Acquired chromosomal anomalies (most commonly translocations) in lymphoma and leukemia usually result in either activation of a quiescent gene (by means of immunoglobulin or T-cell–receptor promotors) and expression of an intact protein product, or creation of a fusion gene encoding a chimeric protein. This review summarizes current immunocytochemical studies of these 2 categories of oncogenic protein, with emphasis on the clinical relevance of their detection in diagnostic samples. Among the quiescent genes activated by rearrangement, expression of cyclin D1 (due to rearrangement of theCCND1 [BCL-1] gene) is a near-specific marker of t(11;14) in mantle cell lymphoma; BCL-2 expression distinguishes follicular lymphoma cells from their nonneoplastic counterparts in reactive germinal centers and appears to be an independent prognostic marker in diffuse large cell lymphoma; andTAL-1 (SCL) expression identifies T-cell acute lymphoblastic neoplasms in which this gene is activated. The protein products of other genes activated by chromosomal rearrangement have a role as markers of either lineage (eg, PAX-5 [B-cell–specific activator protein] for B cells, including B-lymphoblastic neoplasms), or maturation stage (eg, BCL-6 for germinal-center and activated B cells and MUM-1/IRF4 for plasma cells). Currently, no hybrid protein encoded by fusion genes is reliably detectable by antibodies recognizing unique junctional epitopes (ie, epitopes absent from the wild-type constituent proteins). Nevertheless, staining for promyelocytic leukemia (PML) protein will detect acute PML with t(15;17) because the microspeckled nuclear labeling pattern for PML-RARα is highly distinctive. Similarly, antibodies to the anaplastic lymphoma kinase (ALK) tyrosine kinase are valuable (because wild-type ALK is not found in normal lymphoid tissue) in detecting neoplasms (CD30-positive large T-cell lymphomas) with t(2;5) or its variants. Thus, immunocytochemical detection of the products of many rearranged genes in lymphoma and leukemia can be clinically informative and provide information on cellular and subcellular protein expression that cannot be inferred from studies based on messenger RNA.

Translocations, deletions, and other nonrandom chromosomal abnormalities1,2 play a central role in the pathogenesis of many human hematologic malignant diseases. A growing number of studies are revealing the ways in which these chromosomal alterations affect specific genes, such as those encoding nuclear transcription factors. However, any gene implicated in the neoplastic process can act only through the protein it encodes. Many of the consequences of chromosomal changes with respect to abnormal protein expression have been inferred indirectly by analysis of messenger RNA (mRNA) transcription, but it is clear that protein and mRNA levels do not always correlate. For example, germinal-center B cells appear to contain BCL-2 message but little protein,3,4and the same is true for TAL-1 in erythroid cells.5 Many examples of the opposite combination (high protein and little or no message) also exist, such as BCL-2 in mature lymphocytes6,7 and elastase in mature myeloid cells.8 Antibody-based detection of the protein is therefore required, but this frequently cannot be done because of a lack of suitable reagents. Consequently, we are often ignorant not only about patterns of expression of oncogenic proteins in hematologic neoplasms but also about possible abnormalities in the distribution and subcellular localization of these proteins.

Most of the currently recognized genetic alterations in human leukemias and lymphomas result in either activation of a quiescent gene or creation of a hybrid gene encoding a chimeric protein. Table1 summarizes the genetic abnormalities that have been studied using antibodies specific for the protein products of genes involved in such rearrangements. We here review the immunocytochemical studies that have been done with these antibodies and assess, for each protein, whether abnormalities demonstrable by immunocytochemistry are of clinical value in determining diagnosis or predicting prognosis. Immunocytochemical findings that are of value are discussed below, whereas those for which no clear clinical applications have been observed are described briefly in Table2.

The CCND1 (BCL-1) gene and its protein product (cyclin D1)

CCND1 (BCL-1) gene.

The (11;14)(q13;q32) translocation,9 an anomaly often found in mantle cell (centrocytic) lymphoma10,11 and occasionally in other B-cell neoplasms, notably myeloma,12,13 juxtaposes the CCND1(BCL-1) locus encoding cyclin D1 on chromosome 11 to an immunoglobulin-enhancer sequence on chromosome 14.9 TheCCND1 gene is transcriptionally silent in normal lymphohemopoietic tissues14,15; thus, expression of the protein may promote neoplastic cell proliferation by perpetuating the transition from G1 to S.16 

Antibodies to cyclin D1.

Several antibodies recognizing cyclin D1 have been described (Table3),14-18 but hematopathologists know that immunocytochemical detection of cyclin D1 in routinely processed tissues is not always easy.19,20 It is possible that cyclin D1 is so closely associated with other molecules in the nucleus that epitopes are masked. Whatever the cause of the difficulty, reliable staining can usually be obtained by using an optimized technique (eg, by incubating the antibody overnight with sections previously subjected to microwave heating in the presence of EDTA), as described in detail elsewhere.19-21 

Cyclin D1 expression in normal tissues.

Cyclin D1 is not detectable by immunocytochemistry in normal lymphohemopoietic tissues,14 although a variety of stratified squamous epithelia are positive for cyclin D1.14 

Cyclin D1 in lymphoid neoplasia.

Most mantle cell lymphomas express cyclin D1,22-25although the staining intensity and the percentage of positive cells differ from case to case.15 Immunostaining is usually seen in a diffuse pattern in cell nuclei,15,24 but cytoplasmic positivity is also occasionally observed.17 The blastoid form of mantle cell lymphoma,26 characterized by frequent rearrangement of the CCND1 (BCL-1) gene, a high mitotic index, and a poor prognosis,27 shows no difference in the pattern or frequency of cyclin D1 staining.28Expression of cyclin D1 has also been observed in cases of multiple lymphomatous polyposis (Figure 1), the intestinal form of mantle cell lymphoma.11 

Fig. 1.

Cyclin D1 immunostaining of neoplastic cell nuclei in a case of lymphomatoid polyposis, shown at low power (left) and high power (right).

An immunoperoxidase technique in a paraffin section was used. Original magnification left, × 100; right, × 800.

Fig. 1.

Cyclin D1 immunostaining of neoplastic cell nuclei in a case of lymphomatoid polyposis, shown at low power (left) and high power (right).

An immunoperoxidase technique in a paraffin section was used. Original magnification left, × 100; right, × 800.

Close modal

Cyclin D1 expression is not completely specific for mantle cell lymphoma29,30: it has been found in plasmacytoma/myeloma, especially in cases with t(11;14)31,32; in sporadic cases of B-cell chronic lymphocytic leukemia (B-CLL), some of which show t(11;14) and CCND1 rearrangement,33 and in hairy cell leukemia, although levels of cyclin D1 are lower than in mantle cell lymphoma and no CCND1 (BCL-1) gene rearrangements are detected.33,34 Expression of CCND1(BCL-1) mRNA due to t(11;14) has been found in some cases of splenic marginal zone lymphoma,13 but no protein has been detected by immunohistological methods in this disease.35 

Clinical applications of cyclin D1 immunostaining.

Detection of cyclin D1 by immunohistochemistry can be added to other criteria for the diagnosis of mantle cell lymphoma,11,30,36 particularly when a poor biopsy or an unusual growth pattern hinders recognition of the disease. Because mantle cell lymphoma has a range of morphologic features (from a small cell tumor to a blastoid proliferation11,26) and growth patterns (diffuse, nodular, or mantle zone23,37), a near-specific marker is clearly valuable, particularly because survival in this disease is significantly worse than that in other small cell B-cell lymphomas.11,37,38 In addition, cyclin D1 immunostaining may distinguish multiple lymphomatous polyposis from other indolent intestinal lymphomas, and the blastoid variant of mantle cell lymphoma (BCL-1 positive) from B-lymphoblastic lymphoma/leukemia (BCL-1 negative).28 Cyclin D1 immunostaining has also been used to identify cases of mantle cell lymphoma in a leukemic phase (a poor prognostic condition) and to differentiate them from other atypical chronic lymphoproliferative disorders.39 40 

The BCL-2 gene and its protein product

BCL-2 gene.

The (14;18) chromosomal translocation in follicular lymphoma involves the BCL-2 gene at the chromosome 18 breakpoint, which encodes a 26-kd protein with limited homology to an Epstein-Barr viral protein (BHRF-1).41 Because breakpoints occur in the 3′ untranslated region, full-length protein is expressed after juxtaposition of the gene to the immunoglobulin heavy-chain promoter.41 Extensive studies have documented the role of BCL-2 in blocking apoptosis in many cell types and in response to a variety of stimuli.41 Many cellular and viral homologues have also been identified.

Antibodies to BCL-2.

Mouse and hamster monoclonal antibodies (mAbs) to BCL-2 protein have been described (Table 3).7,42 One of these, the mouse monoclonal reagent designated BCL-2/124,7 is used in many of the immunocytochemical studies discussed below.

BCL-2 in normal tissues.

BCL-2 is associated with mitochondria and cell membranes43and is widely distributed in both hemopoietic and nonhemopoietic cells.7,42 However, BCL-2 is absent from B-lymphoid cells in the germinal centers of B-cell follicles and from cortical thymocytes,7,44-46 and it ceases to be detectable when cells are transformed (eg, by mitogens).7 

BCL-2 in hematologic neoplasia.

BCL-2 protein is expressed in most cases of follicular lymphoma,47 both in the majority with t(14;18) and in many without it.48,49 However, a few cases lack both protein expression and rearrangement.50 BCL-2 protein is also expressed in many lymphoid and myeloid neoplasms,7,46,51-56 although it is usually absent or expressed at low levels in Burkitt lymphoma53,57 and anaplastic lymphoma kinase (ALK)–positive anaplastic large cell lymphoma (ALCL),58,59 possibly because of the high rate of cell proliferation.60 BCL-2 protein is also often absent or present at low levels in large B-cell tumors at extranodal sites, including the gastrointestinal tract.51,61,62 In contrast, lymph node–based B-cell neoplasms are commonly positive for BCL-2.54 Furthermore, although low-grade mucosa-associated lymphoid tissue (MALT) lymphomas of the gastrointestinal tract are BCL-2 positive,61 they often contain areas of larger cells that lack BCL-2 (Figure2A),63,64 probably representing early transformation to large cell neoplasms. This may be relevant to observations that large B-cell neoplasms arising from MALT are BCL-2 negative51 and that proliferative markers in nodal lymphomas are inversely related to expression of BCL-2.60 

Fig. 2.

BCL-2 immunostaining of neoplastic lymphoid tissue.

(A) In a low-grade MALT lymphoma, BCL-2 is expressed by the diffuse neoplastic infiltrate lying beneath the intestinal mucosa (Muc). BCL-2 is absent from reactive germinal centers (asterisks) in the tumor and from areas of high-grade transformation. The high-power view (bottom) shows the border between a low-grade area and a high-grade area. The arrows indicate large BCL-2–negative neoplastic cells. Original magnification top, × 200; bottom, × 800. (B) A pseudonegative follicular lymphoma in which the cells in a neoplastic follicle (Foll) lack BCL-2. However, a higher-power examination of the boxed area (center and bottom) shows that many of the BCL-2–positive small interfollicular cells (arrows) are neoplastic centrocytes (small cleaved cells). (Alkaline phosphatase–antialkaline phosphatase [APAAP] technique in paraffin sections; reproduced with permission from American Journal of Pathology47.) Original magnification top, × 250; center, × 400; bottom, × 800.

Fig. 2.

BCL-2 immunostaining of neoplastic lymphoid tissue.

(A) In a low-grade MALT lymphoma, BCL-2 is expressed by the diffuse neoplastic infiltrate lying beneath the intestinal mucosa (Muc). BCL-2 is absent from reactive germinal centers (asterisks) in the tumor and from areas of high-grade transformation. The high-power view (bottom) shows the border between a low-grade area and a high-grade area. The arrows indicate large BCL-2–negative neoplastic cells. Original magnification top, × 200; bottom, × 800. (B) A pseudonegative follicular lymphoma in which the cells in a neoplastic follicle (Foll) lack BCL-2. However, a higher-power examination of the boxed area (center and bottom) shows that many of the BCL-2–positive small interfollicular cells (arrows) are neoplastic centrocytes (small cleaved cells). (Alkaline phosphatase–antialkaline phosphatase [APAAP] technique in paraffin sections; reproduced with permission from American Journal of Pathology47.) Original magnification top, × 250; center, × 400; bottom, × 800.

Close modal

In follicular lymphomas,47 BCL-2 expression may also be heterogeneous, and the BCL-2–negative cells tend to be centroblasts (large noncleaved cells) rather than centrocytes (Figure 2B). As a result, cases at the aggressive end of the morphologic spectrum may appear to be BCL-2 negative when, in reality, few cells express the protein. This “pseudonegative” pattern is accentuated by the tendency of centrocytes to migrate away from neoplastic follicles so that large nodules of BCL-2–negative centroblasts are observed, with intervening smaller neoplastic cells that can be mistaken for normal cells (Figure 2B).47 It is possible that the BCL-2–negative large cell component is more sensitive to chemotherapy than is the small cell component, which may therefore persist and account for the indolent progression of the disease.

BCL-2 protein is found in Reed-Sternberg cells in one third to three fourths of all biopsy specimens from patients with Hodgkin disease (HD),45,65-67 including a few who have also had follicular lymphoma,68 but this presumably reflects its ubiquitous presence in lymphoid cells. However, the neoplastic cells of lymphocyte predominance HD (lymphocytic and histiocytic [L&H] or popcorn cells) tend to be BCL-2 negative,69,70 thereby illustrating the distinct nature of this HD subtype.11 

Clinical applications of BCL-2 immunostaining.

Although the ubiquitous distribution of BCL-2 protein limits its value for diagnosis, it is widely used for distinguishing reactive lymphoid follicles, which are BCL-2 negative, from neoplastic follicles and proliferation centers (found in chronic lymphocytic leukemia46,71-73), both of which are usually BCL-2 positive. In MALT lymphoma, for example, the reactive lymphoid follicles characteristically associated with this tumor (which may be confused with follicular lymphoma) are clearly negative (Figure2A),64 at least until they have undergone follicular colonization by neoplastic cells.74 A similar phenomenon has been observed in mantle cell lymphomas that localize around germinal centers (Figure3C).52 

Fig. 3.

BCL-6 immunostaining of normal and neoplastic lymphoid cells.

(A) BCL-6 expression in germinal centers in a lymph node showing follicular hyperplasia. (B) Diffuse large B-cell lymphoma. At left is a section stained with hematoxylin and eosin, and at right is a sample with BCL-6 immunostaining showing strong nuclear expression. (C) A trapped germinal center in a mantle lymphoma is BCL-2 negative (left) and BCL-6 positive (right). (D) BCL-6 expression by a popcorn/L&H cell (arrow) in nodular lymphocyte predominance HD (APAAP technique in paraffin sections; reproduced with permission from American Journal of Pathology106). Original magnification A, × 100; B left, × 800; B right, × 800; C left, × 200; C right, × 200; D, × 800.

Fig. 3.

BCL-6 immunostaining of normal and neoplastic lymphoid cells.

(A) BCL-6 expression in germinal centers in a lymph node showing follicular hyperplasia. (B) Diffuse large B-cell lymphoma. At left is a section stained with hematoxylin and eosin, and at right is a sample with BCL-6 immunostaining showing strong nuclear expression. (C) A trapped germinal center in a mantle lymphoma is BCL-2 negative (left) and BCL-6 positive (right). (D) BCL-6 expression by a popcorn/L&H cell (arrow) in nodular lymphocyte predominance HD (APAAP technique in paraffin sections; reproduced with permission from American Journal of Pathology106). Original magnification A, × 100; B left, × 800; B right, × 800; C left, × 200; C right, × 200; D, × 800.

Close modal

BCL-2 may also be of value as a marker for identifying infiltration by follicular lymphoma cells in bone marrow trephine biopsy specimens.75,76 Aggregates of normal or reactive lymphoid cells often stain only weakly (although T cells label more strongly than B cells) and have nondescript nuclear morphologic features. In contrast, follicular lymphoma cells show strong cytoplasmic BCL-2 labeling, which tends to emphasize their characteristic cleft nuclear morphologic features (Figure 2B), and this is often a valuable adjunct to the classic criterion of a paratrabecular infiltration pattern. It is also possible that cases of “Burkitt-like” lymphoma can be distinguished from epidemic/sporadic Burkitt lymphoma by the expression of BCL-2 protein,57 as can cases of lymphoblastic lymphoma/leukemia,77 although further studies of this issue are needed.

It has been suggested that the presence of high levels of BCL-2 protein promotes survival of neoplastic cells78 or confers drug resistance79 (or both) and thus is a marker of a poor prognosis. Many clinical studies have sought evidence of such a correlation, and an association between higher cellular BCL-2 levels and poorer disease-free survival at periods up to 10 years was found in several studies of diffuse large cell lymphoma (DLCL).80-86 There is also some evidence for a prognostic influence of BCL-2 levels in acute myeloid leukemia.87-91However, for other hematologic malignant diseases (HD,92follicular lymphoma,84,93,94 chronic lymphocytic leukemia,95,96 and acute lymphoblastic leukemia [ALL]97-99), the data are limited or equivocal.

Diffuse lymphomas therefore represent the most promising area for future studies of BCL-2 expression and offer the longer-term prospect of strategies for specifically inhibiting BCL-2.86However, few if any oncologists currently take BCL-2 levels into account when treating individual patients, and this variable has not been used in any controlled trials to assign patients with lymphoma to more aggressive treatment protocols.

The BCL-6 gene and its protein product

BCL-6 gene.

Studies in knockout mice found that BCL-6 protein is required for germinal-center formation, antibody-affinity maturation, and T-helper-2–mediated responses.100 BCL-6 appears to inhibit differentiation of germinal-center B cells toward plasma cells by binding to signal transducers and activators of transcription 3, thereby preventing the expression of Blimp-1 (a major regulator of plasma cell development).101 

Chromosomal translocations involving the 5′ noncoding domain of theBCL-6 gene102 at band 3q27 are observed in about 40% of diffuse large B-cell lymphomas103 and 10% to 15% of follicular lymphomas,103 juxtaposing the gene to promoters from a variety of partner chromosomes (most commonly in immunoglobulin chain loci). Mutations of the regulatory part of theBCL-6 gene are virtually absent in ALL and mantle cell lymphoma, but they occur frequently in germinal-center and postgerminal-center lymphomas, including follicular lymphomas, DLCLs, and Burkitt lymphomas.104 However, mutations of theBCL-6 gene have also been found in normal germinal-center B cells,105 and it is not clear what role they play in lymphomagenesis.

Antibodies to BCL-6.

Two mAbs (PG-B6p and PG-B6a) have been raised against a recombinant BCL-6 protein (Table 3). PG-B6p, which recognizes a formalin-resistant amino-terminal epitope, labels routinely processed biopsy specimens.106 

BCL-6 in normal tissues.

In lymphoid tissues, BCL-6 protein is expressed in germinal-center B cells in secondary follicles (Table 3 and Figure3A)106-111 in both the dark (centroblast-rich) and light (centrocyte-rich) zones.106-110 It is also detectable in a few CD4-positive T cells, both in the germinal centers and in interfollicular areas,107,108 and in perifollicular CD30+ lymphoid cells.107 In addition, BCL-6 protein is expressed in thymocytes, mainly in the cortex.112 Outside the lymphohemopoietic system, expression of BCL-6 has been observed in squamous epithelia.106,113 In all positive cells, BCL-6 protein localizes to the nucleus in a diffuse or microgranular pattern.106-108 110 

BCL-6 in lymphoid neoplasia.

Strong nuclear expression of BCL-6 occurs frequently in follicular lymphomas, paralleling its presence in germinal-center cells, their normal counterpart. BCL-6 expression is also found in most Burkitt lymphomas and diffuse large B-cell lymphomas (Figure 3B), regardless of whether the gene is rearranged.106-108,110,114 In lymphomas associated with human immunodeficiency virus, expression of BCL-6 and expression of the Epstein-Barr viral protein LMP1 appear to be mutually exclusive.115 

Neoplasms of B cells unrelated to germinal centers (eg, B-cell precursor malignant diseases, B-CLL, mantle cell and marginal zone lymphomas, and hairy cell leukemia) consistently lack BCL-6 protein (Figure 3C),106 in keeping with its absence from the putative normal counterparts of these neoplasms.11,116Among T-cell lymphomas, BCL-6 expression has been detected in about 50% of ALCLs117 and T-cell lymphoblastic lymphomas.112 Reed-Sternberg cells in classic HD are usually BCL-6 negative, but the tumor cells (L&H cells) in lymphocyte predominance HD are strongly positive (Figure 3D),118 in keeping with their probable germinal-center origin.69 119 

Clinical applications of BCL-6 immunostaining.

BCL-6 is a valuable marker of B cells of germinal-center origin.106,110,120 BCL-6 immunostaining, used in conjunction with BCL-2, may be useful in characterizing nodular lymphoid regions, since these can represent neoplastic germinal centers (indicative of follicular lymphoma), trapped residual germinal centers (eg, in MALT and mantle cell lymphomas; Figure 3C), or proliferation centers (in B-CLL).121 Labeling for BCL-6 may also help in the recognition of lymphocyte predominance HD (Figure3D).118 122 

The MUM1/IRF4 gene and its protein product

MUM1/IRF4 gene.

The 14q+ cytogenetic anomaly in multiple myeloma represents a cryptic t(6;14)(q25;p32) that juxtaposes the immunoglobulin heavy-chain locus to a gene known as MUM1 or IRF4.123The MUM1/IRF4 gene encodes a transcription factor thought to play a key role in lymphoid development, since IRF4-deficient mice have a block in peripheral B-cell maturation (absence of germinal centers and plasma cells) associated with a lack of cytotoxic T-cell responses.124 

Antibody to MUM1/IRF4.

A recently generated mAb (MUM1p)125 against human MUM1/IRF4 protein is suitable for immunostaining routinely prepared paraffin sections and for Western blotting, in which it detects a 50-kd band of expected molecular weight.

MUM1/IRF4 in normal tissues.

In normal lymphoid tissue (Table 3), MUM1/IRF4 protein is detected mainly in plasma cells and in a small number of germinal-center B cells, which are usually BCL-6 negative and nonproliferating (Ki67 negative) and found mainly in the light zone of the germinal center.125 In addition, MUM1/IRF4 is expressed in a small percentage of T cells and in most perifollicular CD30-positive cells,125 in keeping with its expression by peripheral blood T cells after stimulation with phytohemagglutinin.125 In all positive cells, MUM1/IRF4 protein expression is primarily nuclear and shows a microgranular and diffuse pattern, with sparing of nucleoli.125 

MUM1/IRF4 in lymphoid neoplasia.

In accordance with its expression at late stages of B-cell differentiation and after T-cell activation, MUM1/IRF4 protein is strongly expressed in lymphoplasmacytic lymphoma/immunocytoma, about 75% of diffuse large B-cell lymphomas, primary effusion lymphomas,126 HD, multiple myeloma, and ALCL.125,127 128 

Clinical applications of MUM1/IRF4 immunostaining.

Because of the restriction of MUM1/IRF4 to late-stage B cells, it is potentially valuable (used in combination with other plasma cell markers, such as CD138) for recognizing myelomas (Figure4A). Furthermore, its consistent expression in neoplastic cells in HD (Figure 4B) can be of diagnostic help when the staining highlights Reed-Sternberg and Hodgkin cells in a tissue biopsy specimen (eg, of bone marrow).

Fig. 4.

Expression of the plasma cell–associated marker MUM1/IRF4 in neoplastic lymphoid cells.

(A) Bone marrow biopsy specimen showing infiltration by multiple myeloma. Monoclonal MUM1/IRF4 is present in the nuclei and the perinuclear area in neoplastic plasma cells. Residual hemopoietic elements are unlabeled. The inset shows higher magnification-view of the same field. (B) Strong labeling of tumor cells in classic HD (APAAP technique in paraffin sections). Original magnification A, × 600; A inset, × 1000; B, × 1000.

Fig. 4.

Expression of the plasma cell–associated marker MUM1/IRF4 in neoplastic lymphoid cells.

(A) Bone marrow biopsy specimen showing infiltration by multiple myeloma. Monoclonal MUM1/IRF4 is present in the nuclei and the perinuclear area in neoplastic plasma cells. Residual hemopoietic elements are unlabeled. The inset shows higher magnification-view of the same field. (B) Strong labeling of tumor cells in classic HD (APAAP technique in paraffin sections). Original magnification A, × 600; A inset, × 1000; B, × 1000.

Close modal

The PAX-5 gene and its protein product

PAX-5 gene.

The PAX-5 gene (a member of the paired-box PAXgene family) encodes the transcription factor B-cell–specific activator protein (BSAP), which plays a key role in B-lymphoid development129 and is also involved in the embryonic development of the central nervous system and testis. In the t(8;14)(p13;q32), which is characteristic of small lymphocytic lymphoma with plasmacytoid differentiation (immunocytoma), the PAX-5 gene (on 9p13) is juxtaposed to the immunoglobulin heavy-chain gene (on 14q32).130 

Antibodies to PAX-5.

A polyclonal antibody to PAX-5 has been described,131 and a mAb is available commercially (Transduction Laboratories, Lexington, KY; Table 3). Both detect PAX-5 protein in paraffin-embedded samples.

Expression of PAX-5 in normal tissues.

In tissue sections, PAX-5 protein can be stained in B-cell follicles but not plasma cells.131 Unlike B-cell–associated transcription factors such as BCL-6 and MUM1, which are also expressed in activated T cells,106,107,125 PAX-5 appears to be B-cell specific131 (Table 3). Experimental studies have shown that PAX-5 protein is expressed in B-lymphoid precursors, but no data on direct immunostaining in human bone marrow have been reported.

Expression of PAX-5 in lymphohemopoietic neoplasms.

PAX-5 has been found in many B-cell lymphomas (with the strongest expression in follicular, mantle cell, and DLCLs)131 but not in T-cell neoplasms.

Clinical applications of PAX-5 immunostaining.

PAX-5 may prove to be a valuable diagnostic marker in paraffin-embedded biopsy specimens of B-lymphoblastic neoplasms because it is expressed strongly in such samples and is negative in T-cell lymphoblastic proliferations (Figure 5A; B.F., unpublished data, 2001). HD sometimes mimics ALCL, and PAX-5 may be useful in such cases because Reed-Sternberg cells are usually positive132 (Figure 5B), whereas ALCLs (both ALK positive and ALK negative) are consistently negative for PAX-5.

Fig. 5.

Expression of the PAX-5 (BSAP) transcription factor in neoplastic lymphoid cells.

(A) Pro-B acute lymphoblastic leukemia cells in a bone marrow biopsy specimen are selectively stained. The inset is a higher magnification, showing the irregular nuclei of PAX-5–positive leukemic cells. (B) Tumor cells of classic HD showing nuclear labeling, as well as cytoplasmic staining. Normal residual mantle zone B cells also express PAX-5 (APAAP technique in paraffin sections). Original magnification A, × 400; A inset, × 800; B, × 1000.

Fig. 5.

Expression of the PAX-5 (BSAP) transcription factor in neoplastic lymphoid cells.

(A) Pro-B acute lymphoblastic leukemia cells in a bone marrow biopsy specimen are selectively stained. The inset is a higher magnification, showing the irregular nuclei of PAX-5–positive leukemic cells. (B) Tumor cells of classic HD showing nuclear labeling, as well as cytoplasmic staining. Normal residual mantle zone B cells also express PAX-5 (APAAP technique in paraffin sections). Original magnification A, × 400; A inset, × 800; B, × 1000.

Close modal

The TAL-1 gene and its protein product

TAL-1 gene.

The TAL-1 gene (also known as SCL orTCL-5)133 at 1p32 encodes a 42-kd nuclear protein134 that is essential for mammalian hematopoiesis133 and normal yolk sac angiogenesis.135 Structural alterations in theTAL-1 gene represent the most frequent molecular lesions in T-cell ALL (T-ALL).1,133 In up to 25% of childhood cases, a submicroscopical 90–kilobase-pair deletion on the 5′ side of the gene133 brings it under the influence of promoters for theSIL gene. Much less commonly, the gene is rearranged by a chromosome translocation; this is usually t(1;14)(p32;q11), which associates the gene with the TCRδlocus.133 136 

Antibodies to TAL-1.

Several mAbs suitable for immunocytochemical detection of TAL-1 have been described (Table 3).137 138 

TAL-1 in normal tissues.

Studies of TAL-1 protein or TAL-1 mRNA found that the gene is expressed in some, but not all, hemopoietic lineages (notably, erythroid precursors, megakaryocytes, and mast cells; Figure6A),137,139,140 but it is not detectable in even the most immature T cells (thymocytes and CD2-positive precursor lymphoid cells in fetal liver).141TAL-1 immunostaining is localized to the nucleus (Figure 6A-C),137 characteristically in the form of small dots (Figure 6C),137 whose number is greater than that of PML-positive structures. In mitotic cells, TAL-1 is consistently cytoplasmic.137 

Fig. 6.

TAL-1 expression in normal hemopoietic cells and neoplastic cell lines.

(A) At left is a sample from fetal liver that shows numerous scattered TAL-1–positive nuclei. At right is a sample with double staining for TAL-1 (in brown) in combination with cytokeratin (CK), transferrin receptor (TfR), or platelet glycoprotein (Plt) (all in red) that identifies the TAL-1–positive cells, many of which are intrasinusoidal (arrow), as erythroid precursors and megakaryocytes. (B) The erythroleukemic HEL cell line shows strong nuclear positivity. Nucleoli are unstained. (C) TAL-1 is also expressed in the K562 erythroid cell line, localized to discrete intranuclear bodies and often in an annular pattern (immunoperoxidase [A-C] and APAAP [A] techniques in cryostat sections [A] and cytospin preparations [B,C]; reproduced with permission from Blood137). Original magnification A left, × 100; A right, × 800; B, × 800; C, × 1000.

Fig. 6.

TAL-1 expression in normal hemopoietic cells and neoplastic cell lines.

(A) At left is a sample from fetal liver that shows numerous scattered TAL-1–positive nuclei. At right is a sample with double staining for TAL-1 (in brown) in combination with cytokeratin (CK), transferrin receptor (TfR), or platelet glycoprotein (Plt) (all in red) that identifies the TAL-1–positive cells, many of which are intrasinusoidal (arrow), as erythroid precursors and megakaryocytes. (B) The erythroleukemic HEL cell line shows strong nuclear positivity. Nucleoli are unstained. (C) TAL-1 is also expressed in the K562 erythroid cell line, localized to discrete intranuclear bodies and often in an annular pattern (immunoperoxidase [A-C] and APAAP [A] techniques in cryostat sections [A] and cytospin preparations [B,C]; reproduced with permission from Blood137). Original magnification A left, × 100; A right, × 800; B, × 800; C, × 1000.

Close modal

TAL-1 in hematologic neoplasia.

The TAL-1 gene is activated in about one fourth of all cases of T-ALL, but the link with expression of TAL-1 protein remains unclear. In several T-ALL lines, mRNA and/or protein was found in the absence of detectable gene rearrangement,137,142 andTAL-1 mRNA has been detected by reverse transcriptase–polymerase chain reaction (RT-PCR) in most cases of T-ALL.143,144 However, this may derive from normal monocytes and T cells in the samples145: the results of an immunocytochemical study suggested that TAL-1 protein is only rarely expressed by leukemic cells in T-ALL when the gene is not rearranged.146 

Clinical applications of TAL-1 immunostaining.

In a study by Chetty et al,147 samples from approximately 50% of T-ALL cases showed nuclear labeling, but the investigations were done in paraffin-embedded tissue, on which the available mAbs do not provide clean labeling. However, immunocytochemical labeling of fresh T-ALL samples that were also studied with molecular biologic techniques suggested that cases in which the TAL-1 gene is rearranged can be detected by immunocytochemistry.146There appears to be no correlation between TAL-1 gene rearrangement and clinical behavior in T-ALL.148 

Several chromosomal translocations cause fusion of unrelated genes rather than overexpression of intact quiescent genes, and these hybrid genes encode tumor-associated chimeric proteins.1 2 This situation has reawakened interest in producing tumor-specific antibodies, but unfortunately, antibodies specific for the novel junctional regions in these chimeric proteins have proved to be difficult to raise. In addition, genes may break at different points and thus give rise to more than one fusion protein, with different junctional regions. In spite of these obstacles, progress has been made by using antibodies directed against nonjunctional parts of chimeric proteins, since such antibodies can detect relocalization (eg, PML) or de novo expression induced by gene fusion (eg, ALK).

The (15;17) translocation and its variants

The PML and RARα genes.

The (15;17) chromosomal translocation, a specific marker for acute promyelocytic leukemia (APL),149 fuses the retinoic acid receptor-α (RARα) gene to a growth suppressor gene (PML) that encodes a protein thought to be involved in regulation of p53 acetylation and premature senescence induced by oncogenic Ras.150 The PML-RARα fusion protein may play a key role in the pathogenesis of APL by blocking terminal differentiation and inhibiting apoptotic cell death,149and studies indicate that it acts by recruiting histone deacetylase through the RARα portion of the chimeric protein.151,152In the rare cases of APL carrying variants of the t(15;17) (Table 1), the RARα gene is fused to the nucleophosmin(NPM), PLZF, or nuclear mitotic apparatus(NuMA) gene.153 

Antibodies to PML.

Most PML isoforms and PML-RARα fusion proteins can be detected by immunocytochemical labeling with mAb PG-M3154 (Table 3), which recognizes an epitope near the cysteine-rich (putative DNA-binding) region. Another anti-PML mAb (5E10)155 has been described, but it is directed against an epitope probably located in a region (between amino acids 448 and 466) that is lost in some PML-RARα forms (eg, type S and bcr3 PML-RARα; Table3).156 Anti-PML antibodies fail to identify endogenous PML in Western blotting analyses,154 possibly because of the small amounts of each PML isoform (13 have been identified157) resolved by gel electrophoresis.

PML in normal tissues.

Wild-type PML protein is expressed ubiquitously, but levels vary greatly according to cell type.154,158-160 PML protein is localized to discrete nuclear dots (average, 10 dots/nucleus) not associated with nucleoli (Figure7A).154,156,161-163 The diameter of these dots ranges from 0.5 to 1 μm,162 and they correspond to nuclear domains (PML oncogenic domains [PODs])164,165 reorganized previously by electron microscopy as “nuclear bodies”166 and by immunocytochemistry as “nuclear dots” or “nuclear domain 10.”167 These macromolecular protein-rich complexes are tightly bound to the nuclear matrix155,163,164 and are known to contain several other proteins, including Sp100, NDP52, PIC1/SUMO-1, and Int-6.165,168-174 Progressive accretion of proteins gives rise to a characteristic ringlike appearance,154,156 162 especially in cells overexpressing PML.

Fig. 7.

Staining for wild-type and hybrid PML proteins.

(A) Speckled nuclear staining, representing wild-type PML, in transfected U937 cells (left) and in a case of FAB-M5 acute leukemia (right). (B) Nuclear microspeckled staining pattern indicating the presence of hybrid PML-RARα protein in 2 cases of APL. Single arrows indicate normal residual marrow cells; and arrowhead, a red blood cell (APAAP and immunofluorescent techniques in cytospin preparations; reproduced with permission from Blood177). (C) Laser-scan confocal microscopy of the KG1 myeloid cell line double stained for PML (green) and PLZF (red). At left is the confocal image. At right is a 3-dimensional reconstruction of the area outlined on the left panel, showing the spatial relation between PML and PLZF in the nuclear bodies. Original magnification A-C, × 1000.

Fig. 7.

Staining for wild-type and hybrid PML proteins.

(A) Speckled nuclear staining, representing wild-type PML, in transfected U937 cells (left) and in a case of FAB-M5 acute leukemia (right). (B) Nuclear microspeckled staining pattern indicating the presence of hybrid PML-RARα protein in 2 cases of APL. Single arrows indicate normal residual marrow cells; and arrowhead, a red blood cell (APAAP and immunofluorescent techniques in cytospin preparations; reproduced with permission from Blood177). (C) Laser-scan confocal microscopy of the KG1 myeloid cell line double stained for PML (green) and PLZF (red). At left is the confocal image. At right is a 3-dimensional reconstruction of the area outlined on the left panel, showing the spatial relation between PML and PLZF in the nuclear bodies. Original magnification A-C, × 1000.

Close modal

PML in hematologic neoplasia.

In leukemias other than APL (and in lymphomas), PML labeling consistently shows a wild-type speckled pattern (Figure 7A, right). In cells containing the PML-RARα fusion protein (eg, in APL blasts and transfected cells), the PML nuclear bodies are disrupted and the wild-type speckled pattern is replaced by a microspeckled pattern (Figure 7B) consisting of many tiny dots (often too numerous to count) in which PML-RARα and PML colocalize.156,161,162,164Treatment of APL cells in vitro or in vivo with all-trans-retinoic-acid (ATRA) or arsenic trioxide (As2O3) restores the wild-type speckled PML pattern.156,161,162,164,175 Wild-type PML is often expressed at higher levels in Reed-Sternberg cells in HD than in neoplastic cells of other lymphoid malignant diseases.160 

Clinical applications of PML immunostaining.

APL can be diagnosed rapidly by showing the microgranular PML immunocytochemical labeling pattern (Figure 7B).176-180This appearance is specific for APL with t(15;17), since the variants t(11;17) and t(5;17) do not disrupt the wild-type speckled pattern.181-184 Immunostaining must be done on fresh samples (smears, cytospin preparations, or frozen sections), since fixation and paraffin embedding change the wild-type speckled pattern to a diffuse pattern.160,177 Immunocytochemistry is especially useful in recognizing the microgranular variant of APL (M3V)—which is characterized by sparse cytoplasmic granules and marked nuclear irregularity, with folding, lobulation, or convolution—because it can be difficult to distinguish M3V from acute myelomonocytic or monocytic leukemia (M4 and M5B).185 The immunocytochemical diagnosis of APL is clinically valuable because the disease responds to treatment with ATRA186,187 and As2O3.188 Variant translocations are not detected by immunostaining for PML, but they are rare and at least some of them, such as t(11;17), do not respond to ATRA treatment.149 189 

The (11;17) and (5;17) translocations.

In the rare t(11;17) and t(5;17) anomalies, the portion of RARα present in the PML-RARα chimeric protein is fused to the Kruppel-like transcription factor PLZF or NuMA (the nuclear mitotic apparatus protein) (both of which are encoded by genes on chromosome 11), or to the amino-terminal of NPM.190-192 There are conflicting reports on the subcellular distribution of the PLZF protein: studies have observed localization to small punctate nuclear domains different from PODs,193 colocalization of PLZF and PML in nuclear bodies,194 and localization to 0.3- to 0.5-μm nuclear bodies (PLZF bodies) adjacent to but functionally distinct from PML nuclear bodies (Figure 7C).195 However, it is agreed that PLZF-RARα and PML-RARα colocalize in the same microspeckles.194,195 PLZF is expressed mainly in CD34+ progenitors196 but not in more mature myeloid cells, and it may also be involved in central nervous system differentiation.197 Because of the rarity of these APL molecular variants, no clinical studies of the value of antibodies to their products have been reported.

The (2;5)(p23;q35) translocation and its variants

The NPM and ALK genes.

The (2;5)(p23;q35) translocation, which is associated with ALCL,198-200 fuses the NPM gene, which encodes the ubiquitously expressed nucleolar phosphoprotein NPM, on chromosome 5 to the ALK gene, which encodes a receptor tyrosine kinase.201,202 The resulting NPM-ALK hybrid protein (also known as p80) contains 40% of the amino-terminal portion of NPM linked to the entire intracytoplasmic domain of ALK.203-206 Six variant translocations in which ALK fuses to a partner other than NPM have been identified in human tumors207-213 (Table4), but the classic NPM-ALK anomaly accounts for more than 80% of ALK fusion genes. TheALK gene appears not to be transcribed in normal lymphoid cells, but the promoter or promoters for NPM or other partners are assumed to induce transcription of the ALKfusion genes in lymphoma cells.201 NPM-ALK kinase activity is induced as a result of cross-linking by the NPM portion or one of the other fusion partners. Initial estimates of the frequency of t(2;5) and the resulting NPM-ALK gene in ALCL varied widely,214-219 but most of this uncertainty has since been resolved through immunocytochemical studies of ALK expression.

Antibodies to ALK.

The laboratory that identified the p80 protein (later shown to be identical to NPM-ALK) prepared an affinity-purified polyclonal antibody to the kinase domain,220-222 and several immunocytochemical studies were conducted with samples made available to other researchers.218,219,223,224 A second polyclonal antibody (anti-ALK 11) has been used on a more limited scale for biochemical and immunocytochemical studies.225,226However, most studies of ALK protein are now done with mAbs. Two such reagents have been described: ALK1 and ALKc. These recognize the cytoplasmic portion of ALK227,228 and are both suitable for immunolabeling paraffin-embedded biopsy specimens and for Western blot analyses.227-230 

Antibodies to NPM.

MAbs against the carboxy-terminal portion of NPM were described more than 15 years ago.231 More recently, 3 paraffin-reactive antibodies recognizing epitopes on the carboxy-terminal portion (which is lost in the NPM-ALK hybrid kinase) or the amino-terminal portion (present in NPM-ALK) have been raised.232 

Expression of ALK and NPM in normal tissues and cell lines.

ALK protein cannot be detected in tissues of human adults, except in a few cells in the nervous system that show weak labeling.227,228 RT-PCR has detected NPM-ALKand ATIC-ALK mRNA at low levels in normal and reactive lymphoid cells,233,234 but ALK-positive circulating cells cannot be detected by immunocytochemistry.227 Cell lines with t(2;5) are ALK positive, as are some neuroblastoma cell lines235 and the rhabdomyosarcoma line Rh30,227 all of which express full-length ALK protein.201,227 In contrast to ALK, NPM can be detected in most human cells by immunocytochemical techniques, usually confined to cell nuclei.232 

Expression of ALK in lymphoid neoplasia.

The absence of ALK protein from normal lymphoid tissue means that positive ALK staining is a near-specific marker for lymphomas containing a hybrid ALK gene (Figure8). When a lymphoma expresses ALK, the subcellular labeling pattern is informative (Table 4). Thus, in cases with the classic t(2;5), ALK is observed not only in the cytoplasm but also in the nucleus (Figure 8A), whereas tumors expressing ALK fusion proteins other than NPM-ALK show no nuclear labeling because they do not dimerize with wild-type NPM (Figure8C).227,228,230,236,237 Furthermore, 2 ALK fusion proteins (CTCL-ALK and moesin-ALK) were found to have unique immunohistochemical staining patterns (granular cytoplasmic and membrane-associated, respectively; Figure 8C).210,212 Staining for the amino-terminal of NPM can also be informative because it is detected in both the cytoplasm and the nucleus in tumor cells with t(2;5),232 whereas staining in lymphomas with variant translocations is restricted to the nucleus.238 Western blotting of cryostat-section extracts can also be used to characterize variant ALK proteins, whose molecular weights differ from that of NPM-ALK.239 

Fig. 8.

Immunostaining of lymphomas for ALK proteins.

(A) Labeling for NPM-ALK in the nucleus and cytoplasm in a lymphoma with anaplastic large cell morphologic characteristics. (B) A t(2;5)-positive large cell lymphoma showing the contrast between NPM-ALK expression (cytoplasmic with some nuclear staining) and CD30 staining (surface membrane and Golgi-associated labeling). The double fluorescent images at bottom show that the same cells express CD30 and ALK but in different patterns. (C) Different patterns of ALK immunostaining in lymphomas. All tumors contain a hybrid ALK protein, except for that marked ALK, an example of the rare subtype of large B-cell lymphoma (CD30-negative and positive for epithelial membrane antigen) in which full-length wild-type ALK protein is expressed. (Immunoperoxidase and immunofluorescence techniques in paraffin sections; reproduced with permission from Blood227,258 and Laboratory Investigation212.) Original magnification A left, × 250; A right, × 1000; B top, × 1000; B bottom (ALK and CD30, × 400; ALK/CD30, × 1000); C, × 400 to × 1000.

Fig. 8.

Immunostaining of lymphomas for ALK proteins.

(A) Labeling for NPM-ALK in the nucleus and cytoplasm in a lymphoma with anaplastic large cell morphologic characteristics. (B) A t(2;5)-positive large cell lymphoma showing the contrast between NPM-ALK expression (cytoplasmic with some nuclear staining) and CD30 staining (surface membrane and Golgi-associated labeling). The double fluorescent images at bottom show that the same cells express CD30 and ALK but in different patterns. (C) Different patterns of ALK immunostaining in lymphomas. All tumors contain a hybrid ALK protein, except for that marked ALK, an example of the rare subtype of large B-cell lymphoma (CD30-negative and positive for epithelial membrane antigen) in which full-length wild-type ALK protein is expressed. (Immunoperoxidase and immunofluorescence techniques in paraffin sections; reproduced with permission from Blood227,258 and Laboratory Investigation212.) Original magnification A left, × 250; A right, × 1000; B top, × 1000; B bottom (ALK and CD30, × 400; ALK/CD30, × 1000); C, × 400 to × 1000.

Close modal

Extensive immunohistologic studies have shown that ALK-positive tumors correspond only partly to what pathologists diagnosed in the past as ALCL.217,219,223,227,228,237 ALK-positive lymphomas typically contain a population of medium to large hallmark neoplastic cells, with a reniform eccentric nucleus and a juxtanuclear hof in the Golgi region.222,230 However, the morphologic variation is wide228-230,240,241 and includes cases with a relatively uniform proliferation of hallmark cells,242tumors in which many bizarre large cells (sometimes with a sarcomatoid appearance) are observed, and neoplasms containing large numbers of macrophages (the lymphohistiocytic pattern) or other reactive cells.11 243-245 

ALK-positive lymphomas may contain areas of small cells that also express ALK protein.228,230 This indicates that the large cells do not represent cytologic transformation of a small cell tumor after acquisition of an ALK translocation. In cutaneous T-cell lymphoma, cellular transformation can create this mixed large and small cell pattern, but these cases are ALK negative and lack theNPM-ALK gene.246 When the small cell component is prominent,247 the tumor is sometimes referred to as the “small cell variant” of ALK-positive lymphoma.228,230,236,237 There is no reason, however, to think of this as a different disease, although spread of the disease to the peripheral blood and bone marrow may be more likely in cases of this sort.248 249 

ALK-positive lymphomas usually express T-cell markers and/or cytotoxic granule proteins, CD30, and epithelial membrane antigen and c-Myc.219,222,228,230,250 Unlike ALK-negative cases, ALK-positive ALCLs are consistently BCL2 negative.58,59 We have found that tumors that are diagnosed morphologically as ALCL but have a B-cell phenotype are always ALK negative,228,251and they probably represent one end of the morphologic spectrum of diffuse large B-cell neoplasia.11,251 Primary CD30-positive cutaneous lymphomas are also ALK negative, in keeping with the absence of the NPM-ALK gene.252Finally, the controversial proposed “Hodgkin-like” subtype of ALCL11 is almost always ALK negative219,227,228,230 (as are Reed-Sternberg cells219,223,224,227 228) and probably represents true HD.

Most important, lymphomas defined by ALK labeling appear to be clinically homogeneous, regardless of whether they have the classic t(2;5) or one of its variants. They are usually found in young men presenting with advanced disease (stage III-IV), are often associated with systemic symptoms (especially fever) and involvement of extranodal sites (especially skin, bone, and soft tissues), respond well to chemotherapy, and have a favorable outcome.221,237,253,254Whether the high proliferative rate of the tumor,255 the host immune response to the ALK protein,256 or both these factors contribute to this pattern must be clarified in additional studies. In contrast, ALK-negative lymphomas with anaplastic large cell morphologic features tend to occur in older patients and usually have an unfavorable prognosis.221,253,254 257 

Rare cases of ALK-positive large B-cell lymphoma have been described. These are characterized by immunoblastic (rather than anaplastic) morphologic features, expression of full-length ALK, and cytoplasmic IgA (Figure 8C).258 The ALK-positive lymphomas with a B-cell phenotype described by Gascoyne et al254 may belong to this rare category, but there is insufficient information to confirm this.

ALK protein is not detectable in most nonhematopoietic neoplasms. The only exceptions are a few cases of rhabdomyosarcoma228and neuroblastoma,235 both of which produce full-length ALK, possibly reflecting their primitive origin. In contrast, in the rare entity known as “inflammatory myofibroblastic tumor,” ALK expression is commonly found and appears to reflect the presence of an ALK fusion protein (eg, TPM3-ALK, TPM4-ALK, or clathrin-ALK).208 259-261 

Clinical applications of anti-ALK antibodies.

Immunohistologic staining for ALK in lymphoid tissue biopsy specimens is now a widely used hematopathologic marker for diagnosing the clinicopathologic entity associated with t(2;5) and its variants, for which the name “ALK-positive lymphoma”228,230,236seems most appropriate (rather than “ALCL”). ALK labeling can also be of value in discerning low levels of tumor tissue infiltration in such cases (eg, in bone marrow trephine specimens249,253) and identifying cases in which the neoplastic cells are obscured by macrophages.229 The small cell variant of ALK-positive lymphoma247 can also be distinguished, because of its positive labeling for ALK, from peripheral T-cell lymphomas.228 Moreover, inflammatory myofibroblastic tumors208 that express the ALK protein as a result of translocations involving the ALK gene can also be diagnosed by using ALK labeling, and the absence of CD30 distinguishes them from ALK-positive lymphomas.

Other translocations.

Two translocations involving fusion of NPM with genes other than ALK have been described, and both are rare. The (5;17)(q32;q21) translocation in APL fuses the NPM gene to the RARα gene,191 and the t(3;5)(q25.1;q34), which is found occasionally in myelodysplastic syndrome/acute myeloid leukemia,262 fuses NPM to the myelodysplasia/myeloid leukemia factor 1 (MLF1) gene. It was reported that the resulting NPM-MLF1 hybrid protein localizes to the nucleus, whereas wild-type MLF1 protein is found in the cytoplasm,262 thus indicating that the NPM moiety targets MLF1 to a novel site.

Studies of the proteins encoded by genes involved in chromosomal alterations in hematologic neoplasms have lagged behind molecular biologic investigations of these anomalies because of the investment of resources required to make satisfactory antibodies, a process that depends on the availability of antigen. It is sometimes possible to raise highly specific antibodies against synthetic peptides (eg, anti-BCL-27 and PML154), but the best immunogens are probably recombinant proteins, the production of which is not simple. Furthermore, although polyclonal reagents have been used for immunocytochemical studies and Western blotting analyses (in which nonspecific reactions can be to some extent ignored), they may introduce problems of nonspecific reactivity, variation between different samples, and limited availability. Therefore, mAbs are preferable, but reagents that satisfy stringent criteria for specificity are not easy to produce. Antibodies should detect fixation-resistant epitopes in paraffin-embedded tissue, but those that meet this requirement are more difficult to produce.

Despite these problems, specific antibodies to the products of genes involved in hematologic neoplasia have provided major new insights, particularly when the protein target is relevant not only to hematologic neoplasia specifically but also to cell physiology in general. The best of example of this is BCL-2, a protein that was first studied in lymphomas with the t(14;18) but that subsequently was found to be extremely important in apoptosis.

Most antibodies discussed here can detect their targets through biochemical methods (Western blotting or immunoprecipitation). These techniques are particularly applicable to the study of hybrid genes, since they allow chimeric proteins (eg, the products of the hybridBCR-ABL and NPM-ALKgenes239,263,264) to be distinguished from wild-type proteins on the basis of their unique molecular sizes. Biochemical analysis can also be useful when a gene (eg,AML1265) encodes several transcripts. However, compared with immunocytochemistry, biochemical methods are more demanding technically and provide only limited information on tissue distribution and subcellular localization of the proteins.

Immunocytochemistry thus has obvious advantages for studying tissue distribution and localization. We have here discussed genetic anomalies that switch on quiescent genes separately from those that create fusion genes. For the anomalies that switch on genes, a major consideration is the expression pattern of the gene product in normal cells. Immunocytochemical studies of widely expressed genes (eg,c-MYC) generally have little clinical value. In contrast, few normal cells express the products of the BCL-1 andTAL-1 genes, and immunocytochemical staining for these proteins has a role in the diagnosis of mantle cell lymphoma and T-ALL, respectively.

Molecules such as BCL-2 and BCL-6 are in an intermediate position: both are expressed by many cells in the absence of gene rearrangement, but immunocytochemical labeling can be useful in certain situations. For example, BCL-2 is a good marker for the t(14;18) if normal germinal-center B cells, which are consistently BCL-2 negative, are compared with their neoplastic counterparts, follicular lymphoma cells, which are usually BCL-2 positive. Thus, immunocytochemical detection of BCL-2 protein has a diagnostic role in distinguishing between reactive and neoplastic follicle centers, even though it is widely expressed outside the germinal center. BCL-6 is not a comparably specific immunocytochemical marker for gene translocation (even in a restricted context), but it has diagnostic value as a molecule associated with germinal-center cells.

Antibodies to chimeric proteins generated by fusion genes are theoretically less suitable for immunocytochemical analyses, since they are usually raised against one of the 2 constituent proteins and therefore react with both wild-type and hybrid proteins. Antibodies specific for junctional epitopes unique to the chimeric protein should avoid this problem, but despite reports of such epitopes on the BCR-ABL,266,267 AML1-ETO,265 and E2A-PBX1268 chimeric proteins—created respectively by t(9;22), t(8;21), and t(1;19)—the only antibodies that appear potentially suitable for immunocytochemical use are monoclonal reagents specific for the E2A-PBX1 junction.268 These, however, apparently have not been used since their initial description.

Therefore, although in theory it should be possible to produce antibodies specific for hybrid proteins, in practice such antibodies remain elusive. If short recombinant protein sequences (or synthetic peptides) are used as immunogens, the junctional region may not adopt the same 3-dimensional conformation that it has in the chimeric protein in vivo. Furthermore, the junctional sequences may not be immunogenic, an issue that has been addressed, at least with respect to T-cell recognition, in several studies. For example, there is evidence that junctional BCR-ABL peptides can be bound by HLA class I molecules,269,270 and several investigators271 have reported that in vitro immunization with a peptide from p210 BCR-ABL can elicit a specific cytotoxic response. However, it is not yet clear whether the BCR-ABL junctional sequence is immunogenic in vivo.272 Peptides eluted from HLA molecules on fresh chronic myeloid leukemia cells include those from the BCR protein but not junctional sequences.273 

Antibodies specific for chimeric oncogene products will therefore always be difficult, if not impossible, to produce. However, the 2 examples discussed here (PML-RARα and NPM-ALK) show that antibodies to normal constituents of a hybrid protein can be used to detect underlying genetic abnormalities. In the case of t(15;17), the intranuclear distribution of the chimeric PML-RARα protein is so distinctively different from that of wild-type PML that the existence of the PML-RARα hybrid gene can reliably be inferred. The presence of the NPM-ALK fusion gene generated by t(2;5) can be assumed when a tumor shows ALK labeling in the nucleus and cytoplasm. When labeling is restricted to the cytoplasm, a variant hybrid ALK gene is likely to be present (since ALK protein is not expressed in normal lymphoid tissues), and labeling and Western blotting with anti-NPM can confirm this. ALK therefore behaves like proteins such as BCL-1 or BCL-2, which are switched on by gene rearrangement. Because of the absence of ALK protein from normal lymphohemopoietic cells, ALK fulfills most of the criteria for that elusive entity—a tumor-specific antigenic marker.

Immunocytochemical labeling for the NPM-ALK chimeric protein illustrates a point of wider relevance: the importance of subcellular localization patterns. Several proteins move from the cytoplasm to the nucleus under physiologic conditions. For example, the dimeric p50/p65 nuclear factor-κβ transcription factor is normally held in the cytoplasm by the inhibitor protein Iκβ, but it translocates to the nucleus when Iκβ is inactivated by phosphorylation.274Epidermal growth factor was also found to relocate to the nucleus, after binding of its ligand.275,276 Such observations are often made when proteins affected by genetic changes in hematologic neoplasia alter their subcellular localization (Table5), and it is frequently concluded that the sites to which proteins relocate must be the sites of their oncogenic action. However, the NPM-ALK chimeric protein, for example, despite its prominent nuclear (and, in particular, nucleolar) localization, almost certainly exerts its lymphomagenic effect within the cytoplasm.277 The chimeric PML-RARα protein shows no tendency to associate with nucleoli; thus, the hybrid NPM-RARα protein that is produced in rare cases of APL (and which presumably also accumulates in nucleoli) probably does not exert its transforming effect at this site.

Another leukemia-associated protein that relocates to a site that may not be relevant to its action is PML, which is displaced in APL cells from its normal target sites (ie, nuclear bodies) through heterodimerization with the PML-RARα fusion protein.156,161,162,164 However, a PML-RARα mutant that cannot dimerize with PML in coimmunoprecipitation experiments and does not delocalize PML from the nuclear bodies is nevertheless also capable of blocking differentiation.278 Moreover, in some cases of APL, there are variant translocations that presumably contribute directly to leukemogenesis but that encode fusion proteins (PLZF-RARα,190 NPM-RARα,191 and NuMA-RARα192) that do not alter the localization of PML, thereby suggesting that interference with retinoid signaling rather than disruption of PML nuclear bodies is the important event.

In conclusion, antibodies specific for the products of genes involved in chromosomal changes in leukemia and lymphoma have proved to be valuable both for studying normal cell physiology (eg, BCL-2) and for diagnosis (eg, BCL-1, PML, and NPM-ALK). The scope of antibody-based studies should continue to expand as new genetic changes in lymphoma and leukemia are identified.279 Furthermore, microarray studies of the expression of large numbers of gene sequences are likely to reveal new potential immunocytochemical markers of clinical relevance associated with subtypes of disease280-282 or prognosis.280 However, researchers choosing candidate proteins against which to raise antibodies must recognize that the level of mRNA extracted from a cell suspension (and particularly from a biopsy specimen) does not necessarily correlate with the level of the corresponding protein in the tumor cells. This consideration is relevant to a microarray study of human B-cell lymphoma.282 Figure 9 shows the mRNA expression data from this study for protein markers previously investigated immunohistologically in DLCL. The mRNA patterns for purified cells appear to correlate with protein expression (eg, the absence of B-cell gene expression in T cells and the expression of CD10 and BCL-2 in purified germinal-center cells). However, expression of the proteins studied is known to range in DLCL from less than 40% of cases (CD10) to essentially all cases (class II, CD20, and CD79), and these documented differences in protein expression are not, at least on first inspection, reflected in the mRNA profiles for the corresponding genes, which show little obvious differences.

Fig. 9.

Microarray patterns of mRNA expression for 7 lymphoid-associated genes in normal and neoplastic lymphoid cells.

These data are from a series reported by Alizadeh et al (http://llmpp.nih.gov/lymphoma) that included samples of diffuse large B-cell lymphomas. On the basis of microarray analysis, these tumors were assigned to 2 categories (with different clinical behavior), with one resembling germinal-center cells and the other having an activated B-cell phenotype. The frequency of positive immunostaining for the 7 markers in diffuse large B-cell lymphoma indicated by published studies and the authors' experience is indicated beneath the microarray images. Act B indicates activated B cells; B, B cells; CLL, B-cell chronic lymphocytic lymphoma; Foll Ly, follicular lymphoma; GC, germinal-center cells; and T, T cells.

Fig. 9.

Microarray patterns of mRNA expression for 7 lymphoid-associated genes in normal and neoplastic lymphoid cells.

These data are from a series reported by Alizadeh et al (http://llmpp.nih.gov/lymphoma) that included samples of diffuse large B-cell lymphomas. On the basis of microarray analysis, these tumors were assigned to 2 categories (with different clinical behavior), with one resembling germinal-center cells and the other having an activated B-cell phenotype. The frequency of positive immunostaining for the 7 markers in diffuse large B-cell lymphoma indicated by published studies and the authors' experience is indicated beneath the microarray images. Act B indicates activated B cells; B, B cells; CLL, B-cell chronic lymphocytic lymphoma; Foll Ly, follicular lymphoma; GC, germinal-center cells; and T, T cells.

Close modal

Thus, the process of translating new molecular biologic findings into practical antibody-based techniques for assessing human tumor samples will continue to require a combination of judicious discrimination in choosing clinically important molecules and skill in making and characterizing specific antibodies. Given the large number of genes from which to choose the right candidates and the lotterylike nature of mAb production, an element of luck would also help!

This article is dedicated to the memory of Carlo Falini. The authors also wish to express their gratitude to their collaborators who have contributed with great skill over the years to the production and characterization of many of the reagents referred to in this review.

Supported by Associazione Italiana per la Ricerca sul Cancro (B.F.) and the Leukaemia Research Fund of Great Britain (D.Y.M.).

The authors contributed equally to this work.

1
Rabbitts
TH
Chromosomal translocations in human cancer.
Nature.
372
1994
143
149
2
Look
AT
Oncogenic transcription factors in the human acute leukemias.
Science.
278
1997
1059
1064
3
Chleq-Deschamps
CM
Le Brun
DP
Huie
P
et al
Topographical dissociation of BCL-2 messenger RNA and protein expression in human lymphoid tissues.
Blood.
81
1993
293
298
4
Kondo
E
Nakamura
S
Onoue
H
et al
Detection of bcl-2 protein and bcl-2 messenger RNA in normal and neoplastic lymphoid tissues by immunohistochemistry and in situ hybridization.
Blood.
80
1992
2044
2051
5
Murrell
AM
Bockamp
EO
Gottgens
B
et al
Discordant regulation of SCL/TAL-1 mRNA and protein during erythroid differentiation.
Oncogene.
11
1995
131
139
6
Tsujimoto
Y
Croce
CM
Analysis of the structure, transcripts, and protein products of bcl-2, the gene involved in human follicular lymphoma.
Proc Natl Acad Sci U S A.
83
1986
5214
5218
7
Pezzella
F
Tse
AGD
Cordell
JL
Pulford
KAF
Gatter
KC
Mason
DY
Expression of the bcl-2 oncogene protein is not specific for the 14;18 chromosomal translocation.
Am J Pathol.
137
1990
225
232
8
Fouret
P
du Bois
RM
Bernaudin
JF
Takahashi
H
Ferrans
VJ
Crystal
RG
Expression of the neutrophil elastase gene during human bone marrow cell differentiation.
J Exp Med.
169
1989
833
845
9
Rosenberg
CL
Wong
E
Petty
EM
et al
PRAD1, a candidate BCL1 oncogene: mapping and expression in centrocytic lymphoma.
Proc Natl Acad Sci U S A.
88
1991
9638
9642
10
Banks
PM
Chan
J
Cleary
ML
et al
Mantle cell lymphoma. A proposal for unification of morphologic, immunologic, and molecular data.
Am J Surg Pathol.
16
1992
637
640
11
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.
84
1994
1361
1392
12
Raynaud
SD
Bekri
S
Leroux
D
et al
Expanded range of 11q13 breakpoints with differing patterns of cyclin D1 expression in B-cell malignancies.
Genes Chromosomes Cancer.
8
1993
80
87
13
Jadayel
D
Matutes
E
Dyer
MJ
et al
Splenic lymphoma with villous lymphocytes: analysis of BCL-1 rearrangements and expression of the cyclin D1 gene.
Blood.
83
1994
3664
3671
14
Bartkova
J
Lukas
J
Strauss
M
Bartek
J
Cell cycle-related variation and tissue-restricted expression of human cyclin D1 protein.
J Pathol.
172
1994
237
245
15
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.
145
1994
86
96
16
Baldin
V
Lukas
J
Marcote
MJ
Pagano
M
Draetta
G
Cyclin D1 is a nuclear protein required for cell cycle progression in G1.
Genes Dev.
7
1993
812
821
17
Banno
S
Yoshikawa
K
Nakamura
S
et al
Monoclonal antibody against PRAD1/cyclin D1 stains nuclei of tumor cells with translocation or amplification at BCL-1 locus.
Jpn J Cancer Res.
85
1994
918
926
18
Lukas
J
Pagano
M
Staskova
Z
Draetta
G
Bartek
J
Cyclin D1 protein oscillates and is essential for cell cycle progression in human tumour cell lines.
Oncogene.
9
1994
707
718
19
Chan
JKC
Reliable immunostaining of cyclin D1 is difficult, but the problems can be overcome.
Histopathology.
34
1999
266
268
20
Miller
KD
Munson
P
Isaacson
PG
Optimizing cyclin D1 immunostaining of mantle cell lymphoma.
Histopathology.
34
1999
268
270
21
Korin
HW
Schwartz
MR
Chirala
M
Younes
M
Optimized cyclin D1 immunoperoxidase staining in mantle cell lymphoma.
Appl Immunohistochem Mol Morphol.
8
2000
57
60
22
Seto
M
Yamamoto
K
Iida
S
et al
Gene rearrangement and overexpression of PRAD1 in lymphoid malignancy with t(11;14)(q13;q32) translocation.
Oncogene.
7
1992
1401
1406
23
Majlis
A
Pugh
WC
Rodriguez
MA
Benedict
WF
Cabanillas
F
Mantle cell lymphoma: correlation of clinical outcome and biologic features with three histologic variants.
J Clin Oncol.
15
1997
1664
1671
24
Nakamura
S
Yatabe
Y
Kuroda
H
et al
Immunostaining of PRAD1/cyclin D1 protein as a marker for the diagnosis of mantle cell lymphoma.
Leukemia.
3
1997
536
537
25
Aguilera
NS
Bijwaard
KE
Duncan
B
et al
Differential expression of cyclin D1 in mantle cell lymphoma and other non-Hodgkin's lymphomas.
Am J Pathol.
153
1998
1969
1976
26
Ott
G
Kalla
J
Ott
MM
et al
Blastoid variants of mantle cell lymphoma: frequent bcl-1 rearrangements at the major translocation cluster region and tetraploid chromosome clones.
Blood.
89
1997
1421
1429
27
Ott
MM
Ott
G
Kuse
R
et al
The anaplastic variant of centrocytic lymphoma is marked by frequent rearrangements of the bcl-1 gene and high proliferation indices.
Histopathology.
24
1994
329
334
28
Soslow
RA
Zukerberg
LR
Harris
NL
Warnke
RA
BCL-1 (PRAD-1/cyclin D-1) overexpression distinguishes the blastoid variant of mantle cell lymphoma from B-lineage lymphoblastic lymphoma.
Mod Pathol.
10
1997
810
817
29
Zukerberg
LR
Medeiros
LJ
Ferry
JA
Harris
NL
Diffuse low-grade B-cell lymphomas. Four clinically distinct subtypes defined by a combination of morphologic and immunophenotypic features.
Am J Clin Pathol.
100
1993
373
385
30
Zucca
E
Stein
H
Coiffier
B
European Lymphoma Task Force (ELTF): report of the workshop on mantle cell lymphoma (MCL).
Ann Oncol.
5
1994
507
511
31
Hoyer
JD
Hanson
CA
Fonseca
R
Greipp
PR
Dewald
GW
Kurtin
PJ
The (11;14)(q13;q32) translocation in multiple myeloma. A morphologic and immunohistochemical study.
Am J Clin Pathol.
113
2000
831
837
32
Pruneri
G
Fabris
S
Baldini
L
et al
Immunohistochemical analysis of cyclin D1 shows deregulated expression in multiple myeloma with the t(11;14).
Am J Pathol.
156
2000
1505
1513
33
Bosch
F
Jares
P
Campo
E
et al
PRAD-1/cyclin D1 gene overexpression in chronic lymphoproliferative disorders: a highly specific marker of mantle cell lymphoma.
Blood.
84
1994
2726
2732
34
Miranda
RN
Briggs
RC
Kinney
MC
Veno
PA
Hammer
RD
Cousar
JB
Immunohistochemical detection of cyclin D1 using optimized conditions is highly specific for mantle cell lymphoma and hairy cell leukemia.
Mod Pathol.
13
2000
1308
1314
35
Savilo
E
Campo
E
Mollejo
M
et al
Absence of cyclin D1 protein expression in splenic marginal zone lymphoma.
Mod Pathol.
11
1998
601
606
36
Nakamura
S
Yatabe
Y
Seto
M
Cyclin D1 overexpression in malignant lymphomas.
Pathol Int.
47
1997
421
429
37
Argatoff
LH
Connors
JM
Klasa
RJ
Horsman
DE
Gascoyne
RD
Mantle cell lymphoma: a clinicopathologic study of 80 cases.
Blood.
89
1997
2067
2078
38
Yatabe
Y
Suzuki
R
Tobinai
K
et al
Significance of cyclin D1 overexpression for the diagnosis of mantle cell lymphoma: a clinicopathologic comparison of cyclin D1-positive MCL and cyclin D1-negative MCL-like B-cell lymphoma.
Blood.
95
2000
2253
2261
39
Levy
V
Ugo
V
Delmer
A
et al
Cyclin D1 overexpression allows identification of an aggressive subset of leukemic lymphoproliferative disorder.
Leukemia.
13
1999
1343
1351
40
Wong
KF
Chan
JK
So
JC
Yu
PH
Mantle cell lymphoma in leukemic phase: characterization of its broad cytologic spectrum with emphasis on the importance of distinction from other chronic lymphoproliferative disorders.
Cancer.
86
1999
850
857
41
Yang
E
Korsmeyer
SJ
Molecular thanatopsis: a discourse on the BCL2 family and cell death.
Blood.
88
1996
386
401
42
Hockenbery
DM
Zutter
M
Hickey
W
Nahm
M
Korsmeyer
SJ
BCL2 protein is topographically restricted in tissues characterized by apoptotic cell death.
Proc Natl Acad Sci U S A.
88
1991
6961
6965
43
Hockenbery
D
Nunez
G
Milliman
C
Schreiber
RD
Korsmeyer
SJ
Bcl-2 is an inner mitochondrial membrane protein that blocks programmed cell death.
Nature.
348
1990
334
336
44
Fujii
Y
Okumura
M
Takeuchi
Y
et al
Bcl-2 expression in the thymus and periphery.
Cell Immunol.
155
1994
335
344
45
Krajewski
S
Bodrug
S
Gascoyne
R
Berean
K
Krajewska
M
Reed
JC
Immunohistochemical analysis of Mcl-1 and Bcl-2 proteins in normal and neoplastic lymph nodes.
Am J Pathol.
145
1994
515
525
46
Wang
T
Lasota
J
Hanau
CA
Miettinen
M
Bcl-2 oncoprotein is widespread in lymphoid tissue and lymphomas but its differential expression in benign versus malignant follicles and monocytoid B-cell proliferations is of diagnostic value.
APMIS.
103
1995
655
662
47
Gaulard
P
d'Agay
MF
Peuchmaur
M
et al
Expression of the bcl-2 gene product in follicular lymphoma.
Am J Pathol.
140
1992
1089
1095
48
Pezzella
F
Gatter
KC
Mason
DY
et al
Bcl-2 protein expression in follicular lymphomas in absence of 14;18 translocation [letter].
Lancet.
336
1990
1510
1511
49
Seite
P
Hillion
J
d'Agay
MF
et al
BCL2 gene activation and protein expression in follicular lymphoma: a report on 64 cases.
Leukemia.
7
1993
410
417
50
Wang
J
Raffeld
M
Medeiros
LJ
et al
Follicular center cell lymphoma with the t(14;18) translocation in which the rearranged BCL-2 gene is silent.
Leukemia.
7
1993
1834
1839
51
Villuendas
R
Piris
MA
Orradre
JL
Mollejo
M
Rodriguez
R
Morente
M
Different bcl-2 protein expression in high-grade B-cell lymphomas derived from lymph node or mucosa-associated lymphoid tissue.
Am J Pathol.
139
1991
989
993
52
Swerdlow
SH
Utz
GL
Williams
ME
Bcl-2 protein in centrocytic lymphoma; a paraffin section study.
Leukemia.
7
1993
1456
1458
53
Gala
JL
Vermylen
C
Cornu
G
et al
High expression of bcl-2 is the rule in acute lymphoblastic leukemia, except in Burkitt subtype at presentation, and is not correlated with the prognosis.
Ann Hematol.
69
1994
17
24
54
Tang
SC
Visser
L
Hepperle
B
Hanson
J
Poppema
S
Clinical significance of bcl-2-MBR gene rearrangement and protein expression in diffuse large-cell non-Hodgkin's lymphoma: an analysis of 83 cases.
J Clin Oncol.
12
1994
149
154
55
Porwit-MacDonald
A
Ivory
K
Wilkinson
S
Wheatley
K
Wong
L
Janossy
G
Bcl-2 protein expression in normal human bone marrow precursors and in acute myelogenous leukemia.
Leukemia.
9
1995
1191
1198
56
Schlaifer
D
Krajewski
S
Galoin
S
et al
Immunodetection of apoptosis-regulating proteins in lymphomas from patients with and without human immunodeficiency virus infection.
Am J Pathol.
149
1996
177
185
57
Spina
D
Leoncini
L
Megha
T
et al
Cellular kinetic and phenotypic heterogeneity in and among Burkitt's and Burkitt-like lymphomas.
J Pathol.
182
1997
145
150
58
Rassidakis
GZ
Sarris
AH
Herling
M
et al
Differential expression of bcl-2 family proteins in ALK-positive and ALK-negative anaplastic large cell lymphoma of T/null-cell lineage.
Am J Pathol.
159
2001
527
535
59
Villalva
C
Bougrine
F
Delsol
G
Brousset
P
Bcl-2 expression in anaplastic large cell lymphoma.
Am J Pathol.
158
2001
1889
1890
60
Takano
Y
Saegusa
M
Ikenaga
M
Okayasu
I
Apoptosis and proliferative activity of non-Hodgkin's lymphomas: comparison with expression of bcl-2, p53 and c-myc proteins.
Pathol Int.
47
1997
90
94
61
Gisbertz
IA
Schouten
HC
Bot
FJ
Arends
JW
Proliferation and apoptosis in primary gastric B-cell non-Hodgkin's lymphoma.
Histopathology.
30
1997
152
159
62
Cogliatti
SB
Griesser
H
Peng
H
et al
Significantly different bcl-2 expression profiles in gastric and non-gastric primary extranodal high-grade B-cell lymphomas.
J Pathol.
192
2000
470
478
63
Ashton-Key
M
Biddolph
SC
Stein
H
Gatter
KC
Mason
DY
Heterogeneity of bcl-2 expression in MALT lymphoma.
Histopathology.
26
1995
75
78
64
Navratil
E
Gaulard
P
Kanavaros
P
et al
Expression of the bcl-2 protein in B cell lymphomas arising from mucosa-associated lymphoid tissue.
J Clin Pathol.
48
1995
18
21
65
Bhagat
SK
Medeiros
LJ
Weiss
LM
Wang
J
Raffeld
M
Stetler-Stevenson
M
bcl-2 expression in Hodgkin's disease. Correlation with the t(14;18) translocation and Epstein-Barr virus.
Am J Clin Pathol.
99
1993
604
608
66
Doussis
IA
Pezzella
F
Lane
DP
Gatter
KC
Mason
DY
An immunocytochemical study of p53 and bcl-2 protein expression in Hodgkin's disease.
Am J Clin Pathol.
99
1993
663
667
67
Gupta
RK
Lister
TA
Bodmer
JG
The t(14;18) chromosomal translocation and Bcl-2 protein expression in Hodgkin's disease.
Leukemia.
8
1994
1337
1341
68
Le Brun
DP
Ngan
BY
Weiss
LM
Huie
P
Warnke
RA
Cleary
ML
The bcl-2 oncogene in Hodgkin's disease arising in the setting of follicular non-Hodgkin's lymphoma.
Blood.
83
1994
223
230
69
Algara
P
Martinez
P
Sanchez
L
et al
Lymphocyte predominance Hodgkin's disease (nodular paragranuloma)—a bcl-2 negative germinal centre lymphoma.
Histopathology.
19
1991
69
75
70
Lones
MA
Pinkus
GS
Shintaku
IP
Said
JW
Bcl-2 oncogene protein is preferentially expressed in Reed-Sternberg cells in Hodgkin's disease of the nodular sclerosis subtype.
Am J Clin Pathol.
102
1994
464
467
71
Utz
GL
Swerdlow
SH
Distinction of follicular hyperplasia from follicular lymphoma in B5-fixed tissues: comparison of MT2 and bcl-2 antibodies.
Hum Pathol.
24
1993
1155
1158
72
Schmid
C
Isaacson
PG
Proliferation centres in B-cell malignant lymphoma, lymphocytic (B-CLL): an immunophenotypic study.
Histopathology.
24
1994
445
451
73
Pezzella
F
Gatter
K
What is the value of bcl-2 protein detection for histopathologists?
Histopathology.
26
1995
89
93
74
Straka
C
Mielke
B
Eichelmann
A
Trede
I
Ho
AD
Möller
P
Bcl-2 gene rearrangements in primary B-cell lymphoma of the gastrointestinal tract reveal follicular lymphoma as a subtype.
Leukemia.
7
1993
268
273
75
Ben-Ezra
JM
King
BE
Harris
AC
Todd
WM
Kornstein
MJ
Staining for Bcl-2 protein helps to distinguish benign from malignant lymphoid aggregates in bone marrow biopsies.
Mod Pathol.
7
1994
560
564
76
Chetty
R
Echezarreta
G
Comley
M
Gatter
K
Immunohistochemistry in apparently normal bone marrow trephine specimens from patients with nodal follicular lymphoma.
J Clin Pathol.
48
1995
1035
1038
77
Soslow
RA
Bhargava
V
Warnke
RA
MIC2, TdT, bcl-2, and CD34 expression in paraffin-embedded high-grade lymphoma/acute lymphoblastic leukemia distinguishes between distinct clinicopathologic entities.
Hum Pathol.
28
1997
1158
1165
78
Cory
S
Regulation of lymphocyte survival by the bcl-2 gene family.
Annu Rev Immunol.
13
1995
513
543
79
Reed
JC
Bcl-2 family proteins: regulators of apoptosis and chemoresistance in hematologic malignancies.
Semin Hematol.
34
1997
9
19
80
Hermine
O
Haioun
C
Lepage
E
et al
Prognostic significance of bcl-2 protein expression in aggressive non-Hodgkin's lymphoma. Groupe d'Etude des Lymphomes de l'Adulte (GELA).
Blood.
87
1996
265
272
81
Hill
ME
MacLennan
KA
Cunningham
DC
et al
Prognostic significance of BCL-2 expression and bcl-2 major breakpoint region rearrangement in diffuse large cell non-Hodgkin's lymphoma: a British National Lymphoma Investigation Study.
Blood.
88
1996
1046
1051
82
Kramer
MH
Hermans
J
Parker
J
et al
Clinical significance of bcl2 and p53 protein expression in diffuse large B-cell lymphoma: a population-based study.
J Clin Oncol.
14
1996
2131
2138
83
Gascoyne
RD
Adomat
SA
Krajewski
S
et al
Prognostic significance of Bcl-2 protein expression and Bcl-2 gene rearrangement in diffuse aggressive non-Hodgkin's lymphoma.
Blood.
90
1997
244
251
84
Martinka
M
Comeau
T
Foyle
A
Anderson
D
Greer
W
Prognostic significance of t(14;18) and bcl-2 gene expression in follicular small cleaved cell lymphoma and diffuse large cell lymphoma.
Clin Invest Med.
20
1997
364
370
85
Sanchez
E
Chacon
I
Plaza
MM
et al
Clinical outcome in diffuse large B-cell lymphoma is dependent on the relationship between different cell-cycle regulator proteins.
J Clin Oncol.
16
1998
1931
1939
86
Sarris
A
Ford
R
Recent advances in the molecular pathogenesis of lymphomas.
Curr Opin Oncol.
11
1999
351
363
87
Campos
L
Rouault
JP
Sabido
O
et al
High expression of bcl-2 protein in acute myeloid leukemia cells is associated with poor response to chemotherapy.
Blood.
81
1993
3091
3096
88
Maung
ZT
MacLean
FR
Reid
MM
et al
The relationship between bcl-2 expression and response to chemotherapy in acute leukaemia.
Br J Haematol.
88
1994
105
109
89
Lauria
F
Raspadori
D
Rondelli
D
et al
High bcl-2 expression in acute myeloid leukemia cells correlates with CD34 positivity and complete remission rate.
Leukemia.
11
1997
2075
2078
90
Deng
G
Lane
C
Kornblau
S
et al
Ratio of bcl-xshort to bcl-xlong is different in good- and poor-prognosis subsets of acute myeloid leukemia.
Mol Med.
4
1998
158
164
91
Kornblau
SM
Thall
PF
Estrov
Z
et al
The prognostic impact of BCL2 protein expression in acute myelogenous leukemia varies with cytogenetics.
Clin Cancer Res.
5
1999
1758
1766
92
Brink
AA
Oudejans
JJ
van den Brule
AJ
et al
Low p53 and high bcl-2 expression in Reed-Sternberg cells predicts poor clinical outcome for Hodgkin's disease: involvement of apoptosis resistance?
Mod Pathol.
11
1998
376
383
93
Wendum
D
Sebban
C
Gaulard
P
et al
Follicular large-cell lymphoma treated with intensive chemotherapy: an analysis of 89 cases included in the LNH87 trial and comparison with the outcome of diffuse large B-cell lymphoma. Groupe d'Etude des Lymphomes de l'Adulte.
J Clin Oncol.
15
1997
1654
1663
94
Lopez-Guillermo
A
Cabanillas
F
McDonnell
TI
et al
Correlation of bcl-2 rearrangement with clinical characteristics and outcome in indolent follicular lymphoma.
Blood.
93
1999
3081
3087
95
Robertson
LE
Plunkett
W
McConnell
K
Keating
MJ
McDonnell
TJ
Bcl-2 expression in chronic lymphocytic leukemia and its correlation with the induction of apoptosis and clinical outcome.
Leukemia.
10
1996
456
459
96
Gottardi
D
De Leo
AM
Alfarano
A
et al
Fludarabine ability to down-regulate Bcl-2 gene product in CD5+ leukaemic B cells: in vitro/in vivo correlations.
Br J Haematol.
99
1997
147
157
97
Coustan-Smith
E
Kitanaka
A
Pui
CH
et al
Clinical relevance of BCL-2 overexpression in childhood acute lymphoblastic leukemia.
Blood.
87
1996
1140
1146
98
Uckun
FM
Yang
Z
Sather
H
et al
Cellular expression of antiapoptotic BCL-2 oncoprotein in newly diagnosed childhood acute lymphoblastic leukemia: a Children's Cancer Group Study.
Blood.
89
1997
3769
3777
99
Salomons
G
Smets
L
Verwijs-Janssen
M
et al
Bcl-2 family members in childhood acute lymphoblastic leukemia: relationships with features at presentation, in vitro and in vivo drug response and long-term clinical outcome.
Leukemia.
13
1999
1574
1580
100
Ye
BH
Cattoretti
G
Shen
Q
et al
The BCL-6 proto-oncogene controls germinal-centre formation and Th2-type inflammation.
Nat Genet.
16
1997
161
170
101
Reljic
R
Wagner
SD
Peakman
LJ
Fearon
DT
Suppression of signal transducer and activator of transcription 3-dependent B lymphocyte terminal differentiation by BCL-6.
J Exp Med.
192
2000
1841
1848
102
Ye
BH
Lista
F
Lo Coco
F
et al
Alterations of a zinc finger-encoding gene, BCL-6, in diffuse large-cell lymphoma.
Science.
262
1993
747
750
103
Lo Coco
F
Ye
BH
Lista
F
et al
Rearrangements of the BCL6 gene in diffuse large cell non-Hodgkin's lymphoma.
Blood.
83
1994
1757
1759
104
Capello
D
Vitolo
U
Pasqualucci
L
et al
Distribution and pattern of BCL-6 mutations throughout the spectrum of B-cell neoplasia.
Blood.
95
2000
651
659
105
Pasqualucci
L
Migliazza
A
Fracchiolla
N
et al
BCL-6 mutations in normal germinal center B cells: evidence of somatic hypermutation acting outside Ig loci.
Proc Natl Acad Sci U S A.
95
1998
11816
11821
106
Flenghi
L
Bigerna
B
Fizzotti
M
et al
Monoclonal antibodies PG-B6a and PG-B6p recognize, respectively, a highly conserved and a formol-resistant epitope on the human BCL-6 protein amino-terminal region.
Am J Pathol.
148
1996
1543
1555
107
Cattoretti
G
Chang
CC
Cechova
K
et al
BCL-6 protein is expressed in germinal-center B cells.
Blood.
86
1995
45
53
108
Flenghi
L
Ye
BH
Fizzotti
M
et al
A specific monoclonal antibody (PG-B6) detects expression of the BCL-6 protein in germinal center B cells.
Am J Pathol.
147
1995
405
411
109
Onizuka
T
Moriyama
M
Yamochi
T
et al
BCL-6 gene product, a 92- to 98-kD nuclear phosphoprotein, is highly expressed in germinal center B cells and their neoplastic counterparts.
Blood.
86
1995
28
37
110
Falini
B
Fizzotti
M
Pileri
S
Liso
A
Pasqualucci
L
Flenghi
L
BCL-6 protein expression in normal and neoplastic lymphoid tissues.
Ann Oncol.
8
1997
101
104
111
Ree
HJ
Kadin
ME
Kikuchi
M
Ko
YH
Suzumiya
J
Go
JH
Bcl-6 expression in reactive follicular hyperplasia, follicular lymphoma, and angioimmunoblastic T-cell lymphoma with hyperplastic germinal centers: heterogeneity of intrafollicular T-cells and their altered distribution in the pathogenesis of angioimmunoblastic T-cell lymphoma.
Hum Pathol.
30
1999
403
411
112
Hyjek
E
Chadburn
A
Liu
YF
Cesarman
E
Knowles
DM
BCL-6 protein is expressed in precursor T-cell lymphoblastic lymphoma and in prenatal and postnatal thymus.
Blood.
97
2001
270
276
113
Kanazawa
N
Moriyama
M
Onizuka
T
Sugawara
K
Mori
S
Expression of bcl-6 protein in normal skin and epidermal neoplasms.
Pathol Int.
47
1997
600
607
114
Skinnider
BF
Horsman
DE
Dupuis
B
Gascoyne
RD
Bcl-6 and Bcl-2 protein expression in diffuse large B-cell lymphoma and follicular lymphoma: correlation with 3q27 and 18q21 chromosomal abnormalities.
Hum Pathol.
30
1999
803
808
115
Carbone
A
Gaidano
G
Gloghini
A
et al
BCL-6 protein in AIDS-related non-Hodgkin's lymphomas: inverse relationship with Epstein-Barr virus-encoded latent membrane protein-1 expression.
Am J Pathol.
150
1997
155
165
116
Kuppers
R
Klein
U
Hansmann
ML
Rajewsky
K
Cellular origin of human B-cell lymphomas.
N Engl J Med.
341
1999
1520
1529
117
Carbone
A
Gloghini
A
Gaidano
G
Dalla-Favera
R
Falini
B
BCL-6 protein expression in human peripheral T-cell neoplasms is restricted to CD30+ anaplastic large-cell lymphomas.
Blood.
90
1997
2445
2450
118
Falini
B
Bigerna
B
Pasqualucci
L
et al
Distinctive expression pattern of the BCL-6 protein in nodular lymphocyte predominance Hodgkin's disease.
Blood.
87
1996
465
471
119
Mason
DY
Banks
PM
Chan
J
et al
Nodular lymphocyte predominance Hodgkin's disease. A distinct clinicopathological entity.
Am J Surg Pathol.
18
1994
526
530
120
Dogan
A
Bagdi
E
Munson
P
Isaacson
PG
CD10 and BCL-6 expression in paraffin sections of normal lymphoid tissue and B-cell lymphomas.
Am J Surg Pathol.
24
2000
846
852
121
Raible
MD
Hsi
ED
Alkan
S
Bcl-6 protein expression by follicle center lymphomas. A marker for differentiating follicle center lymphomas from other low-grade lymphoproliferative disorders.
Am J Clin Pathol.
112
1999
101
107
122
Kraus
MD
Haley
J
Lymphocyte predominance Hodgkin's disease: the use of bcl-6 and CD57 in diagnosis and differential diagnosis.
Am J Surg Pathol.
24
2000
1068
1078
123
Iida
S
Rao
PH
Butler
M
et al
Deregulation of MUM1/IRF4 by chromosomal translocation in multiple myeloma.
Nat Genet.
17
1997
226
230
124
Mittrucker
HW
Matsuyama
T
Grossman
A
et al
Requirement for the transcription factor LSIRF/IRF4 for mature B and T lymphocyte function.
Science.
275
1997
540
543
125
Falini
B
Fizzotti
M
Pucciarini
A
et al
A monoclonal antibody (MUM1p) detects expression of the MUM1/IRF4 protein in a subset of germinal center B cells, plasma cells and activated T cells.
Blood.
95
2000
2084
2092
126
Carbone
A
Gloghini
A
Cozzi
MR
et al
Expression of MUM1/IRF4 selectively clusters with primary effusion lymphoma among lymphomatous effusions: implications for disease histogenesis and pathogenesis.
Br J Haematol.
111
2000
247
257
127
Gaidano
G
Carbone
A
MUM1: a step ahead toward the understanding of lymphoma histogenesis [editorial].
Leukemia.
14
2000
563
566
128
Tsuboi
K
Iida
S
Inagaki
H
et al
MUM1/IRF4 expression as a frequent event in mature lymphoid malignancies.
Leukemia.
14
2000
449
456
129
Nutt
SL
Heavey
B
Rolink
AG
Busslinger
M
Commitment to the B-lymphoid lineage depends on the transcription factor Pax5.
Nature.
401
1999
556
562
130
Iida
S
Rao
PH
Nallasivam
P
et al
The t(9;14)(p13;q32) chromosomal translocation associated with lymphoplasmacytoid lymphoma involves the PAX-5 gene.
Blood.
88
1996
4110
4117
131
Krenacs
L
Himmelmann
AW
Quintanilla-Martinez
L
et al
Transcription factor B-cell-specific activator protein (BSAP) is differentially expressed in B cells and in subsets of B-cell lymphomas.
Blood.
92
1998
1308
1316
132
Foss
HD
Reusch
R
Demel
G
et al
Frequent expression of the B-cell-specific activator protein in Reed-Sternberg cells of classical Hodgkin's disease provides further evidence for its B-cell origin.
Blood.
94
1999
3108
3113
133
Begley
CG
Green
AR
The SCL gene: from case report to critical hematopoietic regulator.
Blood.
93
1999
2760
2770
134
Goldfarb
AN
Goueli
S
Mickelson
D
Greenberg
JM
T-cell acute lymphoblastic leukemia—the associated gene SCL/tal codes for a 42-Kd nuclear phosphoprotein.
Blood.
80
1992
2858
2866
135
Visvader
JE
Fujiwara
Y
Orkin
SH
Unsuspected role for the T-cell leukemia protein SCL/tal-1 in vascular development.
Genes Dev.
12
1998
473
479
136
Bernard
O
Guglielmi
P
Jonveaux
P
et al
Two distinct mechanisms for the SCL gene activation in the t(1;14) translocation of T-cell leukemias.
Genes Chromosomes Cancer.
1
1990
194
208
137
Pulford
K
Lecointe
N
Leroy-Viard
K
Jones
M
Mathieu-Mahul
D
Mason
DY
Expression of TAL-1 proteins in human tissues.
Blood.
85
1995
675
684
138
Hsu
SF
Chang
AC
Wang
JK
Cheng
JT
Production and characterization of monoclonal antibodies against Tal1 oncoprotein.
Hybridoma.
17
1998
251
255
139
Green
AR
Salvaris
E
Begley
CG
Erythroid expression of the ‘helix-loop-helix’ gene, SCL.
Oncogene.
6
1991
475
479
140
Kallianpur
AR
Jordan
JE
Brandt
SJ
The SCL/TAL-1 gene is expressed in progenitors of both the hematopoietic and vascular systems during embryogenesis.
Blood.
83
1994
1200
1208
141
Visvader
J
Begley
CG
Adams
JM
Differential expression of the LYL, SCL and E2A helix-loop-helix genes within the hemopoietic system.
Oncogene.
6
1991
187
194
142
Cheng
JT
Hsu
HL
Hwang
LY
Baer
R
Products of the TAL1 oncogene: basic helix-loop-helix proteins phosphorylated at serine residues.
Oncogene.
8
1993
677
683
143
Bash
RO
Hall
S
Timmons
CF
et al
Does activation of the TAL1 gene occur in a majority of patients with T-cell acute lymphoblastic leukemia? A pediatric oncology group study.
Blood.
86
1995
666
676
144
Robb
L
Begley
CG
The helix-loop-helix gene SCL: implicated in T-cell acute lymphoblastic leukaemia and in normal haematopoietic development.
Int J Biochem Cell Biol.
28
1996
609
618
145
Anderegg
B
Horstmann
M
Ernst
M
Kabisch
H
Transcription of tal-1, a putative oncogene playing an important role in childhood T-ALL, can be shown in normal peripheral blood cells by a highly sensitive RT-PCR assay.
Pediatr Hematol Oncol.
14
1997
349
358
146
Delabesse
E
Bernard
M
Meyer
V
et al
TAL1 expression does not occur in the majority of T-ALL blasts.
Br J Haematol.
102
1998
449
457
147
Chetty
R
Pulford
K
Jones
M
et al
An immunohistochemical study of TAL-1 protein expression in leukaemias and lymphomas with a novel monoclonal antibody, 2TL 242.
J Pathol.
178
1996
311
315
148
Bash
RO
Crist
WM
Shuster
JJ
et al
Clinical features and outcome of T-cell acute lymphoblastic leukemia in childhood with respect to alterations at the TAL1 locus: a Pediatric Oncology Group study.
Blood.
81
1993
2110
2117
149
Grignani
F
Fagioli
M
Alcalay
M
et al
Acute promyelocytic leukemia: from genetics to treatment.
Blood.
83
1994
10
25
150
Pearson
M
Carbone
R
Sebastiani
C
et al
PML regulates p53 acetylation and premature senescence induced by oncogenic Ras.
Nature.
406
2000
207
210
151
Grignani
F
de Matteis
S
Nervi
C
et al
Fusion proteins of the retinoic acid receptor-α recruit histone deacetylase in promyelocytic leukaemia.
Nature.
391
1998
815
818
152
Lin
RJ
Nagy
L
Inoue
S
Shao
W
Miller
WH
Jr
Evans
RM
Role of the histone deacetylase complex in acute promyelocytic leukaemia.
Nature.
391
1998
811
814
153
Pandolfi
PP
PML, PLZF and NPM genes in the molecular pathogenesis of acute promyelocytic leukemia.
Haematologica.
81
1996
472
482
154
Flenghi
L
Fagioli
M
Tomassoni
L
et al
Characterization of a new monoclonal antibody (PG-M3) directed against the amino-terminal portion of the PML gene product: immunocytochemical evidence for high expression of PML proteins on activated macrophages, endothelial cells, and epithelia.
Blood.
85
1995
1871
1880
155
Stuurman
N
de Graaf
A
Floore
A
et al
A monoclonal antibody recognizing nuclear matrix-associated nuclear bodies.
J Cell Sci.
101
1992
773
784
156
Koken
MH
Puvion-Dutilleul
F
Guillemin
MC
et al
The t(15;17) translocation alters a nuclear body in a retinoic acid-reversible fashion.
EMBO J.
13
1994
1073
1083
157
Fagioli
M
Alcalay
M
Pandolfi
PP
et al
Alternative splicing of PML transcripts predicts coexpression of several carboxy-terminally different protein isoforms.
Oncogene.
7
1992
1083
1091
158
Koken
MH
Linares-Cruz
G
Quignon
F
et al
The PML growth-suppressor has an altered expression in human oncogenesis.
Oncogene.
10
1995
1315
1324
159
Terris
B
Baldin
V
Dubois
S
et al
PML nuclear bodies are general targets for inflammation and cell proliferation.
Cancer Res.
55
1995
1590
1597
160
Gambacorta
M
Flenghi
L
Fagioli
M
et al
Heterogeneous nuclear expression of the promyelocytic leukemia (PML) protein in normal and neoplastic human tissues.
Am J Pathol.
149
1996
2023
2025
161
Daniel
MT
Koken
M
Romagne
O
et al
PML protein expression in hematopoietic and acute promyelocytic leukemia cells.
Blood.
82
1993
1858
1867
162
Weis
K
Rambaud
S
Lavau
C
et al
Retinoic acid regulates aberrant nuclear localization of PML-RAR α in acute promyelocytic leukemia cells.
Cell.
76
1994
345
356
163
Chang
KS
Fan
YH
Andreeff
M
Liu
J
Mu
ZM
The PML gene encodes a phosphoprotein associated with the nuclear matrix.
Blood.
85
1995
3646
3653
164
Dyck
JA
Maul
GG
Miller
WH
Jr
Chen
JD
Kakizuka
A
Evans
RM
A novel macromolecular structure is a target of the promyelocyte-retinoic acid receptor oncoprotein.
Cell.
76
1994
333
343
165
Hodges
M
Tissot
C
Howe
K
Grimwade
D
Freemont
PS
Structure, organization, and dynamics of promyelocytic leukemia protein nuclear bodies.
Am J Hum Genet.
63
1998
297
304
166
Brasch
K
Ochs
RL
Nuclear bodies (NBs): a newly “rediscovered” organelle.
Exp Cell Res.
202
1992
211
223
167
Ascoli
CA
Maul
GG
Identification of a novel nuclear domain.
J Cell Biol.
112
1991
785
795
168
Zuchner
D
Sternsdorf
T
Szostecki
C
Heathcote
EJ
Cauch-Dudek
K
Will
H
Prevalence, kinetics, and therapeutic modulation of autoantibodies against Sp100 and promyelocytic leukemia protein in a large cohort of patients with primary biliary cirrhosis.
Hepatology.
26
1997
1123
1130
169
Korioth
F
Gieffers
C
Maul
GG
Frey
J
Molecular characterization of NDP52, a novel protein of the nuclear domain 10, which is redistributed upon virus infection and interferon treatment.
J Cell Biol.
130
1995
1
13
170
Bloch
DB
de la Monte
SM
Guigaouri
P
Filippov
A
Bloch
KD
Identification and characterization of a leukocyte-specific component of the nuclear body.
J Biol Chem.
271
1996
29198
29204
171
Boddy
MN
Howe
K
Etkin
LD
Solomon
E
Freemont
PS
PIC 1, a novel ubiquitin-like protein which interacts with the PML component of a multiprotein complex that is disrupted in acute promyelocytic leukaemia.
Oncogene.
13
1996
971
982
172
Desbois
C
Rousset
F
Bantignies
F
Jalinot
P
Exclusion of Int-6 from PML nuclear bodies by binding to the HTLV-1 Tax oncoprotein.
Science.
273
1996
951
953
173
Sternsdorf
T
Jensen
K
Will
H
Evidence for covalent modification of the nuclear dot-associated proteins PML and Sp100 by PIC1/SUMO-1.
J Cell Biol.
139
1997
1621
1634
174
Müller
S
Matunis
MJ
Dejean
A
Conjugation with the ubiquitin-related modifier SUMO-1 regulates the partitioning of PML within the nucleus.
EMBO J.
17
1998
61
70
175
Müller
S
Miller Jr.
WH
Dejean
A
Trivalent antimonials induce degradation of the PML-RAR oncoprotein and reorganization of the promyelocytic leukemia nuclear bodies in acute promyelocytic leukemia NB4 cells.
Blood.
92
1998
4308
4316
176
Dyck
JA
Warrell Jr.
RP
Evans
RM
Miller
WH
Jr
Rapid diagnosis of acute promyelocytic leukemia by immunohistochemical localization of PML/RAR-α protein.
Blood.
86
1995
862
867
177
Falini
B
Flenghi
L
Fagioli
M
et al
Immunocytochemical diagnosis of acute promyelocytic leukemia (M3) with the monoclonal antibody PG-M3 (anti-PML).
Blood.
90
1997
4046
4053
178
O'Connor
SJ
Forsyth
PD
Dalal
S
et al
The rapid diagnosis of acute promyelocytic leukaemia using PML (5E10) monoclonal antibody.
Br J Haematol.
99
1997
597
604
179
Samoszuk
MK
Tynan
W
Sallash
G
Nasr
S
Monczak
Y
Miller
WH
An immunofluorescent assay for acute promyelocytic leukemia cells.
Am J Clin Pathol.
109
1998
205
210
180
Villamor
N
Costa
D
Aymerich
M
et al
Rapid diagnosis of acute promyelocytic leukemia by analyzing the immunocytochemical pattern of the PML protein with the monoclonal antibody PG-M3.
Am J Clin Pathol.
114
2000
786
792
181
Grimwade
D
Gorman
P
Duprez
E
et al
Characterization of cryptic rearrangements and variant translocations in acute promyelocytic leukemia.
Blood.
90
1997
4876
4885
182
Redner
RL
Rush
EA
Schlesinger
KW
Pollock
SL
Watkins
SC
The t(5;17) APL fusion protein NPM-RAR does not alter PML localization [abstract].
Blood
90(suppl 1)
1997
321a
183
Hummel
JL
Wells
RA
Dube
ID
Licht
JD
Kamel-Reid
S
Deregulation of NPM and PLZF in a variant t(5;17) case of acute promyelocytic leukemia.
Oncogene.
18
1999
633
641
184
Grimwade
D
Biondi
A
Mozziconacci
MJ
et al
Characterization of acute promyelocytic leukemia cases lacking the classic t(15;17): results of the European Working Party. Groupe Francais de Cytogenetique Hematologique, Groupe de Francais d'Hematologie Cellulaire, UK Cancer Cytogenetics Group, and BIOMED 1 European Community-Concerted Action “Molecular Cytogenetic Diagnosis in Haematological Malignancies.”
Blood.
96
2000
1297
1308
185
Brunning
RD
McKenna
RW
Tumors of the Bone Marrow.
1994
Armed Forces Institute of Pathology
Washington, DC
Atlas of Tumor Pathology, Fascicle 9.
186
Tallman
MS
Andersen
JW
Schiffer
CA
et al
All-trans-retinoic acid in acute promyelocytic leukemia.
N Engl J Med.
337
1997
1021
1028
187
Lo Coco
F
Diverio
D
Falini
B
Biondi
A
Nervi
C
Pelicci
PG
Genetic diagnosis and molecular monitoring in the management of acute promyelocytic leukemia.
Blood.
94
1999
12
22
188
Shen
ZX
Chen
GQ
Ni
JH
et al
Use of arsenic trioxide (As2O3) in the treatment of acute promyelocytic leukemia (APL): II. Clinical efficacy and pharmacokinetics in relapsed patients.
Blood.
89
1997
3354
3360
189
Licht
JD
Chomienne
C
Goy
A
et al
Clinical and molecular characterization of a rare syndrome of acute promyelocytic leukemia associated with translocation (11;17).
Blood.
85
1995
1083
1094
190
Chen
Z
Brand
NJ
Chen
A
et al
Fusion between a novel Kruppel-like zinc finger gene and the retinoic acid receptor-α locus due to a variant t(11;17) translocation associated with acute promyelocytic leukaemia.
EMBO J.
12
1993
1161
1167
191
Redner
RL
Rush
EA
Faas
S
Rudert
WA
Corey
SJ
The t(5;17) variant of acute promyelocytic leukemia expresses a nucleophosmin-retinoic acid receptor fusion.
Blood.
87
1996
882
886
192
Wells
RA
Catzavelos
C
Kamel-Reid
S
Fusion of retinoic acid receptor α to NuMA, the nuclear mitotic apparatus protein, by a variant translocation in acute promyelocytic leukaemia.
Nat Genet.
17
1997
109
113
193
Licht
JD
Shaknovich
R
English
MA
et al
Reduced and altered DNA-binding and transcriptional properties of the PLZF-retinoic acid receptor-α chimera generated in t(11;17)-associated acute promyelocytic leukemia.
Oncogene.
12
1996
323
336
194
Koken
MH
Reid
A
Quignon
F
et al
Leukemia-associated retinoic acid receptor α fusion partners, PML and PLZF, heterodimerize and colocalize to nuclear bodies.
Proc Natl Acad Sci U S A.
94
1997
10255
10260
195
Ruthardt
M
Orleth
A
Tomassoni
L
et al
The acute promyelocytic leukaemia specific PML and PLZF proteins localize to adjacent and functionally distinct nuclear bodies.
Oncogene.
16
1998
1945
1953
196
Reid
A
Gould
A
Brand
N
et al
Leukemia translocation gene, PLZF, is expressed with a speckled nuclear pattern in early hematopoietic progenitors.
Blood.
86
1995
4544
4552
197
Cook
M
Gould
A
Brand
N
et al
Expression of the zinc-finger gene PLZF at rhombomere boundaries in the vertebrate hindbrain.
Proc Natl Acad Sci U S A.
92
1995
2249
2253
198
Kaneko
Y
Frizzera
G
Edamura
S
et al
A novel translocation, t(2;5)(p23;q35), in childhood phagocytic large T-cell lymphoma mimicking malignant histiocytosis.
Blood.
73
1989
806
813
199
Le Beau
MM
Bitter
MA
Larson
RA
et al
The t(2;5)(p23;q35): a recurring chromosomal abnormality in Ki-1-positive anaplastic large cell lymphoma.
Leukemia.
3
1989
866
870
200
Mason
DY
Bastard
C
Rimokh
R
et al
CD30-positive large cell lymphomas (‘Ki-1 lymphoma’) are associated with a chromosomal translocation involving 5q35.
Br J Haematol.
74
1990
161
168
201
Morris
SW
Kirstein
MN
Valentine
MB
et al
Fusion of a kinase gene, ALK, to a nucleolar protein gene, NPM, in non-Hodgkin's lymphoma.
Science.
263
1994
1281
1284
202
Morris
SW
Naeve
C
Mathew
P
Kirstein
MN
Cui
XL
Witte
DP
ALK, the chromosome 2 gene locus altered by the t(2;5) in non-Hodgkin's lymphoma, encodes a novel neural-specific receptor tyrosine kinase that is highly related to leukocyte tyrosine kinase (LTK).
Oncogene.
14
1997
2175
2188
203
Shiota
M
Fujimoto
J
Semba
T
Satoh
H
Yamamoto
T
Mori
S
Hyperphosphorylation of a novel 80 kDa protein-tyrosine kinase similar to Ltk in a human Ki-1 lymphoma cell line, AMS3.
Oncogene.
9
1994
1567
1574
204
Fujimoto
J
Shiota
M
Iwahara
T
et al
Characterization of the transforming activity of p80, a hyperphosphorylated protein in a Ki-1 lymphoma cell line with chromosomal translocation t(2;5).
Proc Natl Acad Sci U S A.
93
1996
4181
4186
205
Ladanyi
M
The NPM/ALK gene fusion in the pathogenesis of anaplastic large cell lymphoma.
Cancer Surv
30
1997
59
75
206
Drexler
HG
Gignac
SM
von Wasielewski
R
Werner
M
Dirks
WG
Pathobiology of NPM-ALK and variant fusion genes in anaplastic large cell lymphoma and other lymphomas.
Leukemia.
14
2000
1533
1559
207
Colleoni
GW
Bridge
JA
Garicochea
B
Liu
J
Filippa
DA
Ladanyi
M
ATIC-ALK: a novel variant ALK gene fusion in anaplastic large cell lymphoma resulting from the recurrent cryptic chromosomal inversion, inv(2)(p23q35).
Am J Pathol.
156
2000
781
789
208
Lamant
L
Dastugue
N
Pulford
K
Delsol
G
Mariamé
B
A new fusion gene TPM3-ALK in anaplastic large cell lymphoma created by a (1;2) (q25;p23) translocation.
Blood.
93
1999
3088
3095
209
Ma
Z
Cools
J
Marynen
P
et al
Inv(2)(p23q35) in anaplastic large-cell lymphoma induces constitutive anaplastic lymphoma kinase (ALK) tyrosine kinase activation by fusion to ATIC, an enzyme involved in purine nucleotide biosynthesis.
Blood.
95
2000
2144
2149
210
Touriol
C
Greenland
C
Lamant
L
et al
Further demonstration of the diversity of chromosomal changes involving 2p23 in ALK-positive lymphoma: 2 cases expressing ALK kinase fused to CLTCL (clathrin chain polypeptide-like).
Blood.
95
2000
3204
3207
211
Trinei
M
Lanfrancone
L
Campo
E
et al
A new variant anaplastic lymphoma kinase (ALK)-fusion protein (ATIC- ALK) in a case of ALK-positive anaplastic large cell lymphoma.
Cancer Res.
60
2000
793
798
212
Tort
F
Pinyol
M
Pulford
K
et al
Molecular characterization of a new ALK translocation involving moesin (MSN-ALK) in anaplastic large cell lymphoma.
Lab Invest.
81
2001
419
426
213
Meech
SJ
McGavran
L
Odom
LF
et al
Unusual childhood extramedullary hematologic malignancy with natural killer cell properties that contains tropomyosin 4-anaplastic lymphoma kinase gene fusion.
Blood.
98
2001
1209
1216
214
Downing
JR
Shurtleff
SA
Zielenska
M
et al
Molecular detection of the (2;5) translocation of non-Hodgkin's lymphoma by reverse transcriptase-polymerase chain reaction.
Blood.
85
1995
3416
3422
215
Elmberger
PG
Lozano
MD
Weisenburger
DD
Sanger
W
Chan
WC
Transcripts of the npm-alk fusion gene in anaplastic large cell lymphoma, Hodgkin's disease, and reactive lymphoid lesions.
Blood.
86
1995
3517
3521
216
Lopategui
JR
Sun
LH
Chan
JK
et al
Low frequency association of the t(2;5)(p23;q35) chromosomal translocation with CD30+ lymphomas from American and Asian patients. A reverse transcriptase-polymerase chain reaction study.
Am J Pathol.
146
1995
323
328
217
Wellmann
A
Otsuki
T
Vogelbruch
M
Clark
HM
Jaffe
ES
Raffeld
M
Analysis of the t(2;5)(p23;q35) translocation by reverse transcription-polymerase chain reaction in CD30+ anaplastic large-cell lymphomas, in other non-Hodgkin's lymphomas of T-cell phenotype, and in Hodgkin's disease.
Blood.
86
1995
2321
2328
218
DeCoteau
JF
Butmarc
JR
Kinney
MC
Kadin
ME
The t(2;5) chromosomal translocation is not a common feature of primary cutaneous CD30+ lymphoproliferative disorders: comparison with anaplastic large-cell lymphoma of nodal origin.
Blood.
87
1996
3437
3441
219
Lamant
L
Meggetto
F
Al Saati
T
et al
High incidence of the t(2;5)(p23;q35) translocation in anaplastic large cell lymphoma and its lack of detection in Hodgkin's disease. Comparison of cytogenetic analysis, reverse transcriptase-polymerase chain reaction, and P-80 immunostaining.
Blood.
87
1996
284
291
220
Shiota
M
Fujimoto
J
Takenaga
M
et al
Diagnosis of t(2;5)(p23;q35)-associated Ki-1 lymphoma with immunohistochemistry.
Blood.
84
1994
3648
3652
221
Shiota
M
Nakamura
S
Ichinohasama
R
et al
Anaplastic large cell lymphomas expressing the novel chimeric protein p80NPM/ALK: a distinct clinicopathologic entity.
Blood.
86
1995
1954
1960
222
Nakamura
S
Shiota
M
Nakagawa
A
et al
Anaplastic large cell lymphoma: a distinct molecular pathologic entity: a reappraisal with special reference to p80(NPM/ALK) expression.
Am J Surg Pathol.
21
1997
1420
1432
223
Herbst
H
Anagnostopoulos
J
Heinze
B
Durkop
H
Hummel
M
Stein
H
ALK gene products in anaplastic large cell lymphomas and Hodgkin's disease.
Blood.
86
1995
1694
1700
224
Yee
HT
Ponzoni
M
Merson
A
et al
Molecular characterization of the t(2;5)(p23;q35) translocation in anaplastic large cell lymphoma (Ki-1) and Hodgkin's disease.
Blood.
87
1996
1081
1088
225
Bischof
D
Pulford
K
Mason
DY
Morris
SW
Role of the nucleophosmin (NPM) portion of the non-Hodgkin's lymphoma-associated NPM-anaplastic lymphoma kinase fusion protein in oncogenesis.
Mol Cell Biol.
17
1997
2312
2325
226
Hutchison
RE
Banki
K
Shuster
JJ
et al
Use of an anti-ALK antibody in the characterization of anaplastic large-cell lymphoma of childhood.
Ann Oncol.
8(suppl 1)
1997
37
42
227
Pulford
K
Lamant
L
Morris
SW
et al
Detection of anaplastic lymphoma kinase (ALK) and nucleolar protein nucleophosmin (NPM)-ALK protein in normal and neoplastic cells with the monoclonal antibody ALK1.
Blood.
89
1997
1394
1404
228
Falini
B
Bigerna
B
Fizzotti
M
et al
ALK expression defines a distinct group of T/null lymphoma (“ALK lymphoma”) with a wide morphological spectrum.
Am J Pathol.
153
1998
875
886
229
Pileri
SA
Pulford
K
Mori
S
et al
Frequent expression of the NPM-ALK chimeric fusion protein in anaplastic large-cell lymphoma, lympho-histiocytic type.
Am J Pathol.
150
1997
1207
1211
230
Benharroch
D
Meguerian-Bedoyan
Z
Lamant
L
et al
ALK-positive lymphoma: a single disease with a broad spectrum of morphology.
Blood.
91
1998
2076
2084
231
Ochs
R
Lischwe
M
O'Leary
P
Busch
H
Localization of nucleolar phosphoproteins B23 and C23 during mitosis.
Exp Cell Res.
146
1983
139
149
232
Cordell
JL
Pulford
KAF
Bigerna
B
et al
Detection of normal and chimeric nucleophosmin in human cells.
Blood.
93
1999
632
642
233
Trumper
L
Pfreundschuh
M
Bonib
FV
Daus
H
Detection of the t(2;5)-associated NPM/ALK fusion cDNA in peripheral blood cells of healthy individuals.
Br J Haematol.
103
1998
1138
1144
234
Maes
B
Vanhentenrijk
V
Wlodarska
I
et al
The NPM-ALK and the ATIC-ALK fusion genes can be detected in non-neoplastic cells.
Am J Pathol.
158
2001
2185
2193
235
Lamant
L
Pulford
K
Bischof
D
et al
Expression of the ALK tyrosine kinase gene in neuroblastoma.
Am J Pathol.
156
2000
1711
1721
236
Stein
H
Foss
HD
Durkop
H
et al
CD30+ anaplastic large cell lymphoma: a review of its histopathologic, genetic, and clinical features.
Blood.
96
2000
3681
3695
237
Falini
B
Anaplastic large cell lymphoma: pathological, molecular and clinical features.
Br J Haematol.
114
2001
1
21
238
Falini
B
Pulford
K
Pucciarini
A
et al
Lymphomas expressing ALK fusion protein(s) other than NPM-ALK.
Blood.
94
1999
3509
3515
239
Pulford
K
Falini
B
Cordell
J
et al
Biochemical detection of novel ALK proteins in tissue sections of anaplastic large cell lymphoma.
Am J Pathol.
154
1999
1657
1663
240
Chan
JK
Anaplastic large cell lymphoma: redefining its morphologic spectrum and importance of recognition of the ALK-positive subset.
Adv Anat Pathol.
5
1998
281
313
241
Kinney
MC
Kadin
ME
The pathologic and clinical spectrum of anaplastic large cell lymphoma and correlation with ALK gene dysregulation.
Am J Clin Pathol
111
1 suppl 1
1999
S56
6
242
Pittaluga
S
Wiodarska
I
Pulford
K
et al
The monoclonal antibody ALK1 identifies a distinct morphological subtype of anaplastic large cell lymphoma associated with 2p23/ALK rearrangements.
Am J Pathol.
151
1997
343
351
243
Chan
JK
Buchanan
R
Fletcher
CD
Sarcomatoid variant of anaplastic large-cell Ki-1 lymphoma.
Am J Surg Pathol.
14
1990
983
988
244
Pileri
S
Falini
B
Delsol
G
et al
Lymphohistiocytic T-cell lymphoma (anaplastic large cell lymphoma CD30+/Ki-1+ with a high content of reactive histiocytes).
Histopathology.
16
1990
383
391
245
Kadin
ME
Anaplastic large cell lymphoma and its morphological variants.
Cancer Surv.
30
1997
77
86
246
Li
G
Salhany
KE
Rook
AH
Lessin
SR
The pathogenesis of large cell transformation in cutaneous T-cell lymphoma is not associated with t(2;5)(p23;q35) chromosomal translocation.
J Cutan Pathol.
24
1997
403
408
247
Kinney
MC
Collins
RD
Greer
JP
Whitlock
JA
Sioutos
N
Kadin
ME
A small-cell-predominant variant of primary Ki-1 (CD30)+ T-cell lymphoma.
Am J Surg Pathol.
17
1993
859
868
248
Bayle
C
Charpentier
A
Duchayne
E
et al
Leukaemic presentation of small cell variant anaplastic large cell lymphoma: report of four cases.
Br J Haematol.
104
1999
680
688
249
Sadahira
Y
Hata
S
Sugihara
T
Manabe
T
Bone marrow involvement in NPM-ALK-positive lymphoma: report of two cases.
Pathol Res Pract.
195
1999
657
661
250
Krenacs
L
Wellmann
A
Sorbara
L
et al
Cytotoxic cell antigen expression in anaplastic large cell lymphomas of T- and null-cell type and Hodgkin's disease: evidence for distinct cellular origin.
Blood.
89
1997
980
989
251
Haralambieva
E
Pulford
KA
Lamant
L
et al
Anaplastic large-cell lymphomas of B-cell phenotype are anaplastic lymphoma kinase (ALK) negative and belong to the spectrum of diffuse large B-cell lymphomas.
Br J Haematol.
109
2000
584
591
252
Herbst
H
Sander
C
Tronnier
M
Kutzner
H
Hugel
H
Kaudewitz
P
Absence of anaplastic lymphoma kinase (ALK) and Epstein-Barr virus gene products in primary cutaneous anaplastic large cell lymphoma and lymphomatoid papulosis.
Br J Dermatol.
137
1997
680
686
253
Falini
B
Pileri
S
Zinzani
PL
et al
ALK+ lymphoma: clinico-pathological findings and outcome.
Blood.
93
1999
2697
2706
254
Gascoyne
RD
Aoun
P
Wu
D
et al
Prognostic significance of anaplastic lymphoma kinase (ALK) protein expression in adults with anaplastic large cell lymphoma.
Blood.
93
1999
3913
3921
255
Leoncini
L
Lazzi
S
Scano
D
et al
Expression of the ALK protein by anaplastic large-cell lymphomas correlates with high proliferative activity.
Int J Cancer.
86
2000
777
781
256
Pulford
K
Falini
B
Banham
AH
et al
Immune response to the ALK oncogenic tyrosine kinase in patients with anaplastic large-cell lymphoma.
Blood.
96
2000
1605
1607
257
Shiota
M
Mori
S
The clinicopathological features of anaplastic large cell lymphomas expressing p80NPM/ALK.
Leuk Lymphoma.
23
1996
25
32
258
Delsol
G
Lamant
L
Mariamé
B
et al
A new subtype of large B-cell lymphoma expressing the ALK kinase and lacking the 2;5 translocation.
Blood.
89
1997
1483
1490
259
Ladanyi
M
Aberrant ALK tyrosine kinase signaling. Different cellular lineages, common oncogenic mechanisms.
Am J Pathol.
157
2000
341
345
260
Chan
JK
Cheuk
W
Shimizu
M
Anaplastic lymphoma kinase expression in inflammatory pseudotumors.
Am J Surg Pathol.
25
2001
761
768
261
Lawrence
B
Perez-Atayde
A
Hibbard
MK
et al
TPM3-ALK and TPM4-ALK oncogenes in inflammatory myofibroblastic tumors.
Am J Pathol.
157
2000
377
384
262
Yoneda-Kato
N
Look
AT
Kirstein
MN
et al
The t(3;5)(q25.1;q34) of myelodysplastic syndrome and acute myeloid leukemia produces a novel fusion gene, NPM-MLF1.
Oncogene.
12
1996
265
275
263
Guo
JQ
Lian
JY
Xian
YM
et al
BCR-ABL protein expression in peripheral blood cells of chronic myelogenous leukemia patients undergoing therapy.
Blood.
83
1994
3629
3637
264
Guo
JQ
Lian
J
Glassman
A
et al
Comparison of bcr-abl protein expression and Philadelphia chromosome analyses in chronic myelogenous leukemia patients.
Am J Clin Pathol.
106
1996
442
448
265
Sacchi
N
Schiaffonati
L
Magnani
I
et al
Detection and subcellular localization of an AML1 chimeric protein in the t(8;21) positive acute myeloid leukemia.
Oncogene.
12
1996
437
444
266
van Denderen
J
Hermans
A
Meeuwsen
T
et al
Antibody recognition of the tumor-specific bcr-abl joining region in chronic myeloid leukemia.
J Exp Med.
169
1989
87
98
267
van Denderen
J
ten Hacken
P
Berendes
P
et al
Recognition of the ALL-specific BCR-ABL junction in P190bcr-abl by monoclonal antibody ER-FP1.
Leukemia.
8
1994
1503
1509
268
Sang
B-C
Shi
L
Dias
P
et al
Monoclonal antibodies specific to the acute lymphoblastic leukemia t(1;19)-associated E2A/pbx1 chimeric protein: characterization and diagnostic utility.
Blood.
89
1997
2909
2914
269
Bocchia
M
Korontsvit
T
Xu
Q
et al
Specific human cellular immunity to bcr-abl oncogene-derived peptides.
Blood.
87
1996
3587
3592
270
Buzyn
A
Ostankovitch
M
Zerbib
A
et al
Peptides derived from the whole sequence of BCR-ABL bind to several class I molecules allowing specific induction of human cytotoxic T lymphocytes.
Eur J Immunol.
27
1997
2066
2072
271
Bosch
GJ
Joosten
AM
Kessler
JH
Melief
CJ
Leeksma
OC
Recognition of BCR-ABL positive leukemic blasts by human CD4+ T cells elicited by primary in vitro immunization with a BCR-ABL breakpoint peptide.
Blood.
88
1996
3522
3527
272
Pawelec
G
Max
H
Halder
T
et al
BCR/ABL leukemia oncogene fusion peptides selectively bind to certain HLA-DR alleles and can be recognized by T cells found at low frequency in the repertoire of normal donors.
Blood.
88
1996
2118
2124
273
Papadopoulos
KP
Suciu-Foca
N
Hesdorffer
CS
Tugulea
S
Maffei
A
Harris
PE
Naturally processed tissue- and differentiation stage-specific autologous peptides bound by HLA class I and II molecules of chronic myeloid leukemia blasts.
Blood.
90
1997
4938
4946
274
Jaffray
E
Wood
KM
Hay
RT
Domain organization of I κ B α and sites of interaction with NF-κ B p65.
Mol Cell Biol.
15
1995
2166
2172
275
Holt
SJ
Alexander
P
Inman
CB
Davies
DE
Epidermal growth factor induced tyrosine phosphorylation of nuclear proteins associated with translocation of epidermal growth factor receptor into the nucleus.
Biochem Pharmacol.
47
1994
117
126
276
Holt
SJ
Alexander
P
Inman
CB
Davies
DE
Ligand-induced translocation of epidermal growth factor receptor to the nucleus of NR6/HER fibroblasts is serum dependent.
Exp Cell Res.
217
1995
554
558
277
Mason
DY
Pulford
KAF
Bischof
D
et al
Nucleolar localization of the nucleophosmin-anaplastic lymphoma kinase is not required for malignant transformation.
Cancer Res.
58
1998
1057
1062
278
Grignani
F
Testa
U
Rogaia
D
et al
Effects on differentiation by the promyelocytic leukemia PML/RARα protein depend on the fusion of the PML protein dimerization and RARα DNA binding domains.
EMBO J.
15
1996
4949
4958
279
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.
93
1999
3601
3609
280
Alon
U
Barkai
N
Notterman
DA
et al
Broad patterns of gene expression revealed by clustering analysis of tumor and normal colon tissues probed by oligonucleotide arrays.
Proc Natl Acad Sci U S A.
96
1999
6745
6750
281
Golub
TR
Slonim
DK
Tamayo
P
et al
Molecular classification of cancer: class discovery and class prediction by gene expression monitoring.
Science.
286
1999
531
537
282
Alizadeh
AA
Eisen
MB
Davis
RE
et al
Distinct types of diffuse large B-cell lymphoma identified by gene expression profiling.
Nature.
403
2000
503
511
283
Mitani
S
Sugawara
I
Shiku
H
Mori
S
Expression of c-myc oncogene product and ras family oncogene products in various human malignant lymphomas defined by immunohistochemical techniques.
Cancer.
62
1988
2085
2093
284
Spencer
CA
Groudine
M
Control of c-myc regulation in normal and neoplastic cells.
Adv Cancer Res.
56
1991
1
48
285
Lens
D
Matutes
E
Farahat
N
Morilla
R
Catovsky
D
Differential expression of c-myc protein in B and T lymphocytes.
Leukemia.
8
1994
2102
2110
286
Kittas
C
Korkolopoulou
P
Pangalis
GA
Tsenga
A
Boussiotis
VA
Spandidos
DA
Expression of c-myc p62 protein in non-Hodgkins lymphomas.
Leuk Lymphoma.
1
1990
241
247
287
van Etten
RA
Jackson
P
Baltimore
D
The mouse type IV c-abl gene product is a nuclear protein, and activation of transforming ability is associated with cytoplasmic localization.
Cell.
58
1989
669
678
288
Dhut
S
Chaplin
T
Young
BD
Normal c-abl gene protein—a nuclear component.
Oncogene.
6
1991
1459
1464
289
Wetzler
M
Talpaz
M
van Etten
RA
Hirsh
GC
Beran
M
Kurzrock
R
Subcellular localization of Bcr, Abl, and Bcr-Abl proteins in normal and leukemic cells and correlation of expression with myeloid differentiation.
J Clin Invest.
92
1993
1925
1939
290
Dhut
S
Dorey
EL
Horton
MA
Ganesan
TS
Young
BD
Identification of two normal bcr gene products in the cytoplasm.
Oncogene.
3
1988
561
566
291
Bedi
A
Zehnbauer
BA
Collector
MI
et al
BCR-ABL gene rearrangement and expression of primitive hematopoietic progenitors in chronic myeloid leukemia.
Blood.
81
1993
2898
2902
292
Butler
LM
Slany
R
Cui
X
Cleary
ML
Mason
DY
The HRX proto-oncogene product is widely expressed in human tissues and localizes to nuclear structures.
Blood.
89
1997
3361
3370
293
Berendes
P
Hoogeveen
A
van Dijk
M
van Denderen
J
van Ewijk
W
Specific immunologic recognition of the tumor-specific E2A-PBX1 fusion-point antigen in t(1;19)-positive pre-B cells.
Leukemia.
9
1995
1321
1327
294
de Boer
CJ
Schuuring
E
Dreef
E
et al
Cyclin D1 protein analysis in the diagnosis of mantle cell lymphoma.
Blood.
86
1995
2715
2723
295
Zukerberg
LR
Yang
WI
Arnold
A
Harris
NL
Cyclin D1 expression in non-Hodgkin's lymphomas. Detection by immunohistochemistry.
Am J Clin Pathol.
103
1995
756
760
296
Ennas
MG
Sorio
C
Greim
R
et al
The human ALL-1/MLL/HRX antigen is predominantly localized in the nucleus of resting and proliferating peripheral blood mononuclear cells.
Cancer Res.
57
1997
2035
2041
297
Birchenall-Roberts
MC
Ruscetti
FW
Kasper
JJ
et al
Nuclear localization of v-Abl leads to complex formation with cyclic AMP response element (CRE)-binding protein and transactivation through CRE motifs.
Mol Cell Biol.
15
1995
6088
6099
298
Le Brun
DP
Matthews
BP
Feldman
BJ
Cleary
ML
The chimeric oncoproteins E2A-PBX1 and E2A-HLF are concentrated within spherical nuclear domains.
Oncogene.
15
1997
2059
2067
299
Fornerod
M
Boer
J
van Baal
S
et al
Relocation of the carboxyterminal part of CAN from the nuclear envelope to the nucleus as a result of leukemia-specific chromosome rearrangements.
Oncogene.
10
1995
739
748

Author notes

Brunangelo Falini, Istituto di Ematologia, Policlinico, Monteluce, 06100 Perugia, Italy.

Sign in via your Institution