precise cell counterparts are not known in all instances. Precursor B cells that mature in the bone marrow may undergo apoptosis or develop into mature naive B cells that, following exposure to antigen and blast transformation, may develop into short-lived plasma cells or enter the germinal center (GC), where somatic hypermutation and heavy chain class-switching occur. Centroblasts, the transformed cells of the GC, either undergo apoptosis or develop into centrocytes. Post-GC cells include both long-lived plasma cells and memory/marginal zone B cells. Most B cells are activated within the GC, but T cell–independent activation can take place outside of the GC and also probably leads to memory-type B cells. Monocytoid B cells, many of which lack somatic hypermutation, are not illustrated. AG indicates antigen; and FDC, folllicular dendritic cell. Red bar represents immunoglobulin heavy chain gene (IGH@) rearrangement; blue bar, immunoglobulin light chain gene (IGL) rearrangement; and black insertions in the red and blue bars indicate somatic hypermutation.

Close modal
Figure 3

Diagrammatic representation of T-cell differentiation and function. Lymphoid progenitors enter the thymus where precursor T cells develop into varied types of naive T cells. The cells of the innate immune system include NK cells, γδ T cells, and NK-like T cells. These cells constitute a primitive type of immune response that lacks both specificity and memory. In the adaptive immune system, αβ T cells leave the thymus, where, upon exposure to antigen, they may undergo blast transformation and develop further into CD4+ and CD8+ effector and memory T cells. T cells of the adaptive immune system are heterogeneous and functionally complex, and include naive, effector (regulatory and cytotoxic), and memory T cells. Another specific type of effector T cells is the follicular helper T-cell that is found in GCs (TFH). Upon antigenic stimulation, T-cell responses may occur independent of the GC, or in the context of a GC reaction. The lymphomas of the innate immune system are predominantly extranodal in presentation, mirroring the distribution of the functional components of this system.113  T-cell lymphomas of the adaptive immune system present primarily in adults, and are mainly nodal in origin.

Figure 3

Diagrammatic representation of T-cell differentiation and function. Lymphoid progenitors enter the thymus where precursor T cells develop into varied types of naive T cells. The cells of the innate immune system include NK cells, γδ T cells, and NK-like T cells. These cells constitute a primitive type of immune response that lacks both specificity and memory. In the adaptive immune system, αβ T cells leave the thymus, where, upon exposure to antigen, they may undergo blast transformation and develop further into CD4+ and CD8+ effector and memory T cells. T cells of the adaptive immune system are heterogeneous and functionally complex, and include naive, effector (regulatory and cytotoxic), and memory T cells. Another specific type of effector T cells is the follicular helper T-cell that is found in GCs (TFH). Upon antigenic stimulation, T-cell responses may occur independent of the GC, or in the context of a GC reaction. The lymphomas of the innate immune system are predominantly extranodal in presentation, mirroring the distribution of the functional components of this system.113  T-cell lymphomas of the adaptive immune system present primarily in adults, and are mainly nodal in origin.

Close modal

In response to the new information, pathologists appropriately began to apply it to the classification of lymphomas. In the 1970s, several European and American groups published proposals for the classification of lymphomas.59-62  The first and most significant of these efforts was led by Karl Lennert in Kiel, Germany, who first recognized that many lymphomas resembled germinal center cells, and had used ultrastructural studies to identify follicular dendritic cells in normal and neoplastic lymph nodes, linking lymphomas with a nodular growth pattern to the lymphoid follicle.63  The Kiel classification was published in final form in 1974, fully described in a monograph in 1978,64  and later updated.65  Lymphomas were classified according to a hypothetical scheme of lymphocyte differentiation, and the nomenclature reflected the putative normal counterpart of the neoplastic cells. Although the majority of the neoplasms described were B-cell, several well-defined types of T-cell lymphomas were included. The neoplasms were grouped according to histologic features into low-grade malignancy (predominance of small cells or “-cytes”) and high-grade malignancy (predominance of “-blasts”). This classification became widely used in Europe, but never supplanted the Rappaport classification, popular among clinicians, in the U.S.

During the same era, clinicians were advancing their knowledge regarding the clinical behavior of lymphomas and patterns of spread through advances in the treatment and staging of lymphoid neoplasms.66,67  As treatment regimens improved, the heterogeneity among different types of lymphomas became more apparent.68,69  However, the use of different classification systems in clinical studies made it difficult to compare published results from different centers. Several meetings were held to try to break the deadlock, the last of which was held at Airlie House in Warrenton, Virginia, in 1975.8  The inability of the pathologists to develop consensus and agree on a common approach led to a National Cancer Institute–directed study to evaluate the 6 published schemes. None of the schemes proved clearly superior in predicting survival, and since the pathologists would not agree to using one scheme over the others, the Working Formulation (WF) for the non-Hodgkin lymphomas (NHL) was developed to translate among them.25 

The WF stratified lymphomas according to clinical outcome, based on the survival of patients in clinical trials conducted in the 1970s.25  It co-opted terminology from the pathology literature related to cytologic grade (low grade, intermediate grade, and high grade) and created clinical groupings that were intended to provide a clinical guide for patient management. The categories closely followed those of the Rappaport classification,19  with updated terminology from Lukes and Collins.59  Since the scheme did not use immunophenotyping, which was felt at the time to be beyond the reach of the routine pathology laboratory, and since it attempted to cover all entities in all classifications in a few categories, the categories were of necessity heterogeneous. It is therefore not surprising that pathologists could not use these categories reproducibly.70  The WF substituted the term “large cell” for “histiocytic” and divided large cell lymphomas into 2 types: large cell and large cell immunoblastic. This separation split the diffuse large-cell lymphomas into 2 different treatment groups (intermediate and high grade), based on questionable morphologic distinctions.71  These distinctions were not reproducible, and the clinical groupings could not be validated. In many respects the WF was a step backward, since it did not recognize well-defined entities such as centrocytic lymphoma from the Kiel classification, which is now known as mantle cell lymphoma (MCL), and made no use of widely available immunologic knowledge techniques for classification or diagnosis—for example, lumping both T-cell lymphomas and many diffuse B-cell lymphomas in a single category (diffuse mixed small and large cell).

Although not intended to serve as a freestanding classification scheme, the WF was a convenient guide to therapy, quickly became popular among clinicians, and was adopted for use in many centers in the U.S. for clinical trials. Following the publication of the WF, the political situation was unchanged. The Kiel classification remained popular in Europe and in Asia; the WF largely replaced the Rappaport classification in the U.S., but many centers used the Lukes-Collins classification. This situation caused continued problems for both pathologists and clinicians in interpreting the results of published studies. In addition, in the 1980s and early 1990s, many new diseases were described that were not included in either classification, including anaplastic large cell lymphoma (ALCL), lymphomas of the mucosa-associated lymphoid tissues (MALT), and adult T-cell leukemia/lymphoma.

In 1975, Kohler and Milstein created the hybridoma technology that led to the development of monoclonal antibodies.72  Before this revolution, cell-surface marker and immunohistochemistry studies had been performed with rabbit or sheep polyclonal antisera, but these reagents were hampered by limited supply, lack of standardization, and the relatively few antigens that were recognized. In 1979, the first monoclonal antibody against a human lymphocyte differentiation antigen was generated against an antigen expressed on normal thymocytes.29  The antigen recognized by NA1/34 was later designated as CD1a in the human leukocyte differentiation antigen (HLDA) system.34  The CD nomenclature, which stood for “clusters of differentiation,” came into use through a series of workshops organized to bring order out of chaos in this newly emerging field. The initial workshops dealt only with antigens expressed on the cell membrane, all of which were given a CD designation, but later workshops began to look at antigens expressed in the nucleus and cytoplasm.73  By the most recent eighth meeting of what is now called Human Cell Differentiation Molecules workshop, a total of 350 CD designations had been defined.

In these early years, monoclonal antibodies were applied to cells in suspension, or somewhat later to cryostat tissue sections. However, a paradigm shift occurred when David Mason and others not only adapted the techniques to routine formalin-fixed paraffin embedded (FFPE) sections, but changed the strategy for screening monoclonals to look for those antibodies that would have optimal reactivity in FFPE sections.23,30,74  These advances had a dramatic impact on the field. First, one did not have to ensure that tissues were snap-frozen or made into cell suspension to perform immunophenotypic studies. Second, the better morphology of paraffin sections improved the pathologist's ability to characterize positive and negative cells. This technology was rapidly adopted by pathologists both at academic centers and in community hospitals. Characteristic immunophenotypes for many B-cell and T-cell malignancies were becoming recognized, and the ready accessibility of monoclonal antibodies adapted for use in immunohistochemistry in routine paraffin sections made new discoveries easily transportable and testable in different laboratories.

In parallel, there was equally dramatic progress in understanding the genetics of lymphoid malignancies. Recurrent cytogenetic abnormalities were identified, following the early observations of recurring cytogenetic alterations in myeloid leukemias.75  The first to be recognized were the t(14;18)(q32;q21) of FL, and t(8;14)(q24;q32) of Burkitt lymphoma.27,28,37  Subsequent studies led to the cloning of the genes involved in these translocations. The laboratories of Philip Leder and Carlo Croce in 1982 both identified MYC as the gene that was translocated into the immunoglobulin genes in human Burkitt lymphoma,35,36 and other similar discoveries soon followed, such as the BCL2/IGH@ in FL76  and the CCND1/IGH@ in MCL.77,78  The most common paradigm for translocations involving the immunoglobulin heavy chain gene, IGH@ at 14q24, is that a cellular proto-oncogene comes under the influence of the IGH@ promoter. There are also less frequent but parallel alterations involving the T-cell receptor genes in T-cell malignancies.

The process of rearrangement of the immunoglobulin and T-cell receptor genes during normal lymphoid cell development was discovered, and the technology exploited by using rearrangement of the antigen receptor genes as markers of both lineage and clonality in lymphoid neoplasms.79  It was later shown that B cells that have transited the germinal center show evidence of somatic hypermutation of the IGH@ variable region genes.80  Thus, this knowledge could be exploited, not only to show lineage, but stage of differentiation, at least within the B-cell system.81,82 

In addition, the development of polymerase chain reaction (PCR)– and fluorescence in situ hybridization (FISH)–based strategies meant that genetic testing no longer required karyotyping of viable cells or labor-intensive Southern blot analysis of fresh or snap-frozen material. The new techniques permitted analysis of antigen receptor gene and oncogene rearrangements on FFPE sections, facilitating the testing of routine biopsies for clinical diagnosis.83-86  Another critical advantage of the PCR-based strategies was the ability to investigate genetic alterations at the single-cell level. This was particularly important in the study of Hodgkin lymphoma, in which the neoplastic cells represented only a minor component of the entire tumor mass. PCR techniques coupled with single-cell microdissection led to the demonstration of IGH@ gene rearrangements in Reed-Sternberg cells, ending the long debate regarding the origin of the malignant cell.41,87 

While translocations involving oncogenes or tumor suppressor genes were demonstrated to be of major importance in the pathogenesis of many lymphomas, integration of viral genomes into the neoplastic cells was demonstrated for several lymphoid malignancies. Epstein-Barr virus (EBV) was shown to be especially promiscuous and capable of transformation of B cells, T cells, and NK cells.17,88-90  HTLV1 and HHV-8/KSHV were identified as playing a role in the pathogenesis of adult T-cell leukemia/lymphoma,91  primary effusion lymphoma,92  and the lymphoma associated with multicentric Castleman disease.93-95 

From its initial description until the 1990s, the nature and lineage of the Reed-Sternberg cell and the inflammatory infiltrate that comprise Hodgkin disease was debated—was this an infectious or inflammatory process with bizarre cells infected by a virus, or a malignant process with a prominent inflammatory component—and what was the lineage of the infected or malignant cell? Hypotheses and bits of supporting evidence suggested almost every known immune cell type: reticulum cell, dendritic cell, histiocyte/macrophage, B cell, and T cell. The major problem for many years was the relative infrequency of the abnormal cells compared with reactive cells, making it difficult to apply immunophenotyping and genetic techniques to the question.

In the 1970s and 1980s, a number of cell lines were developed from patients with advanced-stage Hodgkin disease, and cytogenetic analysis of both these cells and fresh tumor specimens demonstrated abnormal karyotypes; together with the clinical aggressiveness of the disease, these observations confirmed that the disease was indeed a malignancy.96  In the early 1980s, Stein and colleagues at Kiel immunized mice with Hodgkin cell lines established by Diehl and associates, and developed the Ki-1 antibody, recognizing the CD30 antigen, which was found to be expressed by all Hodgkin and Reed-Sternberg (HRS) cells and by rare lymphoid blasts in normal lymphoid tissues.97  In vitro studies revealed that CD30 expression could be induced in B and T cells by mitogens and by EBV infection, suggesting that CD30+ HRS cells might be related to lymphoid cells.5,98  In the 1980s, the development of antibodies that detected B cell– and T cell–specific antigens in paraffin sections, providing sufficient morphologic detail to recognize HRS cells, showed expression of the B-cell antigen CD20 on some of the HRS cells in a proportion of patients with classical Hodgkin lymphoma (CHL).99,100  The detection of EBV genomes in HRS cells provided additional support for a lymphoid and possibly B-cell origin.101,102  Further evidence pointing toward a B-cell derivation came from the observation of composite lymphomas and metachronous CHL and NHLs, in which the NHL component was nearly always of B-cell origin, most commonly FL.103  Finally, in the 1990s, the PCR technique and primers specific for target sequences of the IGH@ and T-cell receptor (TCR) loci became available, and a method of isolating single HRS cells from frozen sections by microdissection was developed.80  The application of these techniques revealed that HRS cells were clonal B cells in 98% of the patients studied, thus confirming that they were neoplastic and of B-cell origin.41,82  This latter finding led to the change of the designation of Hodgkin disease into Hodgkin lymphoma in the WHO classification of lymphoid neoplasms.

With the recognition that nearly all instances of CHL show evidence of B-cell lineage, the following question arose: why do HRS cells express so few B-cell antigens, lack expression of immunoglobulin, and display markers not normally ex-pressed by B cells, such as CD15, TARC, or T cell–associated antigens?97,104-107  A number of explanations have been suggested, including the presence of crippling mutations in the immunoglobulin variable region genes,108  methylation of the immunoglobulin promotors,109  defects in B cell–associated transcription factors,110  and up-regulation of genes that suppress the B-cell gene expression program.111,112  The extinction of the B-cell gene expression program in HRS cells is a complex process, and its full elucidation will likely contribute to our understanding of the pathogenesis of CHL.

In 1991, an international group of pathologists (International Lymphoma Study Group [ILSG]) was formed by Peter Isaacson and Harald Stein to promote better understanding between European and American hematopathologists. At the first meeting held in London, the participants discussed a variety of topics, and found common ground on most issues. Shortly thereafter the group published a consensus report on the definition of MCL, and proposed that the term MCL be used to the exclusion of centrocytic, intermediate, or diffuse small cleaved cell lymphoma, which were overlapping but not precisely defined categories in the Kiel, modified Rappaport, and WF classifications, respectively.40  The ILSG defined MCL by a combination of morphology, immunophenotype, and genetic anomalies, and noted that it had a characteristic clinical profile. A similar report followed at the next meeting in 1992, defining nodular lymphocyte-predominant Hodgkin lymphoma (NLPHL) and elucidating criteria to distinguish it from CHL.51 

At this meeting, the group felt it should try to define as many of the currently recognized entities as possible. At the third meeting, in Berlin in 1993, the group was able to reach consensus on a long list of lymphoid neoplasms, which formed the basis of a new classification of lymphoid neoplasms called the Revised European-American Classification of Lymphoid Neoplasms (REAL classification). The REAL classification, published in Blood in 1994,42  was among the 5 most highly cited papers in clinical medicine for the decade. The REAL classification, and its successor the WHO classification,45  represented a new paradigm in the classification of lymphoid neoplasms. The focus was on the identification of “real” diseases, rather than a global theoretical framework, such as survival (WF) or cellular differentiation (Kiel and Lukes-Collins classifications). The REAL classification was based on the building of consensus, and recognized that a comprehensive classification system was beyond the experience of any one individual. The 19 members of the ILSG contributed their diverse perspectives to achieve a unified point of view. In addition, the ILSG made the decision to base the classification exclusively on published data; in order for an entity to be included in the REAL classification, it had to be validated in more than one publication. A number of entities were listed as provisional, based on more limited published data. Recognizing that Hodgkin lymphoma and plasma cell myeloma are both lymphoid in origin, and that many lymphoid neoplasms can have both solid and circulating phases, the classification included all lymphoid neoplasms: Hodgkin lymphoma, so-called NHLs, lymphoid leukemias, and plasma cell disorders. The neoplasms were stratified according to lineage (B-cell and T/NK-cell), and further into precursor (lymphoblastic) versus mature/peripheral T- and B-cell neoplasms, and Hodgkin lymphoma.

The REAL classification defined distinct entities using a constellation of features: morphology, immunophenotype, genetic features, and clinical presentation and course. The REAL classification appreciated the importance of genetic abnormalities in defining disease entities. With advances in monoclonal antibody technology, some genetic abnormalities could be recognized by immunohistochemistry, such as cyclin D1 expression in the diagnosis of MCL, and ALK expression in ALCL. However, for many lymphoma subtypes, particularly the mature T-cell malignancies, the molecular pathogenesis was not yet known. The inclusion of clinical criteria, which played a major role in the classification of T-cell lymphomas, was one of the most novel aspects of the ILSG approach.113  The REAL classification recognized that the site of presentation is often a signpost for underlying biological distinctions, as in extranodal lymphomas of MALT,114  or primary mediastinal large B-cell lymphoma. The ILSG appreciated that accurate diagnosis cannot take place in a vacuum and requires knowledge of the clinical history since biologically distinct entities may appear cytologically similar.

The REAL classification recognized that because of limitations in current knowledge, some broad categories of disease could not be resolved with existing data, such as diffuse large B-cell lymphomas (DLBCLs) and peripheral T-cell lymphomas (PTCLs), unspecified or not otherwise specified (NOS). Although a number of morphologic variants had been described in both groups, evidence that these delineated distinct biological or clinical entities was lacking. The ILSG identified these categories as ripe for further studies. Indeed, as expected, new technologies have helped delineate separate biological entities within DLBCL and to a lesser extent PTCL-NOS. Gene expression profiling dissected 2 major categories from DLBCL, the germinal center B-cell (GCB) and activated B-cell type (ABC) types of DLBCL, with different patterns of transformation and deregulation.44  The relevance of the microenvironment was appreciated, and the various gene signatures could be correlated with clinical outcome and response to therapy.115,116  Gene expression profiling also validated the distinction of primary mediastinal large B-cell lymphoma from other DLBCL, which was a feature of the REAL classification.117,118 

The REAL classification did not attempt to stratify lymphomas according to a concept of “grade,” either histologic or clinical, as had been done in the Kiel classification and the WF. We now recognize that lymphomas are a heterogeneous group of distinct diseases, most unrelated to one another, not a single disease with a spectrum of histologic grade and clinical behavior. In addition, many lymphomas have within them a spectrum of clinical behavior, which sometimes can be predicted by pathologic features (cell size, proliferation) or clinical features (age, stage, the International Prognostic Index [IPI]). Thus, the ILSG felt that it was neither practical nor helpful to attempt to sort these diverse diseases according to either histologic features or clinical aggressiveness. Both clinicians and pathologists need to learn the histologic and clinical spectrum of each distinct disease. In the REAL classification, grading was used within a disease entity, such as FL, but not across a range of different diseases. Moreover, low cytological grade does not necessarily translate into indolent clinical behavior. Both MCL and angioimmunoblastic T-cell lymphoma had been designated as cytologically low grade, but both diseases are associated with an aggressive clinical course. Conversely, ALCL, which cytologically is high grade, has a better prognosis than most other nodal T-cell lymphomas, with an excellent response to chemotherapy and prolonged disease-free survival.43 

Following the publication of the REAL classification, an international study directed by James Armitage sought to determine whether the REAL classification could be readily applied by a group of independent expert pathologists and had clinical relevance.43  Other goals of the International Lymphoma Classification Project were (1) to determine the role of immunophenotyping and clinical data in the diagnosis of disease entities; (2) to determine both intraobserver and interobserver reproducibility in the diagnosis of the various entities; (3) to further investigate the clinical features and/or epidemiology of the various entities; and (4) to determine whether clinical groupings would be practical or useful for clinical trial or practice. The conclusions of that study affirmed the principles of the REAL classification.43  Virtually all cases could be classified in the published scheme. Intraobserver and interobserver rates of reproducibility were excellent. The use of precise disease definitions, as provided by the REAL scheme, enhanced diagnostic accuracy and reduced subjectivity on the part of pathologists. Immunophenotyping was found to be essential for many diagnostic categories, such as most peripheral T-cell lymphomas. Importantly, the significance of identifying individual disease entities was confirmed by survival data. The International Classification Lymphoma Classification project also elucidated the importance of clinical factors such as the IPI in determining clinical outcome and response to therapy within disease entities, confirming the concept that stratification of a diverse group of diseases by either cytologic or clinical “grade” was unrealistic.119 

Shortly after publication of the REAL classification, the WHO decided to update the classification of hematopoietic and lymphoid neoplasms for its “Blue Book” series of Classifications of Tumors. The task for developing the classification was undertaken as a joint project by the Society for Hematopathology (SH) and the European Association of Hematopathology (EAHP), with a goal to achieve consensus and avoid the political divisions of the past. A steering committee was appointed by the 2 societies, which in turn established 10 individual committees to deal with different disease groups within hematopoietic and lymphoid malignancies.120  The WHO classification was developed over a period of 7 years, during which time the proposal was extensively discussed and vetted at a number of international meetings and symposia to ensure worldwide acceptance. Approximately 100 pathologists and clinicians gathered at a clinical advisory meeting in 1997 at Airlie House, where an agreement had so spectacularly failed in 1975, to discuss a number of questions of clinical relevance. A consensus was achieved on most of the questions raised.121  Importantly, the group concluded that clinical groupings for either protocol treatment or routine clinical practice were generally not feasible. In considering new therapies, each disease must be evaluated individually. Indeed, treatment approaches for one type of lymphoid malignancy are not necessarily applicable to other diseases, even of the same cell lineage. This point is exemplified by the broad spectrum of lymphoid malignancies composed of small B lymphocytes (ie, chronic lymphocytic leukemia (CLL)/small lymphocytic leukemia (SLL), hairy cell leukemia, MALT lymphoma, FL, and MCL). Each of these tumors exhibits deregulation of distinct signaling or transformation pathways, and thus there should be no expectation that the optimal therapy for all small B-cell neoplasms should be the same.

While work on the WHO classification was in progress, Paul Kleihues became Director of the International Agency for Research on Cancer (IARC), an agency of the WHO. Recognizing the growing role of genetics in the definition of cancers and the need for international consensus-based classifications, he initiated a new series of monographs focusing on the pathology and genetics of cancer, published by IARC Press.122  The approach to disease definition and consensus development that was introduced in the REAL classification and adopted in the WHO classification of hematologic and lymphoid neoplasms was a paradigm for this approach. The publication of the WHO classification in 2001 marked the beginning of a new era in which all scientists and clinicians as well as pathologists involved in lymphoma research, diagnosis, and patient care would finally speak the same language.

Each lymphoma included in the WHO classification is a distinct disease entity. Defining a disease is a multistep process, but typically begins not in the basic science laboratory, but at the bedside or at the microscope. This process of disease discovery is exemplified by MALT lymphoma, but the same tale could be told for any number of diseases, such as adult T-cell leukemia/lymphoma,91,123  MCL,3,6,56,124, or ALCL.5,125,126 

In 1963, Frand and Ramot drew attention to the high frequency of a unique variety of intestinal lymphoma in non-Ashkenazi Jews and Arabs in Israel.127  The histology of this condition, subsequently called Mediterranean lymphoma, was characterized as ranging from intense lymphoplasmacytic infiltration of the intestinal mucosa to frank large-cell lymphoma. Seligmann et al observed that patients with Mediterranean lymphoma had circulating alpha heavy chain with absent light chain and introduced the term “alpha chain disease (ACD).128  There were doubts whether ACD represented true lymphoma, particularly since some patients could be cured with antibiotics and, accordingly at a WHO conference in 1976, ACD was renamed yet again as an immunoproliferative small intestinal disease (IPSID). In the late 1970s and early 1980s, based on careful morphologic analysis and demonstration that ACD was monoclonal, Isaacson et al showed that IPSID was indeed a low-grade lymphoma.129 

In the period between 1960 and 1980 the term “pseudolymphoma” came increasingly to be used for low-grade lymphoproliferative lesions arising at a variety of extranodal sites, including the stomach, lung, thyroid, and ocular adnexa. In 1983, Isaacson and Wright showed that gastric pseudolymphoma was a monotypic disorder (ie, a lymphoma) remarkably similar to IPSID.130  The properties of both lymphomas simulated those of MALT, giving rise to the term MALT lymphoma. It soon became clear that a number of other extranodal indolent B-cell lymphomas, many of which had previously been called pseudolymphomas, were examples of MALT lymphoma.131,132  Subsequent studies showed that the morphology and immunophenotype of MALT lymphoma recapitulated those of the Peyer patch, more specifically the Peyer patch marginal zone cells.133 

MALT lymphoma usually presents as localized disease (stage 1E) but characteristically disseminates, either within the same organ or to other extranodal sites where MALT lymphomas are known to arise.134,135  MALT lymphomas typically arise from sites such as the stomach, where there is no organized lymphoid tissue. In seeking an explanation for this paradox, Isaacson hypothesized that Helicobacter pylori infection may result in accumulation of MALT from which gastric MALT lymphoma arises.136  Serial studies showed that there is indeed a close association of infection with H pylori and gastric MALT lymphoma, and that the growth of gastric MALT lymphoma cells can be stimulated by H pylori strain–specific T cells.137  Moreover, in 75% of patients, antibiotic eradication of H pylori leads to regression and effective cure of gastric MALT lymphoma.138  There is an interesting parallel with earlier observations of complete response of IPSID to antibiotics, where there is some evidence that Campylobacter jejuni may be the organism involved,139  and other bacterial and viral agents have been implicated in the pathogenesis of other forms of marginal zone lymphoma.140,141 

Numeric chromosomal alterations, especially trisomies 3, 12, and 18, are common in MALT lymphomas. Chromosomal translocations associated with MALT lymphomas include t(11:18)(q21:q21), resulting in the production of a chimeric protein (API2-MALT1);142  and t(1;14)(p22;q32), t(14;18)(q32;q21), and t(3;14)(p14.1;q32), resulting respectively in transcriptional deregulation of BCL10, MALT1, and FOXP1.143-145  The frequencies at which the translocations occur vary markedly with the primary site of disease.146  This is unlike most other low-grade lymphomas, where a single translocation is characteristic. There is mounting evidence, however, that these different translocations are linked by the roles of BCL10 and MALT1 in the NFκB molecular pathway in lymphocytes. Thus, perhaps all of the genetic alternations appear to lead to deregulation of a common pathway.

The WHO classification has been updated in the past 2 years, again as a joint effort of the hematopathology societies, with 2 clinical advisory committees and more than 130 authors from 22 countries (Table 3). Published in September 2008, it builds upon the advances of the past and makes some inroads into better defining heterogeneous or ambiguous categories of disease.1  Some changes include the introduction of provisional borderline categories, the recognition of small clonal lymphoid populations, and the identification of diseases characterized by involvement of specific anatomic sites or by other clinical features such as age. Unresolved issues include defining pathologic predictors of prognosis in common diseases such as FL, DLBCL, and peripheral T-cell lymphomas.

Table 3

WHO classification of the mature B-cell, T-cell, and NK-cell neoplasms (2008)

Mature B-cell neoplasms 
    Chronic lymphocytic leukemia/small lymphocytic lymphoma 
    B-cell prolymphocytic leukemia 
    Splenic marginal zone lymphoma 
    Hairy cell leukemia 
    Splenic lymphoma/leukemia, unclassifiable* 
        Splenic diffuse red pulp small B-cell lymphoma* 
        Hairy cell leukemia-variant* 
    Lymphoplasmacytic lymphoma 
    Waldenström macroglobulinemia 
    Heavy chain diseases 
        Alpha heavy chain disease 
        Gamma heavy chain disease 
        Mu heavy chain disease 
    Plasma cell myeloma 
    Solitary plasmacytoma of bone 
    Extraosseous plasmacytoma 
    Extranodal marginal zone lymphoma of mucosa-associated lymphoid tissue 
        (MALT lymphoma) 
    Nodal marginal zone lymphoma 
    Pediatric nodal marginal zone lymphoma* 
    Follicular lymphoma 
    Pediatric follicular lymphoma* 
    Primary cutaneous follicle center lymphoma 
    Mantle cell lymphoma 
    Diffuse large B-cell lymphoma (DLBCL), NOS 
        T-cell/histiocyte rich large B-cell lymphoma 
        Primary DLBCL of the CNS 
        Primary cutaneous DLBCL, leg type 
        EBV+ DLBCL of the elderly* 
    DLBCL associated with chronic inflammation 
    Lymphomatoid granulomatosis 
    Primary mediastinal (thymic) large B-cell lymphoma 
    Intravascular large B-cell lymphoma 
    ALK+ large B-cell lymphoma 
    Plasmablastic lymphoma 
    Large B-cell lymphoma arising in HHV8-associated multicentric Castleman disease 
    Primary effusion lymphoma 
    Burkitt lymphoma 
    B-cell lymphoma, unclassifiable, with features intermediate between diffuse large B-cell lymphoma and Burkitt lymphoma 
    B-cell lymphoma, unclassifiable, with features intermediate between diffuse 
        large B-cell lymphoma and classical Hodgkin lymphoma 
Mature T-cell and NK-cell neoplasms 
    T-cell prolymphocytic leukemia 
    T-cell large granular lymphocytic leukemia 
    Chronic lymphoproliferative disorder of NK cells* 
    Aggressive NK cell leukemia 
    Systemic EBV+ T-cell lymphoproliferative disease of childhood 
    Hydroa vacciniforme-like lymphoma 
    Adult T-cell leukemia/lymphoma 
    Extranodal NK/T-cell lymphoma, nasal type 
    Enteropathy-associated T-cell lymphoma 
    Hepatosplenic T-cell lymphoma 
    Subcutaneous panniculitis-like T-cell lymphoma 
    Mycosis fungoides 
    Sézary syndrome 
    Primary cutaneous CD30+ T-cell lymphoproliferative disorders 
        Lymphomatoid papulosis 
        Primary cutaneous anaplastic large cell lymphoma 
    Primary cutaneous gamma-delta T-cell lymphoma 
    Primary cutaneous CD8+ aggressive epidermotropic cytotoxic T-cell lymphoma* 
    Primary cutaneous CD4+ small/medium T-cell lymphoma* 
    Peripheral T-cell lymphoma, NOS 
    Angioimmunoblastic T-cell lymphoma 
    Anaplastic large cell lymphoma, ALK+ 
    Anaplastic large cell lymphoma, ALK* 
Hodgkin lymphoma 
    Nodular lymphocyte-predominant Hodgkin lymphoma 
    Classical Hodgkin lymphoma 
        Nodular sclerosis classical Hodgkin lymphoma 
        Lymphocyte-rich classical Hodgkin lymphoma 
        Mixed cellularity classical Hodgkin lymphoma 
        Lymphocyte-depleted classical Hodgkin lymphoma 
Posttransplantation lymphoproliferative disorders (PTLD) 
    Early lesions 
    Plasmacytic hyperplasia 
    Infectious mononucleosis-like PTLD 
    Polymorphic PTLD 
    Monomorphic PTLD (B- and T/NK-cell types) 
    Classical Hodgkin lymphoma type PTLD 
Mature B-cell neoplasms 
    Chronic lymphocytic leukemia/small lymphocytic lymphoma 
    B-cell prolymphocytic leukemia 
    Splenic marginal zone lymphoma 
    Hairy cell leukemia 
    Splenic lymphoma/leukemia, unclassifiable* 
        Splenic diffuse red pulp small B-cell lymphoma* 
        Hairy cell leukemia-variant* 
    Lymphoplasmacytic lymphoma 
    Waldenström macroglobulinemia 
    Heavy chain diseases 
        Alpha heavy chain disease 
        Gamma heavy chain disease 
        Mu heavy chain disease 
    Plasma cell myeloma 
    Solitary plasmacytoma of bone 
    Extraosseous plasmacytoma 
    Extranodal marginal zone lymphoma of mucosa-associated lymphoid tissue 
        (MALT lymphoma) 
    Nodal marginal zone lymphoma 
    Pediatric nodal marginal zone lymphoma* 
    Follicular lymphoma 
    Pediatric follicular lymphoma* 
    Primary cutaneous follicle center lymphoma 
    Mantle cell lymphoma 
    Diffuse large B-cell lymphoma (DLBCL), NOS 
        T-cell/histiocyte rich large B-cell lymphoma 
        Primary DLBCL of the CNS 
        Primary cutaneous DLBCL, leg type 
        EBV+ DLBCL of the elderly* 
    DLBCL associated with chronic inflammation 
    Lymphomatoid granulomatosis 
    Primary mediastinal (thymic) large B-cell lymphoma 
    Intravascular large B-cell lymphoma 
    ALK+ large B-cell lymphoma 
    Plasmablastic lymphoma 
    Large B-cell lymphoma arising in HHV8-associated multicentric Castleman disease 
    Primary effusion lymphoma 
    Burkitt lymphoma 
    B-cell lymphoma, unclassifiable, with features intermediate between diffuse large B-cell lymphoma and Burkitt lymphoma 
    B-cell lymphoma, unclassifiable, with features intermediate between diffuse 
        large B-cell lymphoma and classical Hodgkin lymphoma 
Mature T-cell and NK-cell neoplasms 
    T-cell prolymphocytic leukemia 
    T-cell large granular lymphocytic leukemia 
    Chronic lymphoproliferative disorder of NK cells* 
    Aggressive NK cell leukemia 
    Systemic EBV+ T-cell lymphoproliferative disease of childhood 
    Hydroa vacciniforme-like lymphoma 
    Adult T-cell leukemia/lymphoma 
    Extranodal NK/T-cell lymphoma, nasal type 
    Enteropathy-associated T-cell lymphoma 
    Hepatosplenic T-cell lymphoma 
    Subcutaneous panniculitis-like T-cell lymphoma 
    Mycosis fungoides 
    Sézary syndrome 
    Primary cutaneous CD30+ T-cell lymphoproliferative disorders 
        Lymphomatoid papulosis 
        Primary cutaneous anaplastic large cell lymphoma 
    Primary cutaneous gamma-delta T-cell lymphoma 
    Primary cutaneous CD8+ aggressive epidermotropic cytotoxic T-cell lymphoma* 
    Primary cutaneous CD4+ small/medium T-cell lymphoma* 
    Peripheral T-cell lymphoma, NOS 
    Angioimmunoblastic T-cell lymphoma 
    Anaplastic large cell lymphoma, ALK+ 
    Anaplastic large cell lymphoma, ALK* 
Hodgkin lymphoma 
    Nodular lymphocyte-predominant Hodgkin lymphoma 
    Classical Hodgkin lymphoma 
        Nodular sclerosis classical Hodgkin lymphoma 
        Lymphocyte-rich classical Hodgkin lymphoma 
        Mixed cellularity classical Hodgkin lymphoma 
        Lymphocyte-depleted classical Hodgkin lymphoma 
Posttransplantation lymphoproliferative disorders (PTLD) 
    Early lesions 
    Plasmacytic hyperplasia 
    Infectious mononucleosis-like PTLD 
    Polymorphic PTLD 
    Monomorphic PTLD (B- and T/NK-cell types) 
    Classical Hodgkin lymphoma type PTLD 
*

Provisional entities for which the WHO Working Group felt there was insufficient evidence to recognize as distinct diseases at this time.

These lesions are classified according to the leukemia or lymphoma to which they correspond.

In the past 20 years there has been a greater appreciation of morphologic and immunophenotypic overlap between CHL and some large B-cell lymphomas—usually primary mediastinal large B-cell lymphoma (PMBL) and mediastinal nodular sclerosis subtype of classical Hodgkin lymphoma (NSCHL).103,146  In most cases, one or the other diagnosis can be made, but there may be a true biological gray zone between these entities, both of which occur in young adults and involve the mediastinum and have similarities in gene expression profiles.117,118  The 2008 WHO classification recognizes a provisional category of B-cell neoplasms with features intermediate between DLBCL and CHL.147,148  These tumors occur predominantly in young men and appear to be more aggressive than either PMBL or NSCHL; in some patients, PMBL may be followed by CHL or vice versa.

A second borderline category relates to the distinction of classical Burkitt lymphoma from diffuse large B-cell lymphomas that also carry a MYC translocation. Many of these occur in adults and morphologically resemble Burkitt lymphoma.149  The 2008 WHO classification includes a provisional category of B-cell lymphoma, unclassifiable, with features intermediate between DLBCL and Burkitt lymphoma. Many lymphomas in this grouping have a translocations of both MYC and BCL2 (“double hit”), and have a very aggressive clinical course.150  While gene expression profiling may show similarities with classical Burkitt lymphoma, other data support segregation of these tumors from Burkitt lymphoma.85,151 

The multistep pathway of tumorigenesis has parallels in most organ systems, best documented in the evolution of colonic adenocarcinoma.152  Histologic progression is a well-recognized feature of many lymphoid neoplasms, but the earliest events in lymphoid neoplasia are difficult to recognize. In fact, the lymphoid system has no recognized “benign neoplasms,” a fact that may be related to the propensity of lymphoid cells or to circulate or home, and not remained confined to a single anatomic site.153  The 2008 WHO classification addresses the problem of clonal expansions of B cells, or less often T cells, that appear to have limited potential for histologic or clinical progression. For example, although up to 70% of healthy adults have circulating clonal memory B cells with the t(14;18)(q32;q21), these cells presumably lack other genetic alterations necessary for development of malignant behavior.154,155  A total of 3% to 5% of healthy adults and more than 10% of those referred for evaluation of lymphocytosis have circulating monoclonal B cells with the immunophenotype of CLL (monoclonal B lymphocytosis); many of these clones also have genetic abnormalities associated with good-prognosis CLL, yet they rarely progress to overt, progressive leukemia.156,157 

Among lymphomas, in situ or early lesions have been recognized for both FL and MCL.158,159  The risk for progression to clinically significant lymphoma is not yet fully known for these focal lesions, which are often incidental findings in an otherwise reactive-appearing lymph node. A related condition is localized FL presenting as small polyps in the duodenum; these duodenal FLs rarely if ever progress to nodal or systemic disease.160,161  Duodenal FL cells express intestinal homing receptors that may retain the clonal B cells within the intestinal mucosa.162  Early gastric MALT lymphomas that are dependent on the presence of H pylori–activated T cells for survival and lack genetic alternations might also be considered benign or “in situ” lymphomas.163  Cases of reactive follicular hyperplasia in children have been reported that contain clonal populations of CD10+ germinal center B cells, and yet do not progress to overt lymphoma.164  Likewise, nodal marginal zone lymphomas in children appear to have a low risk of progression.165  In the T-cell system, primary cutaneous CD30+ T-cell lymphoproliferative disorder, lymphomatoid papulosis, is a clonal T-cell proliferation that also has limited malignant potential.166,167 

Through studying these early events in lymphomagenesis, we are learning more about the circulation and homing of normal and malignant B cells and gaining insight into normal B-cell physiology. One of the great attributes of pathology as a discipline is that disease is a model that provides insight into the functioning of the healthy human organism. As we better understand the abnormal or diseased tissues, we better understand normal human physiology.

The 2008 WHO classification also emphasizes the importance of anatomic site in classification. In addition to MALT lymphomas and primary mediastinal large B-cell lymphoma, both B-cell and T-cell lineages involving cutaneous or other extranodal sites such as the central nervous system are now recognized as distinctive.168  Age may also be a factor in disease definition. Pediatric marginal zone lymphomas and pediatric FLs have been described, which tend to be localized and have an excellent prognosis.165,169,170  They differ in pathogenesis from morphologically similar lymphomas in adults, as pediatric FL typically lacks BCL2 expression and the t(14;18)(q32;q21). In contrast, some diseases appear to occur most often at advanced age, such as EBV+ DLBCL in elderly persons, which likely arise because of decreased immune surveillance.171 

In 2 of the most common lymphomas—FL and DLBCL—there is a need for pathologic predictors of outcome and response to treatment. FL has traditionally been graded according to the proportion of centroblasts—a continuous variable that is associated with decreasing survival duration, but that is poorly reproducible among pathologists. It has become increasingly apparent that cases graded as Grade 1 or Grade 2 have similar outcomes that are not affected by aggressive therapy, and that they do not need to be distinguished from one another. Thus, the 2008 WHO lumps cases with few centroblasts as “FL Grade 1-2 (low grade).” Stratifying FL Grade 3 according to the presence or absence of residual centrocytes (3A vs 3B) was optional in the 2001 WHO classification, but is now mandatory. Some studies have identified biological differences between these 2 subtypes, with most instances of FL Grade 3B more closely related to DLBCL at the molecular level.172,173  However, the segregation is imperfect based on current genetic and clinical studies; this stratification should provide the opportunity to collect more information. Finally, it is emphasized that areas of DLBCL in any FL should receive a separate diagnosis of DLBCL and such patients should be treated as DLBCL (ie, there is no such thing as “FL Grade 3[A or B] with diffuse areas”). After separation of specific new subtypes of DLBCL, we are still left with a large group of DLBCL for which pathologic features are lacking to further stratify them for predicting prognosis or response to therapy. Stratification according to gene expression profiles as GCB versus ABC types has prognostic value, but as yet does not direct therapy. This stratification is imperfectly replicated by morphology, immunophenotyping, or cytogenetic analysis, and thus is not officially incorporated into the classification for practical use. Development of targeted therapies and recognition of additional markers of clinical behavior will likely result in additional modifications to this category in the future.

Finally, an intriguing new development is the recent recognition of lineage plasticity in hematopoietic cells and their neoplasms. In normal B cells, down-regulation of the B-cell regulatory factor PAX-5 can result in reprogramming of both immature and mature B cells to monocytes and even T cells. Cases of lymphoblastic neoplasms of both T- and B-cell type have been reported in which clonally related histiocytic neoplasms have developed, and recently a similar phenomenon has been reported in FL.174,175  This phenomenon may complicate interpretation of the significance of antigen-receptor gene rearrangements in assigning lineage to a neoplasm.

The authors dedicate this review to David Y. Mason, Oxford University, who died on February 2, 2008. David Mason was a pivotal figure in the development of immunohistochemistry, and pioneered the development of monoclonal antibodies reactive against epitopes preserved in formalin-fixed paraffin-embedded tissues. As a leader in the establishment of the Human Leucocyte Differentiation Antigen workshops, he helped bring order to a complex and chaotic field. Through his scientific work and reagents that he generously made available on a worldwide basis he helped shape the modern classification of lymphoid malignancies and was one of the most highly cited physicians in the field of clinical oncology. His wit and satire brightened many medical symposia. Indeed he was a scriptwriter for some of the early pre-Monty Python satirical shows, especially “That Was The Week That Was,” and had a long-standing interest in the arts. His enthusiasm for international collaboration led many to visit his laboratory, and he fostered the development of new generations of translational scientists. He will be greatly missed by his students of all ages.

Contribution: E.J., N.L.H., H.S., and P.I. all contributed to this historical review by participating in writing the manuscript and in providing figures and graphics material.

Conflict-of-interest disclosure: The authors declare no competing financial interests.

Correspondence: Elaine S. Jaffe, Laboratory of Pathology, National Cancer Institute, Building 10, Room 2B 42, 10 Center Drive, MSC 1500, Bethesda, MD 20892; e-mail: elainejaffe@nih.gov.

1
Swerdlow
 
SH
Campo
 
E
Harris
 
NL
, et al. 
WHO Classification of Tumours of Haematopoietic and Lymphoid Tissues
2008
4th ed.
Lyon, France
International Agency for Research on Cancer
2
Kyle
 
RA
Rajkumar
 
SV
Multiple myeloma.
Blood
2008
, vol. 
111
 (pg. 
2962
-
2972
)
3
Swerdlow
 
S
Habeshaw
 
J
Murray
 
L
Dhaliwal
 
H
Lister
 
T
Stansfeld
 
A
Centrocytic lymphoma: a distinct clinicopathologic and immunologic entity.
Am J Pathol
1983
, vol. 
113
 pg. 
181
 
4
Jaffe
 
ES
Bookman
 
MA
Longo
 
DL
Lymphocytic lymphoma of intermediate differentiation–mantle zone lymphoma: a distinct subtype of B-cell lymphoma.
Hum Pathol
1987
, vol. 
18
 (pg. 
877
-
880
)
5
Stein
 
H
Mason
 
DY
Gerdes
 
J
, et al. 
The expression of the Hodgkin's disease associated antigen Ki-1 in reactive and neoplastic lymphoid tissue: evidence that Reed-Sternberg cells and histiocytic malignancies are derived from activated lymphoid cells.
Blood
1985
, vol. 
66
 (pg. 
848
-
858
)
6
Raffeld
 
M
Jaffe
 
ES
bcl-1, t(11;14), and mantle cell derived neoplasms.
Blood
1991
, vol. 
78
 (pg. 
259
-
263
)
7
Dawson
 
PJ
The original illustrations of Hodgkin's disease.
Ann Diagn Pathol
1999
, vol. 
3
 (pg. 
386
-
393
)
8
Trumper
 
LH
Brittinger
 
G
Diehl
 
V
Harris
 
NL
Mauch
 
PM
Armitage
 
JO
Coiffier
 
B
Dalla-Favera
 
R
Harris
 
NL
Non-Hodgkin's lymphoma: a history of classification and clinical observations.
Non-Hodgkin's Lymphomas
2004
Philadelphia, PA
Lippincott, Williams and Wilkins
(pg. 
3
-
19
)
9
Brill
 
NE
Baehr
 
G
Rosenthal
 
N
Generalized giant lymph follicle hyperplasia of lymph nodes and spleen: a hitherto undescribed type.
J Amer Med Assoc
1925
, vol. 
84
 (pg. 
668
-
671
)
10
Symmers
 
D
Follicular lymphadenopathy with splenomegaly: a newly recognized disease of the lymphatic system.
Arch Pathol
1927
, vol. 
3
 (pg. 
816
-
820
)
11
Symmers
 
D
Giant follicular lymphadenopathy with or without splenomegaly: its transformation into polymorphous cell sarcoma of the lymph follicles and its association with Hodgkin's disease, lymphatic leukemia and an apparently unique disease of the lymph nodes and spleen—a disease entity believed heretofore undescribed.
Arch Pathol
1938
, vol. 
26
 (pg. 
603
-
647
)
12
Gall
 
EA
Mallory
 
TB
Malignant lymphoma: a clinico-pathologic survey of 618 cases.
Am J Pathol
1942
, vol. 
18
 (pg. 
381
-
429
)
13
Gall
 
EA
Morrison
 
HR
Scott
 
AT
The follicular type of malignant lymphoma: a survey of 63 cases.
Ann Intern Med
1941
, vol. 
1941
 (pg. 
2073
-
2090
)
14
Burkitt
 
D
A sarcoma involving the jaws in African children.
Br J Surg
1958
, vol. 
46
 (pg. 
218
-
223
)
15
Nowell
 
PC
Phytohemagglutinin: an initiator of mitosis in cultures of normal human leukocytes.
Cancer Res
1960
, vol. 
20
 (pg. 
462
-
466
)
16
O'Conor
 
GT
Definition of Burkitt's tumor.
Int J Cancer
1968
, vol. 
3
 (pg. 
411
-
412
)
17
Epstein
 
MA
Achong
 
BG
Barr
 
YM
Virus particles in cultured lymphoblasts from Burkitt's lymphoma.
Lancet
1964
, vol. 
1
 (pg. 
702
-
703
)
18
Rappaport
 
H
Winter
 
W
Hicks
 
E
Follicular lymphoma: a re-evaluation of its position in the scheme of malignant lymphoma, based on a survey of 253 cases.
Cancer
1956
, vol. 
9
 (pg. 
792
-
821
)
19
Rappaport
 
H
Tumors of the hematopoietic system.
Atlas of Tumor Pathology
1966
Washington, DC
Armed Forces Institute of Pathology
 
Vol. Section III (Series I ed)
20
Lukes
 
R
Butler
 
J
Hicks
 
E
Natural history of Hodgkin's disease as related to its pathological picture.
Cancer
1966
, vol. 
19
 (pg. 
317
-
344
)
21
Stein
 
H
Lennert
 
K
Parwaresch
 
MR
Malignant lymphomas of B-cell type.
Lancet
1972
, vol. 
2
 (pg. 
855
-
857
)
22
Lennert
 
K
Mohri
 
N
Histological classification and occurrence of Hodgkin's disease.
Internist
1974
, vol. 
15
 (pg. 
57
-
65
)
23
Taylor
 
CR
Mason
 
DY
The immunohistological detection of intracellular immunoglobulin in formalin-paraffin sections from multiple myeloma and related conditions using the immunoperoxidase technique.
Clin Exp Immunol
1974
, vol. 
18
 (pg. 
417
-
429
)
24
Jaffe
 
ES
Shevach
 
EM
Frank
 
MM
Berard
 
CW
Green
 
I
Nodular lymphoma: evidence for origin from follicular B lymphocytes.
N Engl J Med
1974
, vol. 
290
 (pg. 
813
-
819
)
25
Non-Hodgkin's Lymphoma Pathologic Classification Project
National Cancer Institute sponsored study of classifications of non-Hodgkin's lymphomas: summary and description of a Working Formulation for clinical usage.
Cancer
1982
, vol. 
49
 (pg. 
2112
-
2135
)
26
Southern
 
EM
Detection of specific sequences among DNA fragments separated by gel electrophoresis.
J Mol Biol
1975
, vol. 
98
 (pg. 
503
-
517
)
27
Zech
 
L
Haglund
 
U
Nilsson
 
K
Klein
 
G
Characteristic chromosomal abnormalities in biopsies and lymphoid-cell lines from patients with Burkitt and non-Burkitt lymphomas.
Int J Cancer
1976
, vol. 
17
 (pg. 
47
-
56
)
28
Fukuhara
 
S
Rowley
 
JD
Variakojis
 
D
Golomb
 
HM
Chromosome abnormalities in poorly differentiated lymphocytic lymphoma.
Cancer Res
1979
, vol. 
39
 (pg. 
3119
-
3128
)
29
McMichael
 
AJ
Pilch
 
JR
Galfre
 
G
Mason
 
DY
Fabre
 
JW
Milstein
 
C
A human thymocyte antigen defined by a hybrid myeloma monoclonal antibody.
Eur J Immunol
1979
, vol. 
9
 (pg. 
205
-
210
)
30
Stein
 
H
Gerdes
 
J
Mason
 
DY
The normal and malignant germinal centre.
Clin Haematol
1982
, vol. 
11
 (pg. 
531
-
559
)
31
Stein
 
H
Bonk
 
A
Tolksdorf
 
G
Lennert
 
K
Rodt
 
H
Gerdes
 
J
Immunohistologic analysis of the organization of normal lymphoid tissue and non-Hodgkin's lymphomas.
J Histochem Cytochem
1980
, vol. 
28
 (pg. 
746
-
760
)
32
Poppema
 
S
Bhan
 
A
Reinherz
 
E
McCluskey
 
R
Schlossman
 
S
Distribution of T cell subsets in human lymph nodes.
J Exp Med
1981
, vol. 
153
 (pg. 
30
-
41
)
33
Poppema
 
S
Bhan
 
A
Reinherz
 
E
Posner
 
M
Schlossman
 
S
In situ immunologic characterization of cellular constituents in lymph nodes and spleens involved by Hodgkin's disease.
Blood
1982
, vol. 
59
 (pg. 
226
-
232
)
34
Bernard
 
A
Boumsell
 
L
The clusters of differentiation (CD) defined by the First International Workshop on Human Leucocyte Differentiation Antigens.
Hum Immunol
1984
, vol. 
11
 (pg. 
1
-
10
)
35
Dalla-Favera
 
R
Bregni
 
M
Erikson
 
J
Patterson
 
D
Gallo
 
RC
Croce
 
CM
Human c-myc onc gene is located on the region of chromosome 8 that is translocated in Burkitt lymphoma cells.
Proc Natl Acad Sci U S A
1982
, vol. 
79
 (pg. 
7824
-
7827
)
36
Taub
 
R
Kirsch
 
I
Morton
 
C
, et al. 
Translocation of the c-myc gene into the immunoglobulin heavy chain locus in human Burkitt lymphoma and murine plasmacytoma cells.
Proc Natl Acad Sci U S A
1982
, vol. 
79
 (pg. 
7837
-
7841
)
37
Yunis
 
JJ
Oken
 
MM
Kaplan
 
ME
Ensrud
 
KM
Howe
 
RR
Theologides
 
A
Distinctive chromosomal abnormalities in histologic subtypes of non-Hodgkin's lymphoma.
N Engl J Med
1982
, vol. 
307
 (pg. 
1231
-
1236
)
38
Saiki
 
RK
Scharf
 
S
Faloona
 
F
, et al. 
Enzymatic amplification of beta-globin genomic sequences and restriction site analysis for diagnosis of sickle cell anemia.
Science
1985
, vol. 
230
 (pg. 
1350
-
1354
)
39
Cremer
 
T
Landegent
 
J
Bruckner
 
A
, et al. 
Detection of chromosome aberrations in the human interphase nucleus by visualization of specific target DNAs with radioactive and non-radioactive in situ hybridization techniques: diagnosis of trisomy 18 with probe L1.84.
Hum Genet
1986
, vol. 
74
 (pg. 
346
-
352
)
40
Banks
 
P
Chan
 
J
Cleary
 
M
, et al. 
Mantle cell lymphoma: a proposal for unification of morphologic, immunologic, and molecular data.
Am J Surg Pathol
1992
, vol. 
16
 (pg. 
637
-
640
)
41
Kuppers
 
R
Rajewsky
 
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.
Proc Natl Acad Sci U S A
1994
, vol. 
91
 (pg. 
10962
-
10966
)
42
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
, vol. 
84
 (pg. 
1361
-
1392
)
43
A clinical evaluation of the International Lymphoma Study Group classification of non-Hodgkin's lymphoma
The Non-Hodgkin's Lymphoma Classification Project.
Blood
1997
, vol. 
89
 (pg. 
3909
-
3918
)
44
Alizadeh
 
AA
Eisen
 
MB
Davis
 
RE
, et al. 
Distinct types of diffuse large B-cell lymphoma identified by gene expression profiling.
Nature
2000
, vol. 
403
 (pg. 
503
-
511
)
45
Jaffe
 
ES
Harris
 
NL
Stein
 
H
Vardiman
 
J
Pathology and Genetics of Tumours of Haematopoietic and Lymphoid Tissues.
2001
Lyon, France
IARC Press
46
Hodgkin
 
T
On some morbid experiences of the absorbent glands and spleen.
Medico-Chirurgical Trans
1832
, vol. 
17
 (pg. 
68
-
97
)
47
Dawson
 
PJ
Whatever happened to Dorothy Reed?
Ann Diagn Pathol
2003
, vol. 
7
 (pg. 
195
-
203
)
48
Jackson
 
H
Parker
 
F
Hodgkin's disease, II: pathology.
N Engl J Med
1944
, vol. 
231
 (pg. 
35
-
44
)
49
Lukes
 
RJ
Craver
 
LF
Hall
 
TC
Rappaport
 
H
Rubin
 
P
Report of the nomenclature committee.
Cancer Res
1966
pg. 
26
 
50
Kant
 
JA
Hubbard
 
SM
Longo
 
DL
Simon
 
RM
DeVita
 
VT
Jaffe
 
ES
The pathologic and clinical heterogeneity of lymphocyte-depleted Hodgkin's disease.
J Clin Oncol
1986
, vol. 
4
 (pg. 
284
-
294
)
51
Mason
 
D
Banks
 
P
Chan
 
J
, et al. 
Nodular lymphocyte predominance Hodgkin's disease: a distinct clinico-pathological entity.
Am J Surg Pathol
1994
, vol. 
18
 (pg. 
528
-
530
)
52
Harris
 
NL
Hodgkin's lymphomas: classification, diagnosis, and grading.
Semin Hematol
1999
, vol. 
36
 (pg. 
220
-
232
)
53
Papermaster
 
BW
Good
 
RA
Relative contributions of the thymus and the bursa of Fabricius to the maturation of the lymphoreticur system and immunological potential in the chicken.
Nature
1962
, vol. 
196
 (pg. 
838
-
840
)
54
Cooper
 
MD
Peterson
 
RD
Good
 
RA
Delineation of the thymic and bursal lymphoid systems in the chicken.
Nature
1965
, vol. 
205
 (pg. 
143
-
146
)
55
Shevach
 
EM
Jaffe
 
ES
Green
 
I
Receptors for complement and immunoglobulin on human and animal lymphoid cells.
Transplant Rev
1973
, vol. 
16
 (pg. 
3
-
28
)
56
Jaffe
 
ES
Braylan
 
RC
Nanba
 
K
Frank
 
MM
Berard
 
CW
Functional markers: a new perspective on malignant lymphomas.
Cancer Treat Rep
1977
, vol. 
61
 (pg. 
953
-
962
)
57
Jaffe
 
ES
Braylan
 
RC
Frank
 
MM
Green
 
I
Berard
 
CW
Heterogeneity of immunologic markers and surface morphology in childhood lymphoblastic lymphoma.
Blood
1976
, vol. 
48
 (pg. 
213
-
222
)
58
Tsukimoto
 
I
Wong
 
KY
Lampkin
 
BC
Surface markers and prognostic factors in acute lymphoblastic leukemia.
N Engl J Med
1976
, vol. 
294
 (pg. 
245
-
248
)
59
Lukes
 
R
Collins
 
R
Immunologic characterization of human malignant lymphomas.
Cancer
1974
, vol. 
34
 (pg. 
1488
-
1503
)
60
Gerard-Marchant
 
R
Hamlin
 
I
Lennert
 
K
Rilke
 
F
Stansfeld
 
A
van Unnik
 
J
Classification of non-Hodgkin's lymphomas.
Lancet
1974
, vol. 
ii
 (pg. 
406
-
408
)
61
Bennett
 
MH
Farrer-Brown
 
G
Henry
 
K
Jeliffe
 
AM
Classification of non-Hodgkin's lymphomas.
Lancet
1974
, vol. 
2
 (pg. 
405
-
406
)
62
Dorfman
 
RF
Classification of non-Hodgkin's lymphomas [letter].
Lancet
1974
, vol. 
2
 (pg. 
961
-
962
)
63
Lennert
 
K
Stein
 
H
Kaiserling
 
E
Cytological and functional criteria for the classification of malignant lymphomata.
Br J Cancer Suppl
1975
, vol. 
2
 (pg. 
29
-
43
)
64
Lennert
 
K
Malignant Lymphomas Other than Hodgkin's Disease.
1978
New York
Springer-Verlag
65
Lennert
 
K
Feller
 
A
Histopathology of Non-Hodgkin's Lymphomas
1992
2nd ed.
New York
Springer-Verlag
66
Dorfman
 
RF
Kim
 
H
Relationship of histology to site in the non-Hodgkin's lymphomata: a study based on surgical staging procedures.
Br J Cancer Suppl
1975
, vol. 
2
 (pg. 
217
-
220
)
67
Chabner
 
BA
Johnson
 
RE
Young
 
RC
, et al. 
Sequential nonsurgical and surgical staging of non-Hodgkin's lymphoma.
Cancer
1978
, vol. 
42
 (pg. 
922
-
925
)
68
Van Unnik
 
JA
Breur
 
K
Burgers
 
JM
, et al. 
Non-Hodgkin's lymphomata: clinical features in relation to histology.
Br J Cancer Suppl
1975
, vol. 
2
 (pg. 
201
-
207
)
69
Rosenberg
 
SA
Kaplan
 
HS
Clinical trials in the non-Hodgkin's lymphomata at Stanford University experimental design and preliminary results.
Br J Cancer Suppl
1975
, vol. 
2
 (pg. 
456
-
464
)
70
NCI non-Hodgkin's Classification Project Writing Committee
Classification of non-Hodgkin's lymphomas: reproducibility of major classification systems.
Cancer
1985
, vol. 
55
 (pg. 
91
-
95
)
71
Strauchen
 
JA
Young
 
RC
DeVita
 
VT
Anderson
 
T
Fantone
 
JC
Berard
 
CW
Clinical relevance of the histopathological subclassification of diffuse “histiocytic” lymphoma.
N Engl J Med
1978
, vol. 
299
 (pg. 
1382
-
1387
)
72
Kohler
 
G
Milstein
 
C
Continuous cultures of fused cells secreting antibody of predefined specificity.
Nature
1975
, vol. 
256
 (pg. 
495
-
497
)
73
Zola
 
H
Swart
 
B
Banham
 
A
, et al. 
CD molecules 2006: human cell differentiation molecules.
J Immunol Methods
2007
, vol. 
319
 (pg. 
1
-
5
)
74
Mason
 
DY
Cordell
 
JL
Abdulaziz
 
Z
Naiem
 
M
Bordenave
 
G
Preparation of peroxidase: antiperoxidase (PAP) complexes for immunohistological labeling of monoclonal antibodies.
J Histochem Cytochem
1982
, vol. 
30
 (pg. 
1114
-
1122
)
75
Nowell
 
PC
Hungerford
 
DA
Chromosome studies in human leukemia, II: chronic granulocytic leukemia.
J Natl Cancer Inst
1961
, vol. 
27
 (pg. 
1013
-
1035
)
76
Tsujimoto
 
Y
Cossman
 
J
Jaffe
 
E
Croce
 
CM
Involvement of the bcl-2 gene in human follicular lymphoma.
Science
1985
, vol. 
228
 (pg. 
1440
-
1443
)
77
Tsujimoto
 
Y
Yunis
 
J
Onorato-Showe
 
L
Erikson
 
J
Nowell
 
P
Croce
 
CM
Molecular cloning of the chromosomal breakpoint of B-cell lymphomas and leukemias with the t(11;14) chromosome translation.
Science
1984
, vol. 
224
 (pg. 
1403
-
1406
)
78
Arnold
 
A
Kim
 
HG
Gaz
 
RD
, et al. 
Molecular cloning and chromosomal mapping of DNA rearranged with the parathyroid hormone gene in a parathyroid adenoma.
J Clin Invest
1989
, vol. 
83
 (pg. 
2034
-
2040
)
79
Arnold
 
A
Cossman
 
J
Bakhshi
 
A
Jaffe
 
ES
Waldmann
 
TA
Korsmeyer
 
SJ
Immunoglobulin-gene rearrangements as unique clonal markers in human lymphoid neoplasms.
N Engl J Med
1983
, vol. 
309
 (pg. 
1593
-
1599
)
80
Kuppers
 
R
Zhao
 
M
Hansmann
 
ML
Rajewsky
 
K
Tracing B cell development in human germinal centres by molecular analysis of single cells picked from histological sections.
EMBO J
1993
, vol. 
12
 (pg. 
4955
-
4967
)
81
Kuppers
 
R
Klein
 
U
Hansmann
 
ML
Rajewsky
 
K
Cellular origin of human B-cell lymphomas.
N Engl J Med
1999
, vol. 
341
 (pg. 
1520
-
1529
)
82
Tamaru
 
J
Hummel
 
M
Zemlin
 
M
Kalvelage
 
B
Stein
 
H
Hodgkin's disease with a B-cell phenotype often shows a VDJ rearrangement and somatic mutations in the VH genes.
Blood
1994
, vol. 
84
 (pg. 
708
-
715
)
83
Deane
 
M
Norton
 
JD
Detection of immunoglobulin gene rearrangement in B lymphoid malignancies by polymerase chain reaction gene amplification.
Br J Haematol
1990
, vol. 
74
 (pg. 
251
-
256
)
84
Ngan
 
BY
Nourse
 
J
Cleary
 
ML
Detection of chromosomal translocation t(14;18) within the minor cluster region of bcl-2 by polymerase chain reaction and direct genomic sequencing of the enzymatically amplified DNA in follicular lymphomas.
Blood
1989
, vol. 
73
 (pg. 
1759
-
1762
)
85
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
, vol. 
354
 (pg. 
2419
-
2430
)
86
Wlodarska
 
I
Mecucci
 
C
Stul
 
M
, et al. 
Fluorescence in situ hybridization identifies new chromosomal changes involving 3q27 in non-Hodgkin's lymphomas with BCL6/LAZ3 rearrangement.
Genes Chromosomes Cancer
1995
, vol. 
14
 (pg. 
1
-
7
)
87
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
, vol. 
95
 (pg. 
1443
-
1450
)
88
Jaffe
 
ES
Chan
 
JKC
Su
 
IJ
, et al. 
Report of the workshop on nasal and related extranodal angiocentric T/NK cell lymphomas: definitions, differential diagnosis, and epidemiology.
Am J Surg Pathol
1996
, vol. 
20
 (pg. 
103
-
111
)
89
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
, vol. 
89
 (pg. 
1413
-
1420
)
90
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
, vol. 
96
 (pg. 
443
-
451
)
91
Poiesz
 
B
Ruscetti
 
F
Gazdar
 
A
Detection and isolation of type C retrovirus particles from fresh and cultured lymphocytes of a patient with cutaneous T-cell lymphoma.
Proc Natl Acad Sci U S A
1980
, vol. 
77
 (pg. 
7415
-
7419
)
92
Cesarman
 
E
Chang
 
Y
Moore
 
PS
Said
 
JW
Knowles
 
DM
Kaposi's sarcoma-associated herpesvirus-like DNA sequences in AIDS-related body-cavity-based lymphomas.
N Engl J Med
1995
, vol. 
332
 (pg. 
1186
-
1191
)
93
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
, vol. 
99
 (pg. 
2331
-
2336
)
94
Dupin
 
N
Fisher
 
C
Kellam
 
P
, et al. 
Distribution of human herpesvirus-8 latently infected cells in Kaposi's sarcoma, multicentric Castleman's disease, and primary effusion lymphoma.
Proc Natl Acad Sci U S A
1999
, vol. 
96
 (pg. 
4546
-
4551
)
95
Du
 
MQ
Diss
 
TC
Liu
 
H
, et al. 
KSHV- and EBV-associated germinotropic lymphoproliferative disorder.
Blood
2002
, vol. 
100
 (pg. 
3415
-
3418
)
96
Diehl
 
V
Kirchner
 
HH
Schaadt
 
M
, et al. 
Hodgkin's disease: establishment and characterization of four in vitro cell lies.
J Cancer Res Clin Oncol
1981
, vol. 
101
 (pg. 
111
-
124
)
97
Stein
 
H
Gerdes
 
J
Schwab
 
U
, et al. 
Identification of Hodgkin and Sternberg-reed cells as a unique cell type derived from a newly-detected small-cell population.
Int J Cancer
1982
, vol. 
30
 (pg. 
445
-
459
)
98
Andreesen
 
R
Osterholz
 
J
Lohr
 
GW
Bross
 
KJ
A Hodgkin cell-specific antigen is expressed on a subset of auto- and alloactivated T (helper) lymphoblasts.
Blood
1984
, vol. 
63
 (pg. 
1299
-
1302
)
99
Pinkus
 
G
Said
 
J
Hodgkin's disease, lymphocyte predominance type, nodular: a distinct entity? Unique staining profile of L&H variants of Reed-Sternberg cells defined by monoclonal antibodies to leukocyte common antigen, granulocyte specific antigen, and B-cell specific antigen.
Am J Pathol
1985
, vol. 
116
 (pg. 
1
-
6
)
100
Schmid
 
C
Pan
 
L
Diss
 
T
Isaacson
 
PG
Expression of B-cell antigens by Hodgkin's and Reed-Sternberg cells.
Am J Pathol
1991
, vol. 
139
 (pg. 
701
-
707
)
101
Weiss
 
LM
Movahed
 
LA
Warnke
 
RA
Sklar
 
J
Detection of Epstein-Barr viral genomes in Reed-Sternberg cells of Hodgkin's disease.
N Engl J Med
1989
, vol. 
320
 (pg. 
502
-
506
)
102
Jaffe
 
ES
The elusive Reed-Sternberg cell.
N Engl J Med
1989
, vol. 
320
 (pg. 
529
-
531
)
103
Jaffe
 
ES
Zarate
 
OA
Medeiros
 
LJ
The interrelationship of Hodgkin's disease and non-Hodgkin's lymphomas: lessons learned from composite and sequential malignancies.
Semin Diagn Pathol
1992
, vol. 
9
 (pg. 
297
-
303
)
104
Hsu
 
SM
Yang
 
K
Jaffe
 
ES
Phenotypic expression of Hodgkin's and Reed-Sternberg cells in Hodgkin's disease.
Am J Pathol
1985
, vol. 
118
 (pg. 
209
-
217
)
105
Falini
 
B
Stein
 
H
Pileri
 
S
, et al. 
Expression of lymphoid-associated antigens on Hodgkin's and Reed-Sternberg cells of Hodgkin's disease: an immunocytochemical study on lymph node cytospins using monoclonal antibodies.
Histopathology
1987
, vol. 
11
 (pg. 
1229
-
1242
)
106
Dallenbach
 
FE
Stein
 
H
Expression of T-cell-receptor beta chain in Reed-Sternberg cells.
Lancet
1989
, vol. 
2
 (pg. 
828
-
830
)
107
Tzankov
 
A
Bourgau
 
C
Kaiser
 
A
, et al. 
Rare expression of T-cell markers in classical Hodgkin's lymphoma.
Mod Pathol
2005
, vol. 
18
 (pg. 
1542
-
1549
)
108
Kanzler
 
H
Kuppers
 
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.
J Exp Med
1996
, vol. 
184
 (pg. 
1495
-
1505
)
109
Ushmorov
 
A
Ritz
 
O
Hummel
 
M
, et al. 
Epigenetic silencing of the immunoglobulin heavy-chain gene in classical Hodgkin lymphoma-derived cell lines contributes to the loss of immunoglobulin expression.
Blood
2004
, vol. 
104
 (pg. 
3326
-
3334
)
110
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
, vol. 
97
 (pg. 
496
-
501
)
111
Ehlers
 
A
Oker
 
E
Bentink
 
S
Lenze
 
D
Stein
 
H
Hummel
 
M
Histone acetylation and DNA demethylation of B cells result in a Hodgkin-like phenotype.
Leukemia
2008
, vol. 
22
 (pg. 
835
-
841
)
112
Mathas
 
S
Janz
 
M
Hummel
 
F
, et al. 
Intrinsic inhibition of transcription factor E2A by HLH proteins ABF-1 and Id2 mediates reprogramming of neoplastic B cells in Hodgkin lymphoma.
Nat Immunol
2006
, vol. 
7
 (pg. 
207
-
215
)
113
Jaffe
 
ES
Pathobiology of peripheral T-cell lymphomas.
Hematology Am Soc Hematol Educ Program
2006
(pg. 
317
-
322
)
114
Isaacson
 
P
Spencer
 
J
Malignant lymphoma of mucosa-associated lymphoid tissue.
Histopathol
1987
, vol. 
11
 (pg. 
445
-
462
)
115
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
, vol. 
346
 (pg. 
1937
-
1947
)
116
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
, vol. 
8
 (pg. 
68
-
74
)
117
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
, vol. 
102
 (pg. 
3871
-
3879
)
118
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
, vol. 
198
 (pg. 
851
-
862
)
119
The International Non-Hodgkin's Lymphoma Prognostic Factors Project
A predictive model for aggressive non-Hodgkin's lymphoma.
N Engl J Med
1993
, vol. 
329
 (pg. 
987
-
994
)
120
Jaffe
 
ES
Harris
 
NL
Diebold
 
J
Muller-Hermelink
 
HK
World Health Organization classification of neoplastic diseases of the hematopoietic and lymphoid tissues: A progress report.
Am J Clin Pathol
1999
, vol. 
111
 (pg. 
S8
-
S12
)
121
Harris
 
NL
Jaffe
 
ES
Diebold
 
J
, et al. 
World Health Organization classification of neoplastic diseases of the hematopoietic and lymphoid tissues: report of the Clinical Advisory Committee meeting—Airlie House, Virginia, November 1997.
J Clin Oncol
1999
, vol. 
17
 (pg. 
3835
-
3849
)
122
Kleihues
 
P
Cavenee
 
WK
Pathology and Genetics: Tumours of the Nervous System
2000
Lyon, France
IARC Press
123
Takatsuki
 
K
Adult T-cell leukemia.
Intern Med
1995
, vol. 
34
 (pg. 
947
-
952
)
124
Yang
 
WI
Zukerberg
 
LR
Motokura
 
T
Arnold
 
A
Harris
 
NL
Cyclin D1 (Bcl-1, PRAD1) protein expression in low-grade B-cell lymphomas and reactive hyperplasia.
Am J Pathol
1994
, vol. 
145
 (pg. 
86
-
96
)
125
Stein
 
H
Foss
 
HD
Durkop
 
H
, et al. 
CD30(+) anaplastic large cell lymphoma: a review of its histopathologic, genetic, and clinical features.
Blood
2000
, vol. 
96
 (pg. 
3681
-
3695
)
126
Jaffe
 
ES
Anaplastic large cell lymphoma: the shifting sands of diagnostic hematopathology.
Mod Pathol
2001
, vol. 
14
 (pg. 
219
-
228
)
127
Frand
 
UI
Ramot
 
B
[Malignant lymphomata in Israel: an epidemiological study based on 399 cases].
Harefuah
1963
, vol. 
65
 (pg. 
83
-
86
)
128
Seligmann
 
M
Danon
 
F
Hurez
 
D
Mihaesco
 
E
Preud'homme
 
JL
Alpha-chain disease: a new immunoglobulin abnormality.
Science
1968
, vol. 
162
 (pg. 
1396
-
1397
)
129
Isaacson
 
P
Middle East lymphoma and alpha-chain disease: an immunohistochemical study.
Am J Surg Pathol
1979
, vol. 
3
 (pg. 
431
-
441
)
130
Isaacson
 
P
Wright
 
D
Malignant lymphoma of mucosa associated lymphoid tissue: a distinctive B cell lymphoma.
Cancer
1983
, vol. 
52
 (pg. 
1410
-
1416
)
131
Harris
 
NL
Pilch
 
BZ
Bhan
 
AK
Harmon
 
DC
Goodman
 
ML
Immunohistologic diagnosis of orbital lymphoid infiltrates.
Am J Surg Pathol
1984
, vol. 
8
 (pg. 
83
-
91
)
132
Isaacson
 
PG
Gastric MALT lymphoma: from concept to cure.
Ann Oncol
1999
, vol. 
10
 (pg. 
637
-
645
)
133
Spencer
 
J
Finn
 
T
Pulford
 
K
Mason
 
D
Isaacson
 
P
The human gut contains a novel population of B lymphocytes which resemble marginal zone cells.
Clin Exp Immunol
1985
, vol. 
62
 (pg. 
607
-
612
)
134
Wotherspoon
 
AC
Diss
 
TC
Pan
 
LX
, et al. 
Primary low-grade B-cell lymphoma of the conjunctiva: a mucosa-associated lymphoid tissue type lymphoma Epstein-Barr virus (EBV) in enteropathy-associated T-cell lymphoma (EATL).
Histopathology
1993
, vol. 
23
 (pg. 
417
-
424
)
135
Isaacson
 
PG
Mucosa-associated lymphoid tissue lymphoma.
Semin Hematol
1999
, vol. 
36
 (pg. 
139
-
147
)
136
Wotherspoon
 
AC
Ortiz-Hidalgo
 
C
Falzon
 
MR
Isaacson
 
PG
Helicobacter pylori-associated gastritis and primary B-cell gastric lymphoma.
Lancet
1991
, vol. 
338
 (pg. 
1175
-
1176
)
137
Hussell
 
T
Isaacson
 
PG
Crabtree
 
JE
Spencer
 
J
The response of cells from low-grade B-cell gastric lymphomas of mucosa-associated lymphoid tissue to Helicobacter pylori.
Lancet
1993
, vol. 
342
 (pg. 
571
-
574
)
138
Wotherspoon
 
A
Doglioni
 
C
Diss
 
T
, et al. 
Regression of primary low-grade B-cell gastric lymphoma of mucosa-associated lymphoid tissue type after eradication of Helicobacter pylori.
Lancet
1993
, vol. 
342
 (pg. 
575
-
577
)
139
Lecuit
 
M
Abachin
 
E
Martin
 
A
, et al. 
Immunoproliferative small intestinal disease associated with Campylobacter jejuni.
N Engl J Med
2004
, vol. 
350
 (pg. 
239
-
248
)
140
Roulland
 
S
Suarez
 
F
Hermine
 
O
Nadel
 
B
Pathophysiological aspects of memory B-cell development.
Trends Immunol
2008
, vol. 
29
 (pg. 
25
-
33
)
141
Rowe
 
M
Peng-Pilon
 
M
Huen
 
DS
, et al. 
Upregulation of bcl-2 by the Epstein-Barr virus latent membrane protein LMP1: a B-cell-specific response that is delayed relative to NF-kappa B activation and to induction of cell surface markers.
J Virol
1994
, vol. 
68
 (pg. 
5602
-
5612
)
142
Dierlamm
 
J
Baens
 
M
Wlodarska
 
I
, et al. 
The apoptosis inhibitor gene API2 and a novel 18q gene, MLT, are recurrently rearranged in the t(11;18)(q21;q21) associated with mucosa-associated lymphoid tissue lymphomas.
Blood
1999
, vol. 
93
 (pg. 
3601
-
3609
)
143
Willis
 
TG
Jadayel
 
DM
Du
 
MQ
, et al. 
Bcl10 is involved in t(1;14)(p22;q32) of MALT B cell lymphoma and mutated in multiple tumor types.
Cell
1999
, vol. 
96
 (pg. 
35
-
45
)
144
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
, vol. 
101
 (pg. 
2335
-
2339
)
145
Streubel
 
B
Simonitsch-Klupp
 
I
Mullauer
 
L
, et al. 
Variable frequencies of MALT lymphoma-associated genetic aberrations in MALT lymphomas of different sites.
Leukemia
2004
, vol. 
18
 (pg. 
1722
-
1726
)
146
Jaffe
 
ES
Wilson
 
WH
Mauch
 
PM
Armitage
 
JO
Coiffier
 
B
Dalla-Favera
 
R
Harris
 
NL
Gray zone, synchronous, and metachronous lymphomas: Diseases at the interface of non-Hodgkin's lymphomas and Hodgkin's Lymphoma.
Non-Hodgkin's Lymphoma
2004
Philadelphia, PA
Lippincott, Williams, and Wilkins
(pg. 
69
-
80
)
147
Rudiger
 
T
Jaffe
 
ES
Delsol
 
G
, et al. 
Workshop report on Hodgkin's disease and related diseases (“grey zone” lymphoma).
Ann Oncol
1998
, vol. 
9
 (pg. 
S31
-
S38
)
148
Traverse-Glehen
 
A
Pittaluga
 
S
Gaulard
 
P
, et al. 
Mediastinal gray zone lymphoma: the missing link between classic Hodgkin's lymphoma and mediastinal large B-cell lymphoma.
Am J Surg Pathol
2005
, vol. 
29
 (pg. 
1411
-
1421
)
149
Haralambieva
 
E
Boerma
 
EJ
van Imhoff
 
GW
, et al. 
Clinical, immunophenotypic, and genetic ana-lysis of adult lymphomas with morphologic features of Burkitt lymphoma.
Am J Surg Pathol
2005
, vol. 
29
 (pg. 
1086
-
1094
)
150
Karsan
 
A
Gascoyne
 
RD
Coupland
 
RW
Shepherd
 
JD
Phillips
 
GL
Horsman
 
DE
Combination of t(14;18) and a Burkitt's type translocation in B-cell malignancies.
Leuk Lymphoma
1993
, vol. 
10
 (pg. 
433
-
441
)
151
Dave
 
SS
Fu
 
K
Wright
 
GW
, et al. 
Molecular diagnosis of Burkitt's lymphoma.
N Engl J Med
2006
, vol. 
354
 (pg. 
2431
-
2442
)
152
Smith
 
D
Ballal
 
M
Hodder
 
R
Selvachandran
 
SN
Cade
 
D
The adenoma carcinoma sequence: an indoctrinated model for tumorigenesis, but is it always a clinical reality?
Colorectal Dis
2006
, vol. 
8
 (pg. 
296
-
301
)
153
Jaffe
 
ES
Follicular lymphomas: possibility that they are benign tumors of the lymphoid system.
J Natl Cancer Inst
1983
, vol. 
70
 (pg. 
401
-
403
)
154
Limpens
 
J
de Jong
 
D
van Krieken
 
J
, et al. 
Bcl-2 in benign lymphoid tissue with follicular hyperplasia.
Oncogene
1991
, vol. 
6
 (pg. 
2271
-
2276
)
155
Roulland
 
S
Navarro
 
JM
Grenot
 
P
, et al. 
Follicular lymphoma-like B cells in healthy individuals: a novel intermediate step in early lymphomagenesis.
J Exp Med
2006
, vol. 
203
 (pg. 
2425
-
2431
)
156
Marti
 
GE
Rawstron
 
AC
Ghia
 
P
, et al. 
Diagnostic criteria for monoclonal B-cell lymphocytosis.
Br J Haematol
2005
, vol. 
130
 (pg. 
325
-
332
)
157
Rawstron
 
AC
Bennett
 
FL
O'Connor
 
SJ
, et al. 
Monoclonal B-cell lymphocytosis and chronic lymphocytic leukemia.
N Engl J Med
2008
, vol. 
359
 (pg. 
575
-
583
)
158
Cong
 
P
Raffeld
 
M
Teruya-Feldstein
 
J
Sorbara
 
L
Pittaluga
 
S
Jaffe
 
ES
In situ localization of follicular lymphoma: description and analysis by laser capture microdissection.
Blood
2002
, vol. 
99
 (pg. 
3376
-
3382
)
159
Aqel
 
N
Barker
 
F
Patel
 
K
Naresh
 
KN
In-situ mantle cell lymphoma: a report of two cases.
Histopathology
2008
, vol. 
52
 (pg. 
256
-
260
)
160
Yoshino
 
T
Miyake
 
K
Ichimura
 
K
, et al. 
Increased incidence of follicular lymphoma in the duodenum.
Am J Surg Pathol
2000
, vol. 
24
 (pg. 
688
-
693
)
161
Poggi
 
MM
Cong
 
PJ
Coleman
 
CN
Jaffe
 
ES
Low-grade follicular lymphoma of the small intestine.
J Clin Gastroenterol
2002
, vol. 
34
 (pg. 
155
-
159
)
162
Bende
 
RJ
Smit
 
LA
Bossenbroek
 
JG
, et al. 
Primary follicular lymphoma of the small intestine: alpha4beta7 expression and immunoglobulin configuration suggest an origin from local antigen-experienced B cells.
Am J Pathol
2003
, vol. 
162
 (pg. 
105
-
113
)
163
Liu
 
H
Ruskon-Fourmestraux
 
A
Lavergne-Slove
 
A
, et al. 
Resistance of t(11;18) positive gastric mucosa-associated lymphoid tissue lymphoma to Helicobacter pylori eradication therapy.
Lancet
2001
, vol. 
357
 (pg. 
39
-
40
)
164
Kussick
 
SJ
Kalnoski
 
M
Braziel
 
RM
Wood
 
BL
Prominent clonal B-cell populations identified by flow cytometry in histologically reactive lymphoid proliferations.
Am J Clin Pathol
2004
, vol. 
121
 (pg. 
464
-
472
)
165
Taddesse-Heath
 
L
Pittaluga
 
S
Sorbara
 
L
Bussey
 
M
Raffeld
 
M
Jaffe
 
ES
Marginal zone B-cell lymphoma in children and young adults.
Am J Surg Pathol
2003
, vol. 
27
 (pg. 
522
-
531
)
166
Weiss
 
LM
Wood
 
GS
Trela
 
M
Warnke
 
RA
Sklar
 
J
Clonal T-cell populations in lymphomatoid papulosis: evidence of a lymphoproliferative origin for a clinically benign disease.
N Engl J Med
1986
, vol. 
315
 (pg. 
475
-
479
)
167
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
, vol. 
28
 (pg. 
973
-
980
)
168
Willemze
 
R
Jaffe
 
ES
Burg
 
G
, et al. 
WHO-EORTC classification for cutaneous lymphomas.
Blood
2005
, vol. 
105
 (pg. 
3768
-
3785
)
169
Lorsbach
 
RB
Shay-Seymore
 
D
Moore
 
J
, et al. 
Clinicopathologic analysis of follicular lymphoma occurring in children.
Blood
2002
, vol. 
99
 (pg. 
1959
-
1964
)
170
Finn
 
LS
Viswanatha
 
DS
Belasco
 
JB
, et al. 
Primary follicular lymphoma of the testis in childhood.
Cancer
1999
, vol. 
85
 (pg. 
1626
-
1635
)
171
Oyama
 
T
Ichimura
 
K
Suzuki
 
R
, et al. 
Senile EBV+ B-cell lymphoproliferative disorders: a clinicopathologic study of 22 patients.
Am J Surg Pathol
2003
, vol. 
27
 (pg. 
16
-
26
)
172
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
, vol. 
99
 (pg. 
3806
-
3812
)
173
Bosga-Bouwer
 
AG
van 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
, vol. 
101
 (pg. 
1149
-
1154
)
174
Feldman
 
AL
Arber
 
DA
Pittaluga
 
S
, et al. 
Clonally related follicular lymphomas and histiocytic/dendritic cell sarcomas: evidence for transdifferentiation of the follicular lymphoma clone.
Blood
2008
, vol. 
111
 (pg. 
5433
-
5439
)
175
Feldman
 
AL
Berthold
 
F
Arceci
 
R
, et al. 
Clonal relationship between precursor T-lymphoblastic leukaemia/lymphoma and Langerhans-cell histiocytosis.
Lancet Oncology
2005
, vol. 
6
 (pg. 
435
-
437
)
Sign in via your Institution