Your activity: 84 p.v.
your limit has been reached. plz Donate us to allow your ip full access, Email: sshnevis@outlook.com

Clinical manifestations, pathologic features, and diagnosis of mantle cell lymphoma

Clinical manifestations, pathologic features, and diagnosis of mantle cell lymphoma
Authors:
Arnold S Freedman, MD
Jon C Aster, MD, PhD
Section Editor:
Andrew Lister, MD, FRCP, FRCPath, FRCR
Deputy Editor:
Alan G Rosmarin, MD
Literature review current through: Dec 2022. | This topic last updated: Aug 04, 2022.

INTRODUCTION — Mantle cell lymphoma (MCL) is a mature B cell non-Hodgkin lymphoma with a variable clinical course that can involve lymph nodes, extranodal sites, and/or blood [1,2]. MCL is usually composed of small- to medium-sized lymphoid cells that typically express BCL2, CD5, and nuclear cyclin D1. Nearly all cases of MCL have the t(11;14) chromosomal translocation, which involves rearrangement of an immunoglobulin heavy chain and CCND1 (the gene that encodes cyclin D1), and overexpression of cyclin D1. Patients generally present with advanced stage disease, but disease progression is highly variable.

The epidemiology, clinical presentation, pathology, diagnosis, and prognosis of MCL are discussed in this topic.

The pathobiology and treatment of MCL are presented separately. (See "Pathobiology of mantle cell lymphoma" and "Initial treatment of mantle cell lymphoma".)

EPIDEMIOLOGY — MCL comprises about 7 percent of adult non-Hodgkin lymphomas in the United States and Europe with an incidence of approximately 4 to 8 cases per million persons per year [3-8]. Incidence increases with age and appears to be increasing overall in the United States [9]. Approximately three-quarters of patients are male, and White individuals are affected almost twice as frequently as Black individuals. Median age at diagnosis is 68 years.

PATHOGENESIS — MCL is thought to have two distinct cellular origins, each giving rise to different forms of the disease [10]:

Classic MCL typically involves lymph nodes and extranodal sites, such as the gastrointestinal tract. Classic MCL is believed to arise from naïve B cells that "mis-express" SOX11, which is not expressed in normal B cells. SOX11 has been reported to block B cell differentiation, suggesting that it has a direct role in MCL pathogenesis [11].

The "leukemic" variant of MCL mainly involves the peripheral blood, bone marrow, and/or spleen and often spares lymph nodes. This variant develops from antigen-experienced SOX11-negative B cells and is often clinically indolent, but may acquire secondary abnormalities (eg, TP53 mutations) that lead to a very aggressive course.

Both types of MCL are highly associated with a (11;14) translocation that dysregulates the cyclin D1 gene (CCND1). Acquisition of additional genetic abnormalities, such as translocations involving MYC, can lead to progression to more aggressive forms of MCL with blastoid or pleomorphic morphologies.

Additional details regarding the pathobiology of MCL are discussed separately. (See "Pathobiology of mantle cell lymphoma".)

CLINICAL FEATURES — Most patients with MCL have advanced stage disease at diagnosis (70 percent). Approximately 75 percent of patients initially present with lymphadenopathy, with extranodal disease being the primary presentation in the remaining 25 percent [12]. Common sites of involvement include lymph nodes, spleen (45 to 60 percent), Waldeyer's ring, bone marrow (>60 percent), blood (13 to 77 percent), and extranodal sites, such as the gastrointestinal (GI) tract, breast, pleura, and orbit [12-14].

Up to one-third of patients have systemic B symptoms, such as fever, night sweats, and unintentional weight loss, at presentation (table 1). The definition of systemic B symptoms is presented in more detail separately. (See "Clinical presentation and initial evaluation of non-Hodgkin lymphoma".)

MCL can involve any region of the GI tract, occasionally presenting as lymphomatous intestinal polyposis (picture 1) [15,16]. A prospective clinicopathologic study of 31 cases of GI tract involvement found the following distribution of disease: stomach (57 percent), duodenum (52 percent), jejunum/ileum (87 percent), colon (90 percent), and rectum (69 percent) [17]. Lymphomatous submucosal nodules producing polypoid lesions were found in both the small bowel and colon in 28 of 31 cases. (See "Epidemiology, clinical features, and types of small bowel neoplasms", section on 'Primary gastrointestinal tract lymphoma'.)

SOX11-negative forms of MCL often spare lymph nodes and instead have leukemic presentations. Splenomegaly in the absence of lymphadenopathy is common in such variants [18]. Central nervous system involvement is rare overall (<5 percent of cases) but is more common in patients with the leukemic variant [19-21].

MORPHOLOGY — The histologic pattern of nodal MCL growth may be diffuse, nodular, mantle zone, or a combination of the three. Most cases are composed exclusively of small to medium-sized lymphoid cells, with slightly irregular or "notched" nuclei and inconspicuous nucleoli (picture 2). However, the morphology can range from small, more irregular lymphocytes (mimicking the centrocytes or small cleaved cells of follicular lymphoma), to lymphoblast-like cells (in the blastoid variant, raising the question of lymphoblastic lymphoma), and even occasionally to mixtures of small and large cells or markedly atypical large cells (in the pleomorphic variant) (picture 3 and picture 4 and picture 5) [4,22-25]. Despite the small size and bland appearance of the cells in most cases, there is often more mitotic activity than in other histologically low-grade lymphomas. When malignant effusions are present, the cytologic features of the tumor cells in the effusion are similar to those seen with peripheral blood involvement. Single epithelioid macrophages are often interspersed among the tumor cells in tissues, but clusters of macrophages and granulomas are not seen. Transformed tumor cells with basophilic cytoplasm (centroblast- or immunoblast-like cells) are rare or absent from typical cases.

IMMUNOPHENOTYPE — MCL cells express high levels of surface IgM and IgD and, for unknown reasons, show marked skewing towards lambda light chain expression, which is seen in up to 80 percent of cases. They also express pan-B cell antigens (eg, CD19, CD20), CD5, and FMC7. Rare cases may be CD5– or CD23+ [26-29]. A prominent, expanded irregular meshwork of follicular dendritic cells (FDCs) is found even in diffuse cases [22,30,31]. SOX11, a member of the SRY-related HMG family of transcription factors, is a useful marker for MCL, particularly in rare cases in which cyclin D1 expression is not detected [32-34]. (See 'Cyclin D1' below.)

When MCL involves the gastrointestinal tract (lymphomatous polyposis), the tumor cells express the adhesion molecule alpha-4/beta-7 integrin (CD49d), which is normally involved in lymphocyte homing to the high endothelial venules in the gut-associated lymphoid tissues [35,36]. (See "Treatment of extranodal marginal zone lymphoma of mucosa associated lymphoid tissue (MALT lymphoma)".)

Cyclin D1 — Nuclear staining for cyclin D1 (BCL1) is present in 95 percent of cases [37-40], including those that are CD5 negative [41]. The product of the cyclin D1 gene can be detected in the nuclei of neoplastic mantle cells in paraffin-embedded tissue sections with the immunoperoxidase technique, and is useful in distinguishing MCL from other relatively indolent B cell lymphomas, such as chronic lymphocytic leukemia/small lymphocytic lymphoma (CLL/SLL), follicular lymphoma, lymphoplasmacytic lymphoma, and splenic marginal zone lymphoma [37,38,42].

Cyclin D1 may be overproduced even in cases lacking the t(11;14) (see 'Genetic features' below), suggesting that other types of acquired genetic aberrations (eg, point mutations) may also result in increased expression [39,43-45]. Conversely, in rare instances, cyclin D1 staining is absent in MCLs that have the t(11;14) and high levels of CCND1 expression because of concomitant mutations involving the coding sequence of CCND1 that prevent recognition of cyclin D1 by commonly used antibodies [46]. As is typical of molecular lesions identified in cancer, mis-expression of this single gene is insufficient to cause MCL, as transgenic mice that overexpress cyclin D1 alone do not develop lymphoma [47].

The relationship of cyclin D1 positive MCL and cases resembling MCL that are cyclin D1 negative is unclear. In a study of 151 Japanese patients with MCL morphology, tumors that overexpressed cyclin D1 tended to be composed of larger cells with a higher mitotic index [42]. Also, patients with cyclin D1 positive tumors tended to be older, have more involvement of the gastrointestinal tract, less involvement of the orbit, a higher International Prognostic Index (table 2), and a lower five-year survival (30 versus 86 percent).

On the other hand, a separate study from the United States showed that the expression profiles of cyclin D1 positive and negative "MCL" were very similar, and that tumors that failed to express cyclin D1 instead overexpressed either cyclin D2 or D3 [48], which are highly homologous and functionally identical to cyclin D1. In this well characterized group of cyclin D1 negative and positive tumors, no difference in clinical behavior or outcome was observed. (See "Pathobiology of mantle cell lymphoma", section on 'Cell cycle progression'.)

p53 — Mutations of TP53, which encodes the tumor suppressor p53, are more often found in MCL with blastoid morphology, highly proliferative tumors, and relapsed MCL, and are associated with an adverse prognosis [49-51]. While mutated TP53 cannot be identified based on morphology or proliferative rate, missense mutations in p53 often lead to its accumulation in tumor cells; thus, positive immunohistochemical staining for p53 in routine formalin-fixed paraffin-embedded sections is highly correlated with the presence of TP53 mutations [50].

One study that performed both IHC for p53 and sequencing of TP53 reported that IHC was 82 percent sensitive and 100 percent specific in 11 paired samples; TP53 aberrations were found in 21 percent of 255 patients with MCL and this was associated with a three-fold increased risk of death [52].

GENETIC FEATURES — Immunoglobulin (Ig) heavy and light chain genes are rearranged. The Ig V region genes lack somatic mutations in most cases [53,54], indicating a pre-germinal center stage of differentiation, consistent with an origin from immunologically naïve mantle zone B cells [55].

Overexpression of cyclin D1 in MCL is strongly associated with the t(11;14)(q13;q32), a translocation between the CCND1 locus and the immunoglobulin heavy chain (IgH) locus [56-60]. The t(11;14) is not specific for MCL, as it occurs in a subset of multiple myelomas [61] and rarely in other lymphoid malignancies. This translocation leads to the dysregulated expression of CCND1, the gene that encodes cyclin D1, which is involved in the control of the G1 phase of the cell cycle and is not normally expressed in lymphoid cells [37,62,63]. (See 'Cyclin D1' above and "Pathobiology of mantle cell lymphoma", section on 'Cell cycle progression'.)

Karyotyping of metaphase chromosomes reveals the t(11;14) in only 50 to 65 percent of MCLs, but fluorescence in situ hybridization (FISH) or polymerase chain reaction (PCR) analyses of cases with cyclin D1 overexpression almost always detect CCND1/IgH fusion genes (picture 6) [64-66]. In addition, DNA sequence analysis of MCL has identified frequent mutations involving exon 1 of the CCND1 gene [67]; these are of uncertain functional importance, but translation of the CCND1 mRNA is regulated through its 5' untranslated region, suggesting that some of these mutations may enhance CCND1 expression. (See 'Cyclin D1' above.)

Other genes associated with the cell cycle may also be involved by genomic aberrations, including rearrangements of CCND2 (cyclin D2) in cyclin D1 negative cases, mutations of the cyclin-dependent kinase (CDK) inhibitors, and p16 and p17 (particularly in blastoid variants). Decreased expression of the CDK inhibitor p27 and disturbances of pathways associated with apoptosis [34,68-70] have also been reported.

In one study of 60 cases of the blastoid variant, 80 percent had at least one cytogenetic abnormality in addition to t(11;14) [71]. Other cases have been reported to have a high incidence of tetraploidy and TP53 gene mutations [72-74]. Patients with TP53 mutation or deletion have poor clinical outcomes, including shortened overall survival, following intensive chemotherapy [75].

Acquisition of a translocation involving the oncogene MYC has been associated with shorter survival [76,77]. Alterations in TP53, p16, p18, p21, and p27 may also play a role in the development and evolution of MCL [78]. Additional chromosomal abnormalities have been reported [79,80]. In one study, genomic loss of 8p occurred in 11 and 79 percent of patients with nodal or leukemic disease, respectively [81].

Comparative genomic hybridization studies have uncovered several recurrent copy number changes in MCL [65,82]. In one of these, for example, at least three regions of copy number change were found in each of 30 patients, involving at least two chromosomes [65]. The most common findings were gains in 3q and 6p and losses in 13q. The presence of more than five chromosomal aberrations (relative risk [RR]: 33), gain of Xq (RR: 4.5), and loss of 17p (RR: 4.8) were all significantly associated with a worse prognosis.

Gene expression profiling, comparative genomic hybridization, proteomics, and deep sequencing of MCL genomes may shed additional light on the biology and clinical heterogeneity of MCL [48,67,83-88]. Other studies have identified activating NOTCH1 mutations in a minority of MCL cases, a finding that may predict a worse clinical outcome [67,89]. (See "Pathobiology of mantle cell lymphoma".)

DIAGNOSIS — Patients suspected of having MCL should undergo tissue biopsy. In addition to routine histology and immunohistochemistry, involvement of cyclin D1 should be evaluated by immunohistochemistry. Cytogenetic detection of the t(11;14) by either karyotyping or fluorescence in situ hybridization (FISH) is a useful adjunct test.

On histologic review, tumor cells are usually monomorphous small to medium-sized B lymphocytes with irregular nuclei. The degree of irregularity is usually, but not always, less than that of the centrocytes found in germinal centers and follicular lymphoma. Large cells resembling centroblasts or immunoblasts are absent. Tumor cells are typically CD5+ and CD23–; the vast majority overexpress cyclin D1 [56]. The t(11;14) translocation, which is not specific for MCL, is seen in a little over half of cases by conventional cytogenetics, but in a much higher percentage of cases screened with FISH. SOX11 staining is helpful in rare cases that fail to express cyclin D1.

DIFFERENTIAL DIAGNOSIS — The differential diagnosis for MCL includes other non-Hodgkin lymphomas composed of small to medium-sized cells, most notably chronic lymphocytic leukemia (CLL)/small lymphocytic lymphoma (SLL), follicular lymphoma, marginal zone lymphoma, and lymphoblastic lymphoma.

In situ mantle cell neoplasia — The 2016 revisions of the World Health Organization (WHO) classification incorporated a new diagnostic category, "in situ mantle cell neoplasia" [90], which was previously termed in situ mantle cell lymphoma [91]. In situ mantle cell neoplasia is characterized by the presence of cyclin D1 positive cells, usually located in the inner mantle zones of follicles, in lymph nodes that otherwise lack diagnostic features of MCL. In situ mantle cell neoplasia is often found incidentally, sometimes in association with other B cell lymphomas. Some patients with this finding will prove to have disseminated MCL on further work-up; the "in situ" component in such cases presumably represents early seeding of systemic MCL to a partially involved node. Other patients, however, will not have evidence of MCL on further evaluation [92]. The natural history of this second group of patients is not yet known.

Chronic lymphocytic leukemia — Both MCL and CLL are neoplasms of small to medium-sized lymphoid cells with similar immunophenotypes. While CLL is positive for CD20, CD5, and CD23, MCL is positive for CD20 and CD5 but negative for CD23. Immunohistochemistry for cyclin D1 is very helpful in excluding CLL. Other discriminating markers include SOX11 (typically positive in MCL) and LEF1 (frequently positive in CLL). Additional testing to identify the t(11;14) by fluorescence in situ hybridization (FISH) can also aid in the diagnosis of MCL [93]. (See "Clinical features and diagnosis of chronic lymphocytic leukemia/small lymphocytic lymphoma".)

Follicular lymphoma — On histology, MCL can have a predominantly nodular growth pattern that resembles that of follicular lymphoma. Conversely, an unusual diffuse variant of follicular lymphoma that often presents in inguinal lymph nodes can mimic the histologic appearance of MCL [90]. However, in contrast to follicular lymphoma, MCL cells are usually CD10–, CD5+, CD43+, and cyclin D1+. Like MCL, follicular lymphoma can present with gastrointestinal involvement as lymphomatous polyposis; such tumors are also best distinguished from MCL by immunohistochemistry. (See "Clinical manifestations, pathologic features, diagnosis, and prognosis of follicular lymphoma".)

Marginal zone lymphoma (nodal or extranodal) — Both extranodal marginal zone lymphoma (MZL) and MCL can involve the gastrointestinal tract and are neoplasms of small to medium-sized B lymphocytes. On immunophenotype, MCL expresses CD5 and cyclin D1 while extranodal MZL does not. In addition, MZL often contains monocytoid B cells and shows plasmacytic differentiation, which are not features of MCL. (See "Clinical manifestations, pathologic features, and diagnosis of extranodal marginal zone lymphoma of mucosa associated lymphoid tissue (MALT)".)

Lymphoblastic lymphoma — The blastoid variant of MCL has a high mitotic rate (typically 20 to 30 per 10 high power fields) and is often comprised of intermediate-sized cells with dispersed chromatin, irregular nuclear contours, and scant cytoplasm that mimic the appearance of lymphoblastic lymphoma, which can be of B or T cell origin. These cases are easily distinguished from lymphoblastic lymphoma by immunohistochemistry, as blastoid variant MCL expresses cyclin D1 and mature B cell markers (eg, surface immunoglobulin), whereas B lymphoblastic lymphomas lack surface immunoglobulin and express TdT, and T lymphoblastic lymphomas express TdT and additional T cell markers besides CD5.

PROGNOSIS — The course of MCL is moderately aggressive and variable. Median overall survival in modern trials incorporating intensive therapy is 8 to 10 years, with no plateau in the survival curve. Shorter survival times are seen with less intensive therapy.

Multiple studies have tried to determine prognostic factors to predict which patients will have a more aggressive course [94-103]. While most patients with MCL who do not begin therapy will die of their disease within a few years, a small proportion, increasingly better defined, may remain stable for years. These occasional patients with low stage (table 3), low-risk (table 2) disease may have an indolent course, managed by observation, splenectomy, or treatment with alkylating agents analogous to the treatment of patients with small lymphocytic lymphoma or follicular lymphoma. (See "Initial treatment of mantle cell lymphoma", section on 'Indications for treatment'.)

Several prognostic indices have been applied to patients with MCL. These include:

The International Prognostic Index (IPI) (table 2)

The Follicular Lymphoma International Prognostic Index (FLIPI) (table 4) [95]

The Mantle Cell Lymphoma International Prognostic Index (MIPI) (calculator 1) (table 5) [94,104]

All of these indices incorporate information about the patient's age, lactated dehydrogenase (LDH), and stage. They vary in how they incorporate information about nodal involvement, performance status, and blood counts.

The MIPI was originally described using data from 455 patients with advanced stage MCL enrolled on one of three clinical trials between 1996 and 2004 [94]. Using information regarding age, performance status, LDH, and leukocyte count, patients could be stratified into three risk groups (low, intermediate, and high) with significantly different estimated median overall survival (not reached, 58 months, and 37 months) and survival at five years (60, 35, and 20 percent) (table 5). The prognostic value of the MIPI was later confirmed in a separate cohort of 958 patients with MCL treated on prospective trials between 2004 and 2010 [105]. This latter study confirmed that the MIPI was able to identify three risk groups with significantly different estimated survival at five years (83, 63, and 34 percent). The differences in survival rates seen in this second study likely reflect differences in patient populations, available treatments, and adjunctive care. This also illustrates the importance of healthy scepticism when applying prognostic indices to the care of individual patients.

The blastoid variant of MCL is reported in some studies to be more aggressive [5,27,96-99], whereas patients presenting without anemia or splenomegaly [27,100], those with a normal serum free light chain ratio [106], or whose tumor cells do not overexpress cyclin D1, may have longer survival (see 'Cyclin D1' above) [42].

In one series of 52 patients with MCL, blastoid transformation occurred in 18 (35 percent), with a median survival time following transformation of four months [101]. Sixteen of the 18 had systemic involvement with circulating blastoid cells at the time of transformation. Leukocytosis, elevated serum LDH level, and high proliferative activity (as assessed by Ki-67 staining) were associated with an increased risk of this complication. In separate studies, age >60 and an increased mitotic index [102] or increased Ki-67 staining [107-109] were associated with significantly worse overall survival.

Patients whose MCL has mutant TP53 have inferior outcomes, including reduced overall survival (OS), shorter progression-free survival (PFS), and a higher rate of relapse [75]. In a cohort of 365 patients with MCL, high p53 expression by immunohistochemistry was a strong predictor of inferior overall survival (OS) and earlier treatment failure (TTF) [49]. Importantly, patients with high p53 expression (>50 percent positive lymphoma cells) had inferior OS and shorter TTF; this association was independent of MIPI score and Ki-67 index in multivariate analysis.

SOCIETY GUIDELINE LINKS — Links to society and government-sponsored guidelines from selected countries and regions around the world are provided separately. (See "Society guideline links: Lymphoma diagnosis and staging" and "Society guideline links: Management of mantle cell lymphoma".)

SUMMARY

Mantle cell lymphoma (MCL) – MCL is one of the mature B cell non-Hodgkin lymphomas (NHL). Its behavior is most often that of an aggressive disease. (See 'Introduction' above.)

Epidemiology – MCL accounts for 7 percent of NHL in adults in the United States and Europe. Presentation is usually in the sixth decade and there is a male predominance. (See 'Epidemiology' above.)

Clinical presentation – Most patients present with advanced stage disease. Most have lymphadenopathy (75 percent), while approximately 25 percent will present with symptoms from extranodal disease, such as involvement of the gastrointestinal tract. (See 'Clinical features' above.)

Morphology – The histologic pattern of MCL may be diffuse, nodular, or mantle zone, or a combination of the three. Most cases are composed exclusively of small to medium-sized lymphoid cells, with slightly irregular or "notched" nuclei (picture 7). (See 'Morphology' above.)

Immunophenotype – Mantle cell tumor cells express high levels of surface membrane immunoglobulin M and immunoglobulin D (sIgM±IgD), which is more often of lambda light chain type. They also express pan-B cell antigens (eg, CD19, CD20), CD5, and FMC7. Nuclear staining for cyclin D1 (BCL1) is present in >90 percent of cases, including those that are CD5 negative. (See 'Immunophenotype' above.)

Genetic features – Most cases will demonstrate t(11;14) by conventional cytogenetics or fluorescence in situ hybridization (FISH), although this rearrangement is not specific for MCL. (See 'Genetic features' above.)

Diagnosis – Tissue biopsy is required for diagnosis. Microscopically, this usually reveals a monomorphous pattern of small to medium-sized B lymphocytes with irregular nuclei, nuclear staining for cyclin D1 (BCL1), and t(11;14)(q13;q32) translocation between the CCND1 and immunoglobulin heavy chain (IgH). (See 'Diagnosis' above.)

Differential diagnosis – The differential diagnosis includes other NHLs composed of small to medium-sized cells, including chronic lymphocytic leukemia, follicular lymphoma, and marginal zone lymphoma. (See 'Differential diagnosis' above.)

Prognosis – The course of MCL is moderately aggressive and variable. The most commonly used prognostic scoring systems are the International Prognostic Index (IPI) (table 2), the Follicular Lymphoma International Prognostic Index (FLIPI) (table 4), and the Mantle cell lymphoma International Prognostic Index (calculator 1) (table 5). (See 'Prognosis' above.)

  1. Campo E, Jaffe ES, Cook JR, et al. The International Consensus Classification of Mature Lymphoid Neoplasms: a report from the Clinical Advisory Committee. Blood 2022; 140:1229.
  2. Alaggio R, Amador C, Anagnostopoulos I, et al. The 5th edition of the World Health Organization Classification of Haematolymphoid Tumours: Lymphoid Neoplasms. Leukemia 2022; 36:1720.
  3. Armitage JO, Weisenburger DD. New approach to classifying non-Hodgkin's lymphomas: clinical features of the major histologic subtypes. Non-Hodgkin's Lymphoma Classification Project. J Clin Oncol 1998; 16:2780.
  4. Lennert K. Malignant lymphomas other than Hodgkin's disease, Springer-Verlag, New York 1978.
  5. A clinical evaluation of the International Lymphoma Study Group classification of non-Hodgkin's lymphoma. The Non-Hodgkin's Lymphoma Classification Project. Blood 1997; 89:3909.
  6. Shivdasani RA, Hess JL, Skarin AT, Pinkus GS. Intermediate lymphocytic lymphoma: clinical and pathologic features of a recently characterized subtype of non-Hodgkin's lymphoma. J Clin Oncol 1993; 11:802.
  7. Sant M, Allemani C, Tereanu C, et al. Incidence of hematologic malignancies in Europe by morphologic subtype: results of the HAEMACARE project. Blood 2010; 116:3724.
  8. Smith A, Howell D, Patmore R, et al. Incidence of haematological malignancy by sub-type: a report from the Haematological Malignancy Research Network. Br J Cancer 2011; 105:1684.
  9. Zhou Y, Wang H, Fang W, et al. Incidence trends of mantle cell lymphoma in the United States between 1992 and 2004. Cancer 2008; 113:791.
  10. Jares P, Colomer D, Campo E. Molecular pathogenesis of mantle cell lymphoma. J Clin Invest 2012; 122:3416.
  11. Vegliante MC, Palomero J, Pérez-Galán P, et al. SOX11 regulates PAX5 expression and blocks terminal B-cell differentiation in aggressive mantle cell lymphoma. Blood 2013; 121:2175.
  12. Argatoff LH, Connors JM, Klasa RJ, et al. Mantle cell lymphoma: a clinicopathologic study of 80 cases. Blood 1997; 89:2067.
  13. Romaguera JE, Medeiros LJ, Hagemeister FB, et al. Frequency of gastrointestinal involvement and its clinical significance in mantle cell lymphoma. Cancer 2003; 97:586.
  14. Ferrer A, Salaverria I, Bosch F, et al. Leukemic involvement is a common feature in mantle cell lymphoma. Cancer 2007; 109:2473.
  15. Isaacson PG, MacLennan KA, Subbuswamy SG. Multiple lymphomatous polyposis of the gastrointestinal tract. Histopathology 1984; 8:641.
  16. Chim CS, Hu WH, Loong F, et al. GI manifestations of mantle cell lymphoma. Gastrointest Endosc 2003; 58:931.
  17. Ruskoné-Fourmestraux A, Delmer A, Lavergne A, et al. Multiple lymphomatous polyposis of the gastrointestinal tract: prospective clinicopathologic study of 31 cases. Groupe D'étude des Lymphomes Digestifs. Gastroenterology 1997; 112:7.
  18. Pittaluga S, Verhoef G, Criel A, et al. "Small" B-cell non-Hodgkin's lymphomas with splenomegaly at presentation are either mantle cell lymphoma or marginal zone cell lymphoma. A study based on histology, cytology, immunohistochemistry, and cytogenetic analysis. Am J Surg Pathol 1996; 20:211.
  19. Oinonen R, Franssila K, Elonen E. Central nervous system involvement in patients with mantle cell lymphoma. Ann Hematol 1999; 78:145.
  20. Gill S, Herbert KE, Prince HM, et al. Mantle cell lymphoma with central nervous system involvement: frequency and clinical features. Br J Haematol 2009; 147:83.
  21. Cheah CY, George A, Giné E, et al. Central nervous system involvement in mantle cell lymphoma: clinical features, prognostic factors and outcomes from the European Mantle Cell Lymphoma Network. Ann Oncol 2013; 24:2119.
  22. 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 1993; 100:373.
  23. Lardelli P, Bookman MA, Sundeen J, et al. Lymphocytic lymphoma of intermediate differentiation. Morphologic and immunophenotypic spectrum and clinical correlations. Am J Surg Pathol 1990; 14:752.
  24. 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 1994; 24:329.
  25. 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 1992; 16:637.
  26. Dorfman DM, Pinkus GS. Distinction between small lymphocytic and mantle cell lymphoma by immunoreactivity for CD23. Mod Pathol 1994; 7:326.
  27. Bosch F, López-Guillermo A, Campo E, et al. Mantle cell lymphoma: presenting features, response to therapy, and prognostic factors. Cancer 1998; 82:567.
  28. Kaptain S. CD5-negative mantle cell lymphoma. Mod Pathol 1998.
  29. DiRaimondo F, Albitar M, Huh Y, et al. The clinical and diagnostic relevance of CD23 expression in the chronic lymphoproliferative disease. Cancer 2002; 94:1721.
  30. Stein H, Lennert K, Feller AC, Mason DY. Immunohistological analysis of human lymphoma: correlation of histological and immunological categories. Adv Cancer Res 1984; 42:67.
  31. Harris NL, Nadler LM, Bhan AK. Immunohistologic characterization of two malignant lymphomas of germinal center type (centroblastic/centrocytic and centrocytic) with monoclonal antibodies. Follicular and diffuse lymphomas of small-cleaved-cell type are related but distinct entities. Am J Pathol 1984; 117:262.
  32. Mozos A, Royo C, Hartmann E, et al. SOX11 expression is highly specific for mantle cell lymphoma and identifies the cyclin D1-negative subtype. Haematologica 2009; 94:1555.
  33. Navarro A, Clot G, Royo C, et al. Molecular subsets of mantle cell lymphoma defined by the IGHV mutational status and SOX11 expression have distinct biologic and clinical features. Cancer Res 2012; 72:5307.
  34. Salaverria I, Royo C, Carvajal-Cuenca A, et al. CCND2 rearrangements are the most frequent genetic events in cyclin D1(-) mantle cell lymphoma. Blood 2013; 121:1394.
  35. Mynster T, Hultberg B, Bülow S. Multiple lymphomatous polyposis of the colon and rectum. Report of a case and review of the literature. Scand J Gastroenterol 1994; 29:545.
  36. Hashimoto Y, Nakamura N, Kuze T, et al. Multiple lymphomatous polyposis of the gastrointestinal tract is a heterogenous group that includes mantle cell lymphoma and follicular lymphoma: analysis of somatic mutation of immunoglobulin heavy chain gene variable region. Hum Pathol 1999; 30:581.
  37. Yang WI, Zukerberg LR, Motokura T, et al. Cyclin D1 (Bcl-1, PRAD1) protein expression in low-grade B-cell lymphomas and reactive hyperplasia. Am J Pathol 1994; 145:86.
  38. Zukerberg LR, Yang WI, Arnold A, Harris NL. Cyclin D1 expression in non-Hodgkin's lymphomas. Detection by immunohistochemistry. Am J Clin Pathol 1995; 103:756.
  39. Swerdlow SH, Yang WI, Zukerberg LR, et al. Expression of cyclin D1 protein in centrocytic/mantle cell lymphomas with and without rearrangement of the BCL1/cyclin D1 gene. Hum Pathol 1995; 26:999.
  40. Cheuk W, Wong KO, Wong CS, Chan JK. Consistent immunostaining for cyclin D1 can be achieved on a routine basis using a newly available rabbit monoclonal antibody. Am J Surg Pathol 2004; 28:801.
  41. Liu Z, Dong HY, Gorczyca W, et al. CD5- mantle cell lymphoma. Am J Clin Pathol 2002; 118:216.
  42. 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 2000; 95:2253.
  43. de Boer CJ, Vaandrager JW, van Krieken JH, et al. Visualization of mono-allelic chromosomal aberrations 3' and 5' of the cyclin D1 gene in mantle cell lymphoma using DNA fiber fluorescence in situ hybridization. Oncogene 1997; 15:1599.
  44. Wiestner A, Tehrani M, Chiorazzi M, et al. Point mutations and genomic deletions in CCND1 create stable truncated cyclin D1 mRNAs that are associated with increased proliferation rate and shorter survival. Blood 2007; 109:4599.
  45. Wlodarska I, Dierickx D, Vanhentenrijk V, et al. Translocations targeting CCND2, CCND3, and MYCN do occur in t(11;14)-negative mantle cell lymphomas. Blood 2008; 111:5683.
  46. Iaccarino I, Afify L, Aukema SM, et al. t(11;14)-positive mantle cell lymphomas lacking cyclin D1 (CCND1) immunostaining because of a CCND1 mutation or exclusive expression of the CCND1b isoform. Haematologica 2018; 103:e432.
  47. Lovec H, Grzeschiczek A, Kowalski MB, Möröy T. Cyclin D1/bcl-1 cooperates with myc genes in the generation of B-cell lymphoma in transgenic mice. EMBO J 1994; 13:3487.
  48. Fu K, Weisenburger DD, Greiner TC, et al. Cyclin D1-negative mantle cell lymphoma: a clinicopathologic study based on gene expression profiling. Blood 2005; 106:4315.
  49. Aukema SM, Hoster E, Rosenwald A, et al. Expression of TP53 is associated with the outcome of MCL independent of MIPI and Ki-67 in trials of the European MCL Network. Blood 2018; 131:417.
  50. Jerkeman M, Eskelund CW, Hutchings M, et al. Ibrutinib, lenalidomide, and rituximab in relapsed or refractory mantle cell lymphoma (PHILEMON): a multicentre, open-label, single-arm, phase 2 trial. Lancet Haematol 2018; 5:e109.
  51. Nordström L, Sernbo S, Eden P, et al. SOX11 and TP53 add prognostic information to MIPI in a homogenously treated cohort of mantle cell lymphoma--a Nordic Lymphoma Group study. Br J Haematol 2014; 166:98.
  52. Rodrigues JM, Hassan M, Freiburghaus C, et al. p53 is associated with high-risk and pinpoints TP53 missense mutations in mantle cell lymphoma. Br J Haematol 2020; 191:796.
  53. Walsh SH, Thorsélius M, Johnson A, et al. Mutated VH genes and preferential VH3-21 use define new subsets of mantle cell lymphoma. Blood 2003; 101:4047.
  54. Camacho FI, Algara P, Rodríguez A, et al. Molecular heterogeneity in MCL defined by the use of specific VH genes and the frequency of somatic mutations. Blood 2003; 101:4042.
  55. Hummel M, Tamaru J, Kalvelage B, Stein H. Mantle cell (previously centrocytic) lymphomas express VH genes with no or very little somatic mutations like the physiologic cells of the follicle mantle. Blood 1994; 84:403.
  56. Bertoni F, Zucca E, Cotter FE. Molecular basis of mantle cell lymphoma. Br J Haematol 2004; 124:130.
  57. Campo E, Raffeld M, Jaffe ES. Mantle-cell lymphoma. Semin Hematol 1999; 36:115.
  58. Tsujimoto Y, Yunis J, Onorato-Showe L, et al. Molecular cloning of the chromosomal breakpoint of B-cell lymphomas and leukemias with the t(11;14) chromosome translocation. Science 1984; 224:1403.
  59. De Wolf-Peeters C, Pittaluga S. Mantle-cell lymphoma. Ann Oncol 1994; 5 Suppl 1:35.
  60. Boehm T, Baer R, Lavenir I, et al. The mechanism of chromosomal translocation t(11;14) involving the T-cell receptor C delta locus on human chromosome 14q11 and a transcribed region of chromosome 11p15. EMBO J 1988; 7:385.
  61. Panayiotidis P, Kotsi P. Genetics of small lymphocyte disorders. Semin Hematol 1999; 36:171.
  62. Vandenberghe E, De Wolf-Peeters C, van den Oord J, et al. Translocation (11;14): a cytogenetic anomaly associated with B-cell lymphomas of non-follicle centre cell lineage. J Pathol 1991; 163:13.
  63. 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 1991; 88:9638.
  64. Bertoni F, Rinaldi A, Zucca E, Cavalli F. Update on the molecular biology of mantle cell lymphoma. Hematol Oncol 2006; 24:22.
  65. Allen JE, Hough RE, Goepel JR, et al. Identification of novel regions of amplification and deletion within mantle cell lymphoma DNA by comparative genomic hybridization. Br J Haematol 2002; 116:291.
  66. Sun T, Nordberg ML, Cotelingam JD, et al. Fluorescence in situ hybridization: method of choice for a definitive diagnosis of mantle cell lymphoma. Am J Hematol 2003; 74:78.
  67. Kridel R, Meissner B, Rogic S, et al. Whole transcriptome sequencing reveals recurrent NOTCH1 mutations in mantle cell lymphoma. Blood 2012; 119:1963.
  68. Quintanilla-Martinez L, Davies-Hill T, Fend F, et al. Sequestration of p27Kip1 protein by cyclin D1 in typical and blastic variants of mantle cell lymphoma (MCL): implications for pathogenesis. Blood 2003; 101:3181.
  69. Hofmann WK, de Vos S, Tsukasaki K, et al. Altered apoptosis pathways in mantle cell lymphoma detected by oligonucleotide microarray. Blood 2001; 98:787.
  70. Delfau-Larue MH, Klapper W, Berger F, et al. High-dose cytarabine does not overcome the adverse prognostic value of CDKN2A and TP53 deletions in mantle cell lymphoma. Blood 2015; 126:604.
  71. Parry-Jones N, Matutes E, Morilla R, et al. Cytogenetic abnormalities additional to t(11;14) correlate with clinical features in leukaemic presentation of mantle cell lymphoma, and may influence prognosis: a study of 60 cases by FISH. Br J Haematol 2007; 137:117.
  72. Greiner TC, Moynihan MJ, Chan WC, et al. p53 mutations in mantle cell lymphoma are associated with variant cytology and predict a poor prognosis. Blood 1996; 87:4302.
  73. 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 1997; 89:1421.
  74. Dreyling MH, Bullinger L, Ott G, et al. Alterations of the cyclin D1/p16-pRB pathway in mantle cell lymphoma. Cancer Res 1997; 57:4608.
  75. Eskelund CW, Dahl C, Hansen JW, et al. TP53 mutations identify younger mantle cell lymphoma patients who do not benefit from intensive chemoimmunotherapy. Blood 2017; 130:1903.
  76. Tirier C, Zhang Y, Plendl H, et al. Simultaneous presence of t(11;14) and a variant Burkitt's translocation in the terminal phase of a mantle cell lymphoma. Leukemia 1996; 10:346.
  77. Nagy B, Lundán T, Larramendy ML, et al. Abnormal expression of apoptosis-related genes in haematological malignancies: overexpression of MYC is poor prognostic sign in mantle cell lymphoma. Br J Haematol 2003; 120:434.
  78. Cuneo A, Bigoni R, Rigolin GM, et al. Cytogenetic profile of lymphoma of follicle mantle lineage: correlation with clinicobiologic features. Blood 1999; 93:1372.
  79. Espinet B, Solé F, Woessner S, et al. Translocation (11;14)(q13;q32) and preferential involvement of chromosomes 1, 2, 9, 13, and 17 in mantle cell lymphoma. Cancer Genet Cytogenet 1999; 111:92.
  80. Monni O, Oinonen R, Elonen E, et al. Gain of 3q and deletion of 11q22 are frequent aberrations in mantle cell lymphoma. Genes Chromosomes Cancer 1998; 21:298.
  81. Martinez-Climent JA, Vizcarra E, Sanchez D, et al. Loss of a novel tumor suppressor gene locus at chromosome 8p is associated with leukemic mantle cell lymphoma. Blood 2001; 98:3479.
  82. Schraders M, Pfundt R, Straatman HM, et al. Novel chromosomal imbalances in mantle cell lymphoma detected by genome-wide array-based comparative genomic hybridization. Blood 2005; 105:1686.
  83. Ghobrial IM, McCormick DJ, Kaufmann SH, et al. Proteomic analysis of mantle-cell lymphoma by protein microarray. Blood 2005; 105:3722.
  84. Rubio-Moscardo F, Climent J, Siebert R, et al. Mantle-cell lymphoma genotypes identified with CGH to BAC microarrays define a leukemic subgroup of disease and predict patient outcome. Blood 2005; 105:4445.
  85. Rizzatti EG, Falcão RP, Panepucci RA, et al. Gene expression profiling of mantle cell lymphoma cells reveals aberrant expression of genes from the PI3K-AKT, WNT and TGFbeta signalling pathways. Br J Haematol 2005; 130:516.
  86. Rinaldi A, Kwee I, Taborelli M, et al. Genomic and expression profiling identifies the B-cell associated tyrosine kinase Syk as a possible therapeutic target in mantle cell lymphoma. Br J Haematol 2006; 132:303.
  87. Salaverria I, Zettl A, Beà S, et al. Specific secondary genetic alterations in mantle cell lymphoma provide prognostic information independent of the gene expression-based proliferation signature. J Clin Oncol 2007; 25:1216.
  88. Kienle D, Katzenberger T, Ott G, et al. Quantitative gene expression deregulation in mantle-cell lymphoma: correlation with clinical and biologic factors. J Clin Oncol 2007; 25:2770.
  89. Beà S, Valdés-Mas R, Navarro A, et al. Landscape of somatic mutations and clonal evolution in mantle cell lymphoma. Proc Natl Acad Sci U S A 2013; 110:18250.
  90. Swerdlow SH, Campo E, Pileri SA, et al. The 2016 revision of the World Health Organization classification of lymphoid neoplasms. Blood 2016; 127:2375.
  91. World Health Organization Classification of Tumours of Haematopoietic and Lymphoid Tissues, Swerdlow SH, Campo E, Harris NL, et al. (Eds), IARC Press, Lyon 2008.
  92. Carvajal-Cuenca A, Sua LF, Silva NM, et al. In situ mantle cell lymphoma: clinical implications of an incidental finding with indolent clinical behavior. Haematologica 2012; 97:270.
  93. Barna G, Reiniger L, Tátrai P, et al. The cut-off levels of CD23 expression in the differential diagnosis of MCL and CLL. Hematol Oncol 2008; 26:167.
  94. Hoster E, Dreyling M, Klapper W, et al. A new prognostic index (MIPI) for patients with advanced-stage mantle cell lymphoma. Blood 2008; 111:558.
  95. Møller MB, Pedersen NT, Christensen BE. Mantle cell lymphoma: prognostic capacity of the Follicular Lymphoma International Prognostic Index. Br J Haematol 2006; 133:43.
  96. Fisher RI, Dahlberg S, Nathwani BN, et al. A clinical analysis of two indolent lymphoma entities: mantle cell lymphoma and marginal zone lymphoma (including the mucosa-associated lymphoid tissue and monocytoid B-cell subcategories): a Southwest Oncology Group study. Blood 1995; 85:1075.
  97. Meusers P, Engelhard M, Bartels H, et al. Multicentre randomized therapeutic trial for advanced centrocytic lymphoma: anthracycline does not improve the prognosis. Hematol Oncol 1989; 7:365.
  98. Berger F, Felman P, Sonet A, et al. Nonfollicular small B-cell lymphomas: a heterogeneous group of patients with distinct clinical features and outcome. Blood 1994; 83:2829.
  99. Matutes E, Parry-Jones N, Brito-Babapulle V, et al. The leukemic presentation of mantle-cell lymphoma: disease features and prognostic factors in 58 patients. Leuk Lymphoma 2004; 45:2007.
  100. Andersen NS, Jensen MK, de Nully Brown P, Geisler CH. A Danish population-based analysis of 105 mantle cell lymphoma patients: incidences, clinical features, response, survival and prognostic factors. Eur J Cancer 2002; 38:401.
  101. Räty R, Franssila K, Jansson SE, et al. Predictive factors for blastoid transformation in the common variant of mantle cell lymphoma. Eur J Cancer 2003; 39:321.
  102. Tiemann M, Schrader C, Klapper W, et al. Histopathology, cell proliferation indices and clinical outcome in 304 patients with mantle cell lymphoma (MCL): a clinicopathological study from the European MCL Network. Br J Haematol 2005; 131:29.
  103. Hartmann E, Fernàndez V, Moreno V, et al. Five-gene model to predict survival in mantle-cell lymphoma using frozen or formalin-fixed, paraffin-embedded tissue. J Clin Oncol 2008; 26:4966.
  104. Geisler CH, Kolstad A, Laurell A, et al. The Mantle Cell Lymphoma International Prognostic Index (MIPI) is superior to the International Prognostic Index (IPI) in predicting survival following intensive first-line immunochemotherapy and autologous stem cell transplantation (ASCT). Blood 2010; 115:1530.
  105. Hoster E, Klapper W, Hermine O, et al. Confirmation of the mantle-cell lymphoma International Prognostic Index in randomized trials of the European Mantle-Cell Lymphoma Network. J Clin Oncol 2014; 32:1338.
  106. Furtado M, Shah N, Levoguer A, et al. Abnormal serum free light chain ratio predicts poor overall survival in mantle cell lymphoma. Br J Haematol 2013; 160:63.
  107. Katzenberger T, Petzoldt C, Höller S, et al. The Ki67 proliferation index is a quantitative indicator of clinical risk in mantle cell lymphoma. Blood 2006; 107:3407.
  108. Garcia M, Romaguera JE, Inamdar KV, et al. Proliferation predicts failure-free survival in mantle cell lymphoma patients treated with rituximab plus hyperfractionated cyclophosphamide, vincristine, doxorubicin, and dexamethasone alternating with rituximab plus high-dose methotrexate and cytarabine. Cancer 2009; 115:1041.
  109. Hoster E, Rosenwald A, Berger F, et al. Prognostic Value of Ki-67 Index, Cytology, and Growth Pattern in Mantle-Cell Lymphoma: Results From Randomized Trials of the European Mantle Cell Lymphoma Network. J Clin Oncol 2016; 34:1386.
Topic 4702 Version 32.0

References