Mantle Cell Lymphoma

Back

Practice Essentials

Mantle cell lymphoma (MCL) is a lymphoproliferative disorder derived from a subset of naive pregerminal center cells localized in primary follicles or in the mantle region of secondary follicles. MCL represents 2-10% of all non-Hodgkin lymphomas.

Clinical presentation

The male-to-female ratio in MCL is 3:1, and the age range at presentation is 35-85 years, with a median of 68 years. Findings on the history include the following:

Physical examination findings include the following:

Workup

Studies and procedures for diagnosing and staging MCL are as follows:

Blood studies may yield the following results in MCL:

On immunophenotyping, tumor cells in MCL are monoclonal B cells with the following characteristics:

Management

Reliably curative treatments for MCL are lacking. An inexorable pattern of progression is characteristic, with a median time to treatment failure of less than 18 months. Primary MCL therapy may consist of the following:

Considerations in regimen selection are as follows:

The following regimens have been studied for relapsed or refractory MCL:

Salvage chemotherapy combinations include the following:

The role of ASCT consolidation after salvage therapy remains controversial and may benefit only a subset of patients with relapsed MCL. On the other hand, data for nonmyeloablative transplantation are very promising.

Overview

Mantle cell lymphoma (MCL) is recognized in the Revised European-American Lymphoma and World Health Organization classifications as a distinct clinicopathologic entity.[1, 2] MCL was not recognized by previous lymphoma classification schemes; it was frequently categorized as diffuse small-cleaved cell lymphoma (by the International Working Formulation) or centrocytic lymphoma (by the Kiel classification).[3] In the International Lymphoma Classification Project, it accounted for 8% of all non-Hodgkin lymphomas (NHLs).

For more information, see Non-Hodgkin Lymphoma and Pediatric Non-Hodgkin Lymphoma.

Pathophysiology

Mantle cell lymphoma (MCL) is a lymphoproliferative disorder derived from a subset of naive pregerminal center cells localized in primary follicles or in the mantle region of secondary follicles. Most cases of MCL are associated with chromosome translocation t(11;14)(q13;q32). This translocation involves the immunoglobulin heavy-chain gene on chromosome 14 and the BCL1 locus on chromosome 11.

The molecular consequence of translocation is overexpression of the protein cyclin D1 (coded by the PRAD1 gene located close to the breakpoint). Cyclin D1 plays a key role in cell cycle regulation and progression of cells from G1 phase to S phase by activation of cyclin-dependent kinases.

Epidemiology

Mantle cell lymphoma (MCL) is a type of non-Hodgkin lymphoma (NHL). NHL represents approximately 4% of all cancer diagnoses and the American Cancer Society estimated that in 2019, 74,200 new cases will occur.[4]  MCL is a relatively uncommon form of NHL, comprising 3-10% of NHL with an annual incidence of 0.5 to 1 cases per 100,000 population. The exact international prevalence of MCL is difficult to estimate because of the lack of uniform classification and procedures used for diagnosis.

Overall, whites are at higher risk of developing NHLs than blacks and Asian Americans . The male-to-female ratio is 3:1, and the age range at presentation is 35-85 years, with median age being 68 years.

Etiology

No causative factor has been identified for mantle cell lymphoma (MCL) or for most patients with non-Hodgkin lymphoma (NHL) of other types. NHL has been associated with viral infection (Epstein-Barr virus, HIV, human T-lymphotropic virus type 1, human herpesvirus 6), environmental factors (pesticides, hair dyes), and primary and secondary immunodeficiency.

Nonrandom chromosomal and molecular rearrangements play a major role in the pathogenesis of many lymphomas. The association of t(11;14)(q13;q32) with MCL suggests a causative role.

Clinical Presentation

History

Findings on the history include the following:

Physical examination

Physical examination findings include the following:

Complications

Complications from disease progression may include the following:

Differential Diagnosis

The differential diagnoses for MCL include the following:

Workup

Laboratory studies

Blood studies may yield the following results in mantle cell lymphoma (MCL):

Computed tomography

Body CT scanning is important for initial staging and for assessing the patient's response to treatment.

Immunocytochemistry

Tumor cells are monoclonal B cells that express surface immunoglobulin, immunoglobulin M, or immunoglobulin D. Cells are characteristically CD5+ and pan B-cell antigen positive (eg, CD19, CD20, CD22) but lack expression of CD10 and CD23. Cyclin D1 is overexpressed. Immunophenotyping helps differentiate MCL from other small B-cell lymphomas (see the Table, below).[5]

Table. Differential Diagnosis of Mantle Cell Lymphoma by Immunophenotyping



View Table

See Table

Cytogenetics

Most cases of MCL are associated with a chromosome translocation between chromosome 11 and 14, t(11;14)(q13;q32).[6, 7]

In a study of sox11, a transcription factor involved in embryonic neurogenesis and tissue remodeling, Chen et al concluded that nuclear expression of sox11 is highly associated with MCL, but it is independent of t(11;14)(q13;q32) in non–mantle cell B-cell neoplasms.[7] Chen et al assessed expression of sox11 and evaluated its association with t(11;14) and overexpression of cyclin D1 in 211 cases of B-cell neoplasms.

The investigators noted nuclear staining of sox11 in 95% (54/57) of MCLs (98% classical and 50% variant types). Of the 3 MCLs that were negative for the nuclear sox11 staining, 2 were positive for t(11;14).[7] The remaining 114 cases of B-cell lymphomas had variable cytoplasmic positive staining without nuclear positivity.

In addition, no nuclear staining of sox11 was found in 30 plasma cell myelomas, including 12 cases with t(11;14)(q13;q32), but intense nuclear staining of sox11 was present in 50% (5/10) of a subset of hairy cell leukemias, as well as an overexpression of cyclin D1.[7] Chen et al noted that the association with cyclin D1 overexpression in hairy cell leukemia may suggest sox11 involvement in cyclin D1 upregulation in hairy cell leukemia.[7]

Diagnostic procedures

Perform lymph node biopsy and aspiration together because aspiration alone is insufficient to establish a diagnosis. Use bone marrow aspirate/biopsy results for staging rather than diagnostic purposes.

Histologic findings

In the lymph node, MCL is characterized by expansion of the mantle zone that surrounds the lymph node germinal centers by small-to-medium atypical lymphocytes. These cells have irregular and indented nuclei, moderately coarse chromatin, and scant cytoplasm, resembling smaller cells of follicular lymphoma. However, mitoses are more numerous and large cells are infrequent.

A nodular appearance may be evident from expansion of the mantle zone in 30-50% of patients early in the disease. As disease progresses, the germinal centers become effaced, with obliteration of lymph node architecture.

A blastic variant of MCL, demonstrating numerous medium-to-large blastlike cells, has been reported and is associated with a more aggressive clinical course.

In bone marrow sections, neoplastic cells may infiltrate in a focal, often paratrabecular or diffuse pattern. Diagnosis of MCL should not be based on the examination of bone marrow alone; obtaining a lymph node biopsy is required.

Treatment

Approach considerations

Treatment of mantle cell lymphoma (MCL) remains a difficult problem because of the lack of reliably curative treatments and the paucity of prospective clinical trials.[6] Although 50-90% of these patients respond to combination chemotherapy, relatively few (30%) have a complete response. More aggressive chemotherapy regimens are currently under investigation and may yield higher complete response rates.

An inexorable pattern of progression is characteristic, with a median time to treatment failure of less than 18 months. Median survival usually ranges from 2-5 years, and only 5-10% of patients survive 10 years.

Stage I or II localized MCL is an extremely rare presentation, and literature on its management is retrospective and anecdotal. For stage II (bulky) and stages III-IV MCL, National Comprehensive Cancer Network (NCCN) guidelines recommend induction therapy with any of several regimens; patients who show a complete or partial response should then be considered for high-dose therapy followed by autologous stem cell rescue.[8]

A consensus statement by the Lymphoma Working Party of the European Society for Blood and Marrow Transplantation and the European MCL Network (EBMT/EMCL) on the role of stem cell transplantation in the management of MCL supports autologous SCT as the standard first-line consolidation therapy, and recommends that complete or partial remission be achieved before autologous SCT is performed. The EBMT/EMCL supports considering allogeneic SCT for patients who experience relapse after autologous SCT.[9]

Vaughn et al reported that allogeneic hematopoietic cell transplantation (HCT) provides a long-term survival benefit for patients with relapsed MCL, including those with refractory disease or multiple relapses. In this study, patients underwent HCT after nonmyeloablative conditioning with 2 Gy of total body irradiation with or without fludarabine and/or rituximab.The 5-year rates of overall survival (OS) and progression-free survival (PFS) in 70 patients were 55% and 46%, respectively. The 10-year rates of OS and PFS in 33 patients were 44% and 41%, respectively.[10]

Surgery is rarely indicated for therapeutic purposes in MCL. Exceptions include palliative procedures, such as relief of GI obstruction.

Regimens for primary MCL therapy

Primary MCL therapy may consist of the following:

A study by Griffiths et al found that the addition of rituximab to a standard first-line chemotherapy regimen significantly improved survival in older patients (mean age, 75 y) with MCL.[12]

Single alkylating agents

This therapy (eg, chlorambucil, 0.1-0.2 mg/kg for 3-6 wk) may be preferable for elderly patients or for those with serious comorbid medical problems who require treatment for lymphoma.

CVP and CHOP 

A randomized prospective trial that compared CVP with CHOP showed no advantage of adding doxorubicin, with similar response and survival rates. In some retrospective analyses, doxorubicin-containing regimens were associated with a longer event-free survival.

CVP is administered every 21 days, in the following regimen:

CHOP is administered every 21 days, in the following regimen:

Hyper-CVAD (with or without rituximab)

Hyper-CVAD with or without rituximab is a first-line regimen. Single-institution data (ie, M.D. Anderson Cancer Center) using hyper-CVAD plus rituximab yielded encouraging results as front-line therapy, especially in patients younger than 65 years.

Frontline therapy with hyper-CVAD plus rituximab (R-hyper-CVAD) in patients with MCL shows a higher complete response rate and response duration than any other regimen (100% response rate with 89% complete response).[13] At 36 months, the failure-free survival rate was greater than 80% in patients younger than 65 years, versus less than 50% in patients older than 65 years. In addition to age (ie, >65 y), beta2-microglobulin was found to be a very strong prognostic factor, especially in patients older than 65 years. Although very encouraging, this regimen is intensive and relatively toxic; data must be confirmed in randomized trials.

The hyper-CVAD drug regimen is a total of 8 cycles: 4 cycles of course A and 4 cycles of course B. Each cycle is started upon hematologic recovery, usually every 3 weeks.

Course A is as follows:

Course B is as follows:

Premedication and supportive measures are recommended in combination with the R-hyper-CVAD regimen. With high-dose methotrexate, give hydration with sodium bicarbonate for 48 hours. Prophylactic use of dexamethasone 0.1% (Maxidex ophthalmic solution), 1-2 drops every 4 hours while the patient is awake, for 7 days (during high-dose cytarabine administration) helps prevent conjunctivitis. Antibiotic prophylaxis may also be given. Additionally, doses should be modified according to the protocol being used.

R-CHOP

Another regimen is CHOP plus rituximab (R-CHOP). In a phase 2 randomized trial of CHOP versus R-CHOP in patients with previously untreated MCL, the complete response rate was higher with R-CHOP (34% vs 7%). The time to treatment failure was also in favor of R-CHOP, but the time to progression showed no significant difference. This study was reported by the German Low-Grade Lymphoma Study Group at the 2004 American Society of Clinical Oncology meeting.

Bortezomib

In October 2014 the US Food and Drug Administration (FDA) approved bortezomib for previously untreated patients with MCL. Bortezomib was already approved for patients with relapsed/refractory MCL.

Extension of the drug's indication was based on a phase III clinical trial of 487 previously untreated patients with MCL. The study compared the combination of bortezomib, rituximab, cyclophosphamide, doxorubicin, and prednisone with the standard R-CHOP regimen. The bortezomib combination improved progression-free survival by 11 months. The complete response rate for patients receiving the bortezomib combination regimen was 44%, compared with R-CHOP at 34%.[14]

Hyper-CVAD with autologous stem cell transplantation

Hyper-CVAD with or without rituximab followed by autologous stem cell transplantation (ASCT) was tested at the M.D. Anderson Cancer Center as a frontline regimen. It did not appear superior to hyper-CVAD over time, especially after the addition of rituximab to hyper-CVAD.[15]

Regimens for relapsed or refractory MCL

The following regimens have been studied for relapsed or refractory MCL:

R-hyper-CVAD

This therapy is currently being tested in patients with relapsed MCL in whom fludarabine or CHOP failed, but the results are not yet available. However, based on the frontline data, this is an acceptable option in patients with relapse. Future combinations of R-hyper-CVAD with other biologicals or new agents are potentially promising options.

Hyper-CVAD with or without rituximab followed by ASCT

Studies have shown that ASCT either as frontline consolidation or in the context of relapse provides high response rates and may improve disease-free survival. Unfortunately, this therapy is still typically associated with a continuous pattern of relapse.

Nucleoside analogues and combinations

Fludarabine as a single agent has demonstrated activity in MCL. It has been used, with or without rituximab, in the following combinations:

A higher complete response rate and/or longer response duration has been suggested when fludarabine is used in combination with an alkylator (eg, FC), with an anthracycline (eg, FND or fludarabine plus idarubicin), or both (eg, FCM). Such combinations could be used in refractory or relapse settings, with comparable response rates and duration of response.

The addition of rituximab to all of these regimens is clearly beneficial. For example, with FCM in a series of 38 patients with relapsed MCL, the overall response rate was 65% with rituximab versus 33% without rituximab, and the complete response rate was 35% with rituximab versus 0% without rituximab.[17]

Other nucleoside analogs have activity in MCL. For example, cladribine was found to be superior in combination with mitoxantrone.

Salvage chemotherapy combinations followed by ASCT

Rituximab, ifosfamide, carboplatin, etoposide (R-ICE) or etoposide, methylprednisolone (Solu-Medrol), high-dose cytarabine (Ara-C), cisplatin (ESHAP) followed by ASCT has been used. However, ASCT consolidation after salvage therapy remains controversial and may benefit only a subset of patients with relapsed MCL. On the other hand, data for nonmyeloablative transplantation are very promising, with some long-term survivors, including patients in whom prior high-dose therapy had failed.

Modified VR-CAP/R+ara-C (bortezomib, rituximab, cyclophosphamide, doxorubicin, and prednisone, alternating with rituximab and high-dose cytarabine), has been used for transplant-eligible patients with MCL. Most patients had intermediate- or high-risk disease by both (mantle-cell lymphoma international prognostic index (MIPI)-B and MIPI-C category. Complete response to induction was achieved in 32 (86%) of 37 evaluable patients; 2 achieved partial response, and 3 had primary refractory disease. Stem cell collection was successful in 1 attempt in 30 of 32 patients. The median follow-up of survivors measured from start of treatment is 17.4 months. Five patients have progressed, and 4 have died (2 owing to lymphoma, 2 from toxicity).[18]

Bortezomib

Goy et al and O'Connor established the therapeutic activity of bortezomib (Velcade) in relapsed and refractory MCL, and their work has been extended and confirmed in multicenter trials in the US and Canada with single-agent bortezomib, bortezomib in combination with chemotherapy and/or rituximab, and as a component of front-line therapy for MCL and other lymphomas.[19, 20]

The FDA has approved bortezomib for MCL in patients who have received at least one previous therapy. This approval was based on findings from the PINNACLE trial, a prospective, phase 2, multicenter, single arm, open-label study of 155 patients. Overall response rate was 31% with 8% complete response. Median duration of response was 9.3 months in responding patients and 15.4 in patients with CR. 41.[21]

The bortezomib regimen consists of 1.3 mg/m2 IV push twice per week (days 1, 4, 8, 11) followed by a 10-day treatment-free period (21 day cycle) for 8 cycles. Patients with stable disease or partial responses could receive treatment for up to 1 y, not to exceed maximum 17 cycles.

Starting therapy with subcutaneous (SC) administration of bortezomib may be considered for patients who have or are at high risk for peripheral neuropathy. Moreau et al observed the incidence of grade 2 or greater peripheral neuropathy was 24% for SC compared with 41% for IV; grade 3 or higher occurred in 6% when administered SC vs 16% for IV administration.[22]  

Lenalidomide

In June 2013, the FDA approved lenalidomide (Revlimid) for the treatment of MCL patients whose disease has relapsed or progressed after two prior therapies, one of which included bortezomib. The recommended dose and schedule for lenalidomide treatment is 25 mg PO once daily on days 1-21 of repeated 28-day cycles.

Approval was based on a single-arm, multicenter clinical trial involving 134 patients who had relapsed after, or were refractory to, bortezomib or a bortezomib-containing regimen. Overall response rate was 26%, and complete response (or complete response unconfirmed) was achieved by 9 patients. Partial response was achieved by 25 patients. Among these 34 patients, median duration of response was 16.6 months.[23]

The effectiveness of lenalidomide in combination with ibrutinib and venetoclax, especially in the management of p53-mutated cases, remains inconclusive.[24]

Ibrutinib

Ibrutinib (Imbruvica) was approved by the FDA in 2013 for treatment of MCL in patients who have received at least 1 prior therapy. It is a Bruton’s tyrosine kinase (BTK) inhibitor. BTK is a signaling molecule of the B-cell antigen receptor (bcr) and cytokine receptor pathways that results in activation of pathways necessary for B-cell trafficking, chemotaxis, and adhesion. The recommended dose is 560 mg PO once daily.

Approval was based on the results of a multicenter, international, single-arm trial of 111 patients with previously treated MCL. The efficacy results demonstrated a 68% overall response rate; 21% of patients achieved a complete response and 47% of patients achieved a partial response. The median duration of response was 17.5 months.[25]

Ibrutinib and palbociclib 

A phase I trial of ibrutinib plus palbociclib in previously treated mantle cell lymphoma found that maximum tolerated doses were ibrutinib 560 mg daily plus palbociclib 100 mg days 1-21 of each 28-day cycle. The dose-limiting toxicity was grade 3 rash. The most common grade 3-4 toxicities included neutropenia (41%), thrombocytopenia (30%), hypertension (15%), febrile neutropenia (15%), and lung infection (11%). The overall and complete response rates were 67% and 37%, respectively, and with a median follow-up of 25.6 months, the 2-year progression-free survival was 59.4% and the 2-year response duration was 69.8%. A phase II multicenter clinical trial to further characterize efficacy is ongoing.[26]

Acalabrutinib

Acalabrutinib (Calquence) is a novel irreversible second-generation BTK inhibitor that was shown to be more potent and selective than ibrutinib. In October 2017, the FDA granted an accelerated approval to acalabrutinib as a treatment for adult patients with MCL following at least 1 prior therapy. The approval was based on findings from the 124-patient ACE-LY-004 phase II trial, in which the investigator-assessed objective response rate (ORR) was 81% with acalabrutinib (95% confidence index [CI], 73%-87%). The complete response rate with acalabrutinib was 40% and the partial response rate was 41%.[27]

Zanubrutinib

In November 2019, the FDA granted accelerated approval to another BTK inhibitor, zanubrutinib (Brukinsa), for MCL in patients who have received at least 1 prior therapy. Approval was based on results from two phase II, open-label, single-arm trials, which showed an overall response rate of 84% In one study, MCL patients (n=86) had a 59% complete response and a 24% partial response rate. In the other study, MCL patients (n=32) had a 22% complete and 62% partial response rate.[28] For both studies the primary endpoint was overall response rate, based on the 2014 Lugano classification and assessed by an independent review committee. 

Radioimmunotherapy

Both iodine-131–based tositumomab (Bexxar)[29] and yttrium-90–based ibritumomab tiuxetan (Zevalin)[30] have shown activity as single agents for salvage therapy of MCL. Studies have reported responses with radioimmunotherapy (RIT) in MCL, including some complete responses that lasted more than 1 year. Additional ongoing studies are exploring combinations of RIT with chemotherapy for untreated or relapsed MCL. Strategies including RIT as part of high-dose therapy have shown encouraging results.

The use of RIT as part of a nonmyeloablative allotransplantation conditioning regimen is another promising strategy currently being tested in clinical trials.

Rituximab

The anti-CD20 monoclonal antibody rituximab, used as a single agent, has activity in MCL, yielding a response rate of 35%, a complete response rate of 10-15%, and a median duration of response of approximately 1 year in rituximab-naive patients. The potential role of rituximab as a maintenance therapy for patients with MCL is not yet well defined. The benefit of rituximab has been confirmed in combination with all chemotherapy regimens tested.

Borgerding et al reported responses to repeat treatment with rituximab in approximately two thirds of patients with MCL who had responded to initial treatment. However, lasting remissions in these cases were achieved only by high-dose chemotherapy with stem cell transplantation.[31]

Rituximab and thalidomide

Promising results have been shown using rituximab (standard dose) and thalidomide (200 mg/d, with incremental dose increases to 400 mg on day 15) continued as maintenance therapy until progression or relapse.[32] In a small series, the response rate was 81% (complete response rate of 31%) and the median progression-free survival was 20.4 months (95% confidence interval, 17.3-23.6). The estimated 3-year survival rate was 75%. This approach would be an appealing alternative in elderly patients.

High-dose chemotherapy with autologous bone marrow or stem cell transplantation

High-dose chemotherapy with autologous bone marrow or stem cell transplantation has not proved curative for MCL when used as second-line therapy.[33] Long-term survival data are currently unavailable in the setting of high-dose chemotherapy applied as consolidation therapy to patients in first complete remission.[34]

Consultations

The following specialists may need to be consulted:

Diet

Consultation with a dietitian may be necessary for patients with poor oral intake or marked weight loss. Special attention and support is required for patients receiving chemotherapy, such as appetite stimulants or diet supplements. Patients who are neutropenic require education about food hygiene.

Follow-up

Usually, treatment and follow-up care of patients with MCL is performed in an outpatient setting. Follow-up may include the following:

Admit patients for complications, disease progression, or adverse chemotherapy effects (eg, neutropenic fever or mucositis).

Complications of treatment

Complications from chemotherapy may include the following:

Prognosis

Mantle cell lymphoma (MCL) is associated with a poor prognosis. It exhibits a moderately aggressive course similar to that of intermediate-grade non-Hodgkin lymphoma (NHL). Unlike intermediate-grade lymphomas, it is rarely curable with currently available standard treatment.

Despite response rates of 50-70% with many regimens, MCL typically progresses after chemotherapy. Median time to treatment failure is less than 18 months. The median survival time is approximately 3 years (range, 2-5 y); the 10-year survival rate is only 5-10%.

Developing reliable prognostic markers has been difficult in the absence of better-standardized therapy. Younger age and limited disease are favorable prognostic features.

The blastoid variant, commonly associated with TP53 mutations, has been associated with a worse prognosis. Gene expression profile analysis identified MCL patient subsets with more than 5 years' difference in median survival, based on cyclin D1 and other proliferation signature genes.

Patient Education

Clearly explain all available treatment options and provide detailed instruction about the adverse effects of chemotherapy. Consider enrollment of the patient into a clinical trial. Provide psychosocial counseling.

For patient education information, see the Blood and Lymphatic System Center, as well as Lymphoma.

Guidelines Summary

Guidelines contributors: Priyank P Patel, MD, Hematology/Oncology Fellow, Roswell Park Cancer Institute, University at BuffaloFrancisco J Hernandez-Ilizaliturri, MD; Chief, Lymphoma and Myeloma Section; Professor of Medicine, Department of Medical Oncology; Director of The Lymphoma Translational Research Program; Associate Professor of Immunology, Roswell Park Cancer Institute 

Non-Hodgkin Lymphoma (NHL) Classification Schemas

The three most commonly used classification schemas for non-Hodgkin lymphoma (NHL) are as follows:

The Working Formulation, originally proposed in 1982, classified and grouped lymphomas by morphology and clinical behavior (ie, low, intermediate, or high grade) with 10 subgroups labeled A to J.[1] In 1994, the Revised European-American Lymphoma (REAL) classification attempted to apply immunophenotypic and genetic features in identifying distinct clinicopathologic NHL entities.[2]

The World Health Organization (WHO) classification, first introduced in 2001 and updated in 2008 and 2016, further elaborates upon the REAL approach. This classification divides NHL into two groups: those of B-cell origin and those of T-cell/natural killer (NK)–cell origin.[35]

Although considered obsolete, the National Cancer Institute’s Working Formulation (IWF) classification is still used mainly for historical data comparisons.[3]

World Health Organization Classification

The WHO modification of the REAL classification of NHL is based on morphology and cell lineage. Within the B-cell and T-cell categories, two subdivisions are recognized: precursor neoplasms, which correspond to the earliest stages of differentiation, and more mature differentiated neoplasms.[35]

The WHO classification subtypes for NHL precursors are as follows:

The WHO classification subtypes for mature B-cell neoplasms are as follows:

Diagnosis

MCL is diagnosed in accordance with the WHO criteria for hematological neoplasms and detection of cyclin D1 expression or the t(11;14) translocation along with mature B-cell proliferation. The National Comprehensive Cancer Network (NCCN) recommends the following studies to establish a diagnosis of MCL[8] :

The following studies may be useful in select circumstances:

Risk stratification

The European Society for Medical Oncology (ESMO) recommends the 2008 MCL International Prognostic Index (MIPI) for risk stratification.[36]  The MIPI includes the following risk factors[37] :

Based on the MIPI score, patients can be categorized as follows[37] :

In addition, both the NCCN and ESMO recommend assaying Ki-67 proliferative antigen to evaluate cell proliferation. Low Ki-67 (<  30%) is associated with a more favorable prognosis; however, this finding is not used to guide treatment decisions.[8, 36]

Treatment

The NCCN and ESMO offer similar treatment recommendations, as follows[8, 36] :

For induction therapy, the NCCN lists the following aggressive regimens[8] :

Less aggressive induction regimens include the following: 

For consolidation therapy, the NCCN recommends the following as first-line:

Second-line therapy regimens following short response duration of prior chemoimmunotherapy:

Second-line therapy regimens following extended response duration of prior chemoimmunotherapy:

How is mantle cell lymphoma (MCL) categorized?What is mantle cell lymphoma (MCL)?Which clinical history findings are characteristic of mantle cell lymphoma (MCL)?Which physical findings are characteristic of mantle cell lymphoma (MCL)?Which tests are used to diagnose and stage mantle cell lymphoma (MCL)?Which results on blood studies are characteristic of mantle cell lymphoma (MCL)?What are the immunophenotyping characteristics of mantle cell lymphoma (MCL)?How is primary mantle cell lymphoma (MCL) treated?Which factors should be considered in chemotherapy regimen selection for mantle cell lymphoma (MCL)?Which chemotherapy regimens have been studied for the treatment of relapsed or refractory mantle cell lymphoma (MCL)?Which salvage chemotherapy combinations are used in the treatment of mantle cell lymphoma (MCL)?What is the pathophysiology of mantle cell lymphoma (MCL)?What is the prevalence of mantle cell lymphoma (MCL)?What causes mantle cell lymphoma (MCL)?What are the signs and symptoms of mantle cell lymphoma (MCL)?What are the most common physical findings of mantle cell lymphoma (MCL)?What are the less common physical findings of mantle cell lymphoma (MCL)?What are the possible complications of mantle cell lymphoma (MCL)?What conditions are included in the differential diagnoses of mantle cell lymphoma (MCL)?What is the role of lab tests in the workup of mantle cell lymphoma (MCL)?What is the role of CT in the workup of mantle cell lymphoma (MCL)?What is the role of immunocytochemistry in the workup of mantle cell lymphoma (MCL)?What is the role of cytogenetics in the workup of mantle cell lymphoma (MCL)?What is the role of biopsy in the workup of mantle cell lymphoma (MCL)?Which histologic findings are characteristic of mantle cell lymphoma (MCL)?What is the role of fludarabine in mantle cell lymphoma (MCL) treatment?What is the role of ibrutinib in the treatment of mantle cell lymphoma (MCL)?What is the role of acalabrutinib in the treatment of mantle cell lymphoma (MCL)?What is the role of radioimmunotherapy in the treatment of mantle cell lymphoma (MCL)?What is the role of high-dose chemotherapy with autologous bone marrow or stem cell transplantation in the treatment of mantle cell lymphoma (MCL)?How is mantle cell lymphoma (MCL) treated?Which therapies are used in the treatment of primary mantle cell lymphoma (MCL)?What is the role of single alkylating agents in the treatment of mantle cell lymphoma (MCL)?What is the CVP chemotherapy regimen for mantle cell lymphoma (MCL)?What is the CHOP chemotherapy regimen for mantle cell lymphoma (MCL)?What is the role of the hyper-CVAD chemotherapy regimen in the treatment of mantle cell lymphoma (MCL) involve?What is the hyper-CVAD chemotherapy regimen for mantle cell lymphoma (MCL)?What is the role of the R-CHOP chemotherapy regimen in the treatment of mantle cell lymphoma (MCL)?What is the role of bortezomib in the treatment of mantle cell lymphoma (MCL)?What is the efficacy of the hyper-CVAD chemotherapy regimen followed by ASCT as a frontline treatment for mantle cell lymphoma (MCL)?How is relapsed or refractory mantle cell lymphoma (MCL) treated?What is the role of the R-hyper-CVAD chemotherapy regimen in the treatment of relapsed mantle cell lymphoma (MCL)?What is the efficacy of the hyper-CVAD chemotherapy regimen followed by ASCT in the treatment of mantle cell lymphoma (MCL)?What is the role of nucleoside analogues and combinations in the treatment of mantle cell lymphoma (MCL)?What is the role of ASCT in the treatment of mantle cell lymphoma (MCL)?What is the role of bortezomib in the treatment of mantle cell lymphoma (MCL)?What is the role of lenalidomide in the treatment of mantle cell lymphoma (MCL)?What is the role of rituximab in the treatment of mantle cell lymphoma (MCL)?What is the role of combined rituximab and thalidomide in the treatment of mantle cell lymphoma (MCL)?Which specialist consultations may be beneficial to patients with mantle cell lymphoma (MCL)?What is included in the long-term follow-up care for mantle cell lymphoma (MCL)?What are the possible complications of mantle cell lymphoma (MCL) treatment?What is the prognosis of mantle cell lymphoma (MCL)?What is included in patient education about mantle cell lymphoma (MCL)?What are the most commonly used classification schemas for non-Hodgkin lymphoma (NHL)?What is the WHO classification of non-Hodgkin lymphoma (NHL)?What are the NCCN guidelines on the diagnosis of mantle cell lymphoma (MCL)?What is the MCL global Prognostic Index (MIPI)?What are the NCCN and ESMO treatment guidelines for mantle cell lymphoma (MCL)?Which aggressive induction therapy regimens for mantle cell lymphoma (MCL) are recommended by NCCN?Which less aggressive induction therapy regimens for mantle cell lymphoma (MCL) are recommended by NCCN?What are the NCCN guidelines for first-line consolidation therapy for mantle cell lymphoma (MCL)?What are the NCCN guidelines for second-line therapy for mantle cell lymphoma (MCL)?

Author

Muhammad Rashid Abbasi, MD, Former Assistant Professor of Medicine, Albert Einstein College of Medicine; Consulting Staff, Department of Internal Medicine, Division of Hematology/Oncology, Jacobi Medical Center, Morristown Memorial Hospital, and St Clare's Hospital

Disclosure: Nothing to disclose.

Coauthor(s)

Joseph A Sparano, MD, Professor, Department of Medicine (Oncology), Professor, Department of Obstetrics and Gynecology and Women's Health, Albert Einstein College of Medicine; Associate Chairman for Clinical Research, Department of Oncology, Montefiore Medical Center; Associate Director for Clinical Research, Albert Einstein Cancer Center

Disclosure: Nothing to disclose.

Specialty Editors

Francisco Talavera, PharmD, PhD, Adjunct Assistant Professor, University of Nebraska Medical Center College of Pharmacy; Editor-in-Chief, Medscape Drug Reference

Disclosure: Received salary from Medscape for employment. for: Medscape.

Chief Editor

Emmanuel C Besa, MD, Professor Emeritus, Department of Medicine, Division of Hematologic Malignancies and Hematopoietic Stem Cell Transplantation, Kimmel Cancer Center, Jefferson Medical College of Thomas Jefferson University

Disclosure: Nothing to disclose.

References

  1. Swerdlow SH, Campo E, Pileri SA, Harris NL, Stein H, Siebert R, et al. The 2016 revision of the World Health Organization classification of lymphoid neoplasms. Blood. 2016 May 19. 127 (20):2375-90. [View Abstract]
  2. Harris NL, Jaffe ES, Stein H, et al. A revised European-American classification of lymphoid neoplasms: a proposal from the International Lymphoma Study Group. Blood. 1994 Sep 1. 84(5):1361-92. [View Abstract]
  3. The Non-Hodgkin's Lymphoma Pathologic Classification Project. National Cancer Institute sponsored study of classifications of non- Hodgkin's lymphomas: summary and description of a working formulation for clinical usage. Cancer. 1982 May 15. 49(10):2112-35. [View Abstract]
  4. American Cancer Society. Cancer Facts & Figures 2019. Available at https://www.cancer.org/content/dam/cancer-org/research/cancer-facts-and-statistics/annual-cancer-facts-and-figures/2019/cancer-facts-and-figures-2019.pdf. Accessed: March 11, 2019.
  5. Gao J, Peterson L, Nelson B, Goolsby C, Chen YH. Immunophenotypic variations in mantle cell lymphoma. Am J Clin Pathol. 2009 Nov. 132(5):699-706. [View Abstract]
  6. Weigert O, Unterhalt M, Hiddemann W, Dreyling M. Mantle cell lymphoma: state-of-the-art management and future perspective. Leuk Lymphoma. 2009 Dec. 50(12):1937-50. [View Abstract]
  7. Chen YH, Gao J, Fan G, Peterson LC. Nuclear expression of sox11 is highly associated with mantle cell lymphoma but is independent of t(11;14)(q13;q32) in non-mantle cell B-cell neoplasms. Mod Pathol. 2010 Jan. 23(1):105-12. [View Abstract]
  8. [Guideline] National Comprehensive Cancer Network. NCCN Clinical Practice Guidelines in Oncology: B-cell Lymphomas, Version 2.2019. NCCN. Available at https://www.nccn.org/professionals/physician_gls/pdf/b-cell.pdf. March 6, 2019; Accessed: March 13, 2019.
  9. Robinson S, Dreger P, Caballero D, Corradini P, Geisler C, Ghielmini M, et al. The EBMT/EMCL consensus project on the role of autologous and allogeneic stem cell transplantation in mantle cell lymphoma. Leukemia. 2015 Feb. 29 (2):464-73. [View Abstract]
  10. Vaughn JE, Sorror ML, Storer BE, Chauncey TR, Pulsipher MA, Maziarz RT, et al. Long-term sustained disease control in patients with mantle cell lymphoma with or without active disease after treatment with allogeneic hematopoietic cell transplantation after nonmyeloablative conditioning. Cancer. 2015 Jul 24. [View Abstract]
  11. Ruan J, Martin P, Shah B, Schuster SJ, Smith SM, Furman RR, et al. Lenalidomide plus Rituximab as Initial Treatment for Mantle-Cell Lymphoma. N Engl J Med. 2015 Nov 5. 373 (19):1835-44. [View Abstract]
  12. Griffiths R, Mikhael J, Gleeson M, Danese M, Dreyling M. Addition of rituximab to chemotherapy alone as first-line therapy improves overall survival in elderly patients with mantle cell lymphoma. Blood. 2011 Nov 3. 118(18):4808-16. [View Abstract]
  13. Foran JM, Rohatiner AZ, Cunningham D, et al. European phase II study of rituximab (chimeric anti-CD20 monoclonal antibody) for patients with newly diagnosed mantle-cell lymphoma and previously treated mantle-cell lymphoma, immunocytoma, and small B-cell lymphocytic lymphoma. J Clin Oncol. 2000 Jan. 18(2):317-24. [View Abstract]
  14. Mulcahy N. FDA Approves Bortezomib (Velcade) for Untreated MCL. Medscape Medical News. Available at http://www.medscape.com/viewarticle/833054. October 9, 2014; Accessed: November 4, 2017.
  15. Kasamon YL, Jones RJ, Brodsky RA, et al. Immunologic recovery following autologous stem-cell transplantation with pre- and posttransplantation rituximab for low-grade or mantle cell lymphoma. Ann Oncol. 2009 Oct 30. epub ahead of print. [View Abstract]
  16. Cohen BJ, Moskowitz C, Straus D, et al. Cyclophosphamide/fludarabine (CF) is active in the treatment of mantle cell lymphoma. Leuk Lymphoma. 2001 Sep-Oct. 42(5):1015-22. [View Abstract]
  17. Hiddemann W, Dreyling M, Unterhalt M, et al. Rituximab plus chemotherapy in follicular and mantle cell lymphomas. Semin Oncol. 2003 Feb. 30(1 Suppl 2):16-20. [View Abstract]
  18. Smith SD, Gandhy S, Gopal AK, Reddy P, Shadman M, Till BG, et al. Modified VR-CAP, Alternating With Rituximab and High-dose Cytarabine: An Effective Pre-transplant Induction Regimen for Mantle Cell Lymphoma. Clin Lymphoma Myeloma Leuk. 2019 Jan. 19 (1):48-52. [View Abstract]
  19. Goy A, Younes A, McLaughlin P, et al. Update on a phase (ph) 2 study of bortezomib in patients (pts) with relapsed or refractory indolent or aggressive non-Hodgkin's lymphomas (NHL). ASCO Annual Meeting Proceedings (Post-Meeting Edition). J Clin Oncol. 2004. 22 (July 15 suppl):14S (6581).
  20. O'Connor OA. Marked clinical activity of the novel proteasome inhibitor bortezomib in patients with relapsed follicular (RL) and mantle cell lymphoma (MCL). ASCO Annual Meeting Proceedings (Post-Meeting Edition). J Clin Oncol. 2004. 22 (July 15 suppl):14S (6582).
  21. Fisher RI, Bernstein SH, Kahl BS, Djulbegovic B, Robertson MJ, de Vos S, et al. Multicenter phase II study of bortezomib in patients with relapsed or refractory mantle cell lymphoma. J Clin Oncol. 2006 Oct 20. 24(30):4867-74. [View Abstract]
  22. Moreau P, Pylypenko H, Grosicki S, Karamanesht I, Leleu X, Grishunina M, et al. Subcutaneous versus intravenous administration of bortezomib in patients with relapsed multiple myeloma: a randomised, phase 3, non-inferiority study. Lancet Oncol. 2011 May. 12(5):431-40. [View Abstract]
  23. FDA Approves Lenalidomide. US Food and Drug Administration. Available at https://wayback.archive-it.org/7993/20161022171115/http://www.fda.gov/Drugs/InformationOnDrugs/ApprovedDrugs/ucm355438.htm. Accessed: November 4, 2017.
  24. Morabito F, Skafi M, Recchia AG, Kashkeesh A, Hindiyeh M, Sabatleen A, et al. Lenalidomide for the treatment of mantle cell lymphoma. Expert Opin Pharmacother. 2019 Jan 4. 1-8. [View Abstract]
  25. Wang ML, Rule S, Martin P, Goy A, Auer R, et al. Targeting BTK with ibrutinib in relapsed or refractory mantle-cell lymphoma. N Engl J Med. 2013 Aug 8. 369 (6):507-16. [View Abstract]
  26. Martin P, Bartlett NL, Blum KA, Park S, Maddocks K, Ruan J, et al. A phase I trial of ibrutinib plus palbociclib in previously treated mantle cell lymphoma. Blood. 2019 Jan 28. [View Abstract]
  27. Wang M, Rule S, Zinzani PL, Goy A, Casasnovas O, Smith SD, et al. Acalabrutinib in relapsed or refractory mantle cell lymphoma (ACE-LY-004): a single-arm, multicentre, phase 2 trial. Lancet. 2017 Dec 11. [View Abstract]
  28. Brukinsa (zanubrutinib) [package insert]. San Matteo, CA: BeiGene USA, Inc. November 2019. Available at
  29. Cheung MC, Maceachern JA, Haynes AE, Meyer RM, Imrie K, Members of the Hematology Disease Site Group of Cancer Care Ontario’s Program in Evidence-Based Care. I-Tositumomab in lymphoma. Curr Oncol. 2009 Sep. 16 (5):32-47. [View Abstract]
  30. Wang M, Oki Y, Pro B, et al. Phase II study of yttrium-90-ibritumomab tiuxetan in patients with relapsed or refractory mantle cell lymphoma. J Clin Oncol. 2009 Nov 1. 27(31):5213-8. [View Abstract]
  31. Borgerding A, Hasenkamp J, Glass B, Wulf G, Trumper L. Rituximab retherapy in patients with relapsed aggressive B cell and mantle cell lymphoma. Ann Hematol. 2010 Mar. 89(3):283-9. [View Abstract]
  32. Kaufmann H, Raderer M, Wöhrer S. Antitumor activity of rituximab plus thalidomide in patients with relapsed/refractory mantle cell lymphoma. Blood. 2004 Oct 15. 104(8):2269-71. [View Abstract]
  33. Freedman AS, Neuberg D, Gribben JG, et al. High-dose chemoradiotherapy and anti-B-cell monoclonal antibody-purged autologous bone marrow transplantation in mantle-cell lymphoma: no evidence for long-term remission. J Clin Oncol. 1998 Jan. 16(1):13-8. [View Abstract]
  34. Khouri IF, Saliba RM, Okoroji GJ, et al. Long-term follow-up of autologous stem cell transplantation in patients with diffuse mantle cell lymphoma in first disease remission: the prognostic value of beta2-microglobulin and the tumor score. Cancer. 2003 Dec 15. 98(12):2630-5. [View Abstract]
  35. Swerdlow SH, et al. WHO Classification of Tumours of Haematopoietic and Lymphoid Tissues. Revised 4th Edition. Lyon, France: International Agency for Research on Cancer; 2017.
  36. [Guideline] Dreyling M, Campo E, Hermine O, Jerkeman M, Le Gouill S, Rule S, et al. Newly diagnosed and relapsed mantle cell lymphoma: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol. 2017 Jul 1. 28 (suppl_4):iv62-iv71. [View Abstract]
  37. Hoster E, Dreyling M, Klapper W, et al. A new prognostic index (MIPI) for patients with advanced-stage mantle cell lymphoma. Blood. 2008 Jan 15. 111 (2):558-65. [View Abstract]
  38. Agazzi A. Report on the 56th ASH Annual Meeting, San Francisco, 4-9 December 2014. Ecancermedicalscience. 2015. 9:514. [View Abstract]
  39. Coiffier B, Ribrag V. Exploring mammalian target of rapamycin (mTOR) inhibition for treatment of mantle cell lymphoma and other hematologic malignancies. Leuk Lymphoma. 2009 Dec. 50(12):1916-30. [View Abstract]
  40. Bogner C, Dechow T, Ringshausen I, et al. Immunotoxin BL22 induces apoptosis in mantle cell lymphoma (MCL) cells dependent on Bcl-2 expression. Br J Haematol. 2010 Jan. 148(1):99-109. [View Abstract]
  41. Kirschbaum M, Frankel P, Popplewell L, Zain J, Delioukina M, Pullarkat V, et al. Phase II Study of Vorinostat for Treatment of Relapsed or Refractory Indolent Non-Hodgkin's Lymphoma and Mantle Cell Lymphoma. J Clin Oncol. 2011 Mar 20. 29(9):1198-203. [View Abstract]
  42. Lowes R. FDA OKs Ibrutinib (Imbruvica) for Mantle Cell Lymphoma. Medscape [serial online]. Available at http://www.medscape.com/viewarticle/814307. November 13, 2013; Accessed: November 4, 2017.
  43. U.S. Food and Drug Administration. FDA approves Imbruvica for rare blood cancer. U.S. Food and Drug Administration. Available at http://www.fda.gov/NewsEvents/Newsroom/PressAnnouncements/ucm374761.htm#. Accessed: November 18, 2013.
  44. Wu J, Liu C, Tsui ST, Liu D. Second-generation inhibitors of Bruton tyrosine kinase. J Hematol Oncol. 2016 Sep 2. 9 (1):80. [View Abstract]
Disease CD5 CD20 CD23 CD10 CD103 FMC7 Cyclin D1 Sig*
MCL++++/–++
B-CLL/SLL++++
PLL–/++++/–+++
MZL§+++/–+
SLVL||++–/++/–++
LPL+–/+++
FL#++++/–++
HCL**+++–/+++
* SIg = surface immunoglobulins



B-CLL/SLL = B-cell chronic lymphocytic leukemia/small lymphocytic leukemia



PLL = prolymphocytic leukemia



§ MZL = marginal zone leukemia



|| SLVL = splenic lymphoma with villous lymphocytes



LPL = lymphoplasmacytic lymphoma



# FL = Follicular lymphoma



** HCL = Hairy cell leukemia