Categories
Uncategorized

Mantle cell lymphoma II

Mantle Cell Lymphoma is a rare B-cell malignancy that can invade almost any structure in the body and recur after short-lived clinical responses. 

The pathogenesis and clinical features are well defined, but management has not yet been optimized. 

Induction with traditional immune-chemotherapy regimens that are used in other non- Hodgkin lymphomas rarely generate durable remissions. 

Mantle cell lymphoma (MCL) is an uncommon but aggressive lymphoma, comprising about 6% of all non-Hodgkin lymphomas, with a typical median survival of 5-7 years 

Patients are much more commonly male and elderly with a median age of onset of 68 years.

The disease is less frequent in Asian countries.

Most cases are advanced at presentation and often exhibit complete responses to initial treatment followed by frequent relapses 

This neoplasm is characterized by mature B-lymphocytes that infiltrate the lymph nodes, bone marrow, peripheral blood, and extranodal sites.

Histologically it appears homogenous with a background of pink histiocytes that stain positively for cyclin D1, surface immunoglobulin B-cell markers, and CD5 

The t (11;14) (q13;q32) translocation is present in over 90% of MCL, and other translocations have been reported. 

Disease heterogeneity features: a blastoid variant with a high Kiel 67 (Ki67) proliferation index, as well as a more indolent variant that can be appropriately managed with a watch and wait approach until symptomatic.

Younger, fit patients can be managed with intensive cytarabine-containing chemotherapy alternating with hyperfractionated cyclophosphamide, vincristine, doxorubicin, and dexamethasone combined with rituximab (R-HyperCVAD) with or without autologous stem cell transplantation for consolidation. 

Other patients receive CHOP-like (cyclophosphamide, doxorubicin, vincristine, prednisone) therapy plus rituximab. 

Substitution of vincristine with the proteasome inhibitor bortezomib has emerged as a validated treatment for both relapsed and untreated disease.

Awareness of prognostic factors through the validated MIPI score, identification of potential transplant candidates, and careful assessment of response to and tolerance of immunochemotherapies, are all crucial determinants of both survival and quality of life.

MCL can be distinguished by immunohistochemistry (IHC) with a profile usually showing positivity for B-cell markers as well as CD5, FMC7, CD43, and cyclin D1. 

Most cases are negative for CD10 and CD23. 

The reciprocal translocation t (11;14) results in the overexpression of cyclin D1,

In less than 5% of cases, cyclin D1 and t (11;14) may be negative in MCL if there is an otherwise typical IHC profile, with over half of these cases revealing rearranged CCND2 and consequent overexpression of cyclin D2 mRNA,

Most pathologic findings and clinical features of cyclin D1-negative MCL appear similar to those of cyclin D1-positive cases and are managed the same.

The pathogenesis of mantle cell lymphoma (MCL) occurs through two major events that cause a gain and loss of function. 

Chromosome 11q13 holds the proto-oncogene CCND1, which encodes cyclin D1, and its translocation to chromosome 14q32, the locus of the immunoglobulin heavy chain complex (IGH), deregulates the cell cycle at the G1/S phase transition and makes cyclin D1 constitutively overexpressed in otherwise normal B lymphocytes. 

Gain of function occurs via t (11;14) translocation, causing BCL-1 over-transcription. 

Loss of function occurs via 11q22-23 deletion, alters the DNA damage pathway response. 

Cyclin D1 protein has a major oncogenic effect through two different mechanisms.

MCL carries a high degree of genomic instability, with secondary chromosomal alterations. 

MCL is thought to derive from naïve B-cells in the pre-germinal center (mantle zone) since the initial translocation event t (11;14) occurs during recombination of the V(D)J segments of the IGH variable region (IGHV) in the bone marrow. 

Antigen selection is now thought to play an important role in pathogenesis for the 15-40% of MCLs that carry IGHV hypermutations similar to those in chronic lymphocytic leukemia.

Cyclin D1 binds to CDK4 and CDK6, which phosphorylates retinoblastoma 1 (RB1), thereby activating the transcription factor E2F while promoting cyclin E/CDK2 activation to trigger entry into the S phase of the cell cycle.

The round cell variant has a CLL-like clinical presentation that tends to behave more like an indolent lymphoma, usually showing a rather low cell proliferation (Ki-67 roughly 10%). 

A blastoid variant occurring 5% of the time shows a much more aggressive clinical course. 

Biological factors like high cell proliferation (determined by Ki-67 staining) or p53 mutations and p16 deletions are closely related to this MCL subtype. 

More than 80% of MCL still presents with somewhat intermediate characteristics.

Prognostic marker system categorize MCL patients:

relative survival probability by grading the following risk factors: age, performance status, lactate dehydrogenase (LDH) level, and white blood cell (WBC) count. 

A MIPI combined biologic index (MIPIb) score of less than 5.7 classifies patients into a low-risk group comprising 44% of patients with a median overall survival (OS) not reached; a score of 5.7 to less than 6.2 yields an intermediate-risk group comprising 35% of patients with median OS of 51 months; and a score of 6.2 or greater falls into the high-risk group with 21% of the patients and median OS of 29 months. 

The number of extranodal sites is not an independent prognostic factor in the MIPI score. 

Tumor cell proliferation, assessed on by Ki-67, showed a median value of 14.5% (range 1.2–91%). 

Ki-67 value is prognostically significant using a cutoff point of 10%, and is independent of the MIPI. 

Ki-67 of 30% has been proposed by the NCCN as a cutoff for determining the aggressiveness of the disease.

MIPIb Index Score Risk and overall survival:

<5.7 Low risk 44% Not reached

5.7 to 6.2 Intermediate risk 35% 51 months

6.2 High risk 21% 29 months

Localized presentation, which is extremely rare, can be managed with observation, radiation therapy, or a combination of radiation and chemotherapy. 

The use of radiotherapy with or without chemotherapy engendered significantly better progression-free survival (PFS) at 5 years (73% vs. 13%) with a trend towards overall survival benefit, when compared to patients who did not receive radiation.

Radiotherapy as a primary treatment for stage 1-2 MCL patients analyzed retrospectively showed curative results for 3.6% of patients, with 3-year OS of 93%.

Bulky Stage II, Stage III, and Stage IV

In highly select cases in which the patient is asymptomatic, advanced disease can also be managed with a watch and wait approach.

R-HyperCVAD (rituximab, cyclophosphamide, vincristine, doxorubicin, and dexamethasone alternating with high-dose methotrexate and cytarabine) is considered a preferred aggressive regimen in patients who can tolerate considerable toxicity

A phase II study of 97 treatment-naïve patients with advanced MCL showed failure-free survival of 64% and overall survival of 82% at 3 years, and median overall survival not yet reached at 10 years follow-up

Newly diagnosed patients under the age of 70 who received R-HyperCVAD yielded PFS of 4.8 years and OS of 6.8 years, with 2-year PFS of 63% and OS of 76%.

In a phase III randomized trial of the German Low Grade Lymphoma study group that evaluated the addition of rituximab to CHOP chemotherapy (cyclophosphamide, doxorubicin, vincristine, prednisone) in treatment-naïve patients with advanced MCL aged 65 and younger. 

R-CHOP showed significantly better overall response rate (94% vs. 75%), complete remission rate (34% vs. 7%) and median time to treatment failure (21 months vs. 14 months) than CHOP. 

However, PFS and OS outcomes were not superior. R-CHOP can also be alternated with R-DHAP (rituximab, dexamethasone, cisplatin, cytarabine) or R-ICE (rituximab, ifosfamide, carboplatin, etoposide) in sequence.

For elderly patients or those with poor performance status, bendamustine plus rituximab (BR) has shown superior PFS than R-CHOP (35 months vs. 22 months.

Another less aggressive induction regimen to gain FDA approval is VR-CAP (bortezomib, rituximab, cyclophosphamide, doxorubicin, prednisone): When compared to R-CHOP in patients who underwent front-line VR-CAP showed superior PFS (24.7 vs. 14.4 months, p<0.001) and CR rate (48% vs. 41%) with a trend towards OS benefit (64% vs. 54%) in a phase III randomized study.

Following successful induction with R-HyperCVAD or CHOP-like immunochemotherapy, high-dose therapy (HDT) with one cycle of etoposide, cytarabine, and rituximab and one cycle of carmustine, etoposide, and cyclophosphamide has been effectively followed by autologous stem cell rescue.

Thirty-three patients treated after achieving first remission with HyperCVAD with or without rituximab achieved 5-year disease-free survival of 42% and OS of 77%with 100% OS rate in those patients with a low serum beta-2 microglobulin level. 

Long-term follow-up showed a median PFS of 42 months and OS of 93 months.

There is improved PFS in transplanted patients who were induced with HyperCVAD (with or without rituximab) rather than CHOP (with or without rituximab).

Post-induction maintenance therapy with rituximab has been shown to provide extended disease control for patients who cannot undergo high-dose therapy and stem cell transplantation. 

Additionally studies have also shown promising data for rituximab maintenance after R-CHOP induction.

It remains unclear whether first-line consolidation with high-dose chemotherapy with autologous stem cell rescue (HDT/ASCR) provides an advantage over rituximab maintenance in patients of any age. 

Second-line consolidation therapy can be pursued with autologous or allogeneic stem cell transplantation (SCT) following non-myeloablative or myeloablative conditioning regimens. 

Graft-versus-host disease (GVHD) prophylaxis consisted of a combination of cyclosporin or tacrolimus with mini-dose methotrexate and/or methylprednisolone. 

Allo-SCT in 44 patients resulted in lower recurrence rates but higher treatment-related mortality rates than the 68 patients who underwent high-dose therapy with autologous stem cell transplantation.

The improved OS among the allogeneic SCT group was not statistically significant but the improved DFS was significant.

Regimens suitable for use in first relapse include: bendamustine with or without rituximab, . Cladibrine, FC (fludarabine plus cyclophosphamide) with or without rituximab, FCMR (fludarabine, cyclosphamide, mitoxantrone, and rituximab), FMR (fludarabine, mitoxantrone, and rituximab), PCR (pentostatin, cyclophosphamide, and rituximab) and PEPC (prednisone, etoposide, procarbazine, cyclophosphamide with or without rituximab.

Bortezomib is approved for relapsed or refractory MCL based on the phase II PINNACLE trial, in which bortezomib induced an overall response rate of 33% (CR in 8%), with a median duration of response of 9 months and median time to progression of 6 months.

Bortezomib combined with rituximab has shown activity in heavily pre-treated patients with relapsed/refractory disease.

The immunomodulating agent, lenalidomide, has shown efficacy in relapsed or refractory aggressive non-Hodgkin lymphoma, with subset analysis of MCL patients showing an ORR of 35% and CR rate of 12% at a median follow-up of 12 months in a phase II trial.

In this study the duration of response was 16 months, and the median PFS was approximately 9 months.

The small-molecule Bruton’s tyrosine kinase (BTK) inhibitor, ibrutinib, has shown promising activity in several B-cell malignancies.

 A multicenter phase II trial of 111 patients who had been previously treated with bortezomib and/or rituximab-containing regimens, 72% of whom had advanced disease and 42% of whom had high-risk MIPI scores, showed an ORR of 68% with CR rate of 21% after 15 months of ibrutinib.

The median duration of response was 17.5 months, median PFS was 14 months, and estimated OS rate at 18 months was 58%. 

The response rates appeared to increase with longer duration of therapy. 

The most common adverse event rated grade ≥3 were neutropenia (16%), thrombocytopenia (11%), anemia (10%), pneumonia (6%), diarrhea (6%), fatigue (5%) and dyspnea (5%). 

The use of ibrutinib causes a transient lymphocytosis that resolves after an average of 8 weeks, and caused grade ≥3 bleeding events in 5% of patients.

Ibrutinib can extend the lives of heavily treated, relapsed, or refractory patients with mantle cell lymphoma (MCL), many become resistant to the drug. 

30% of the patients who developed resistance to ibrutinib expressed high levels of activated phosphatidylinositol-3 kinase (PI3K-AKT) and cyclin-dependent kinase 4 (CDK4). 

Analysis of longitudinal functional genomics in MCL showed that 30% of the patients who developed resistance to ibrutinib expressed high levels of activated phosphatidylinositol-3 kinase (PI3K-AKT) and cyclin-dependent kinase 4 (CDK4): PI3K-AKT proteins promote survival; CDK4 drives MCL cells through the cell cycle.

These two mechanisms appeared to override ibrutinib’s inhibitory action in resistant cells. 

Stepwise treatment with palbociclib followed by ibrutinib, or palbociclib followed by the PI3K pathway inhibitor, idelalisib, which is FDA-approved to treat CLL, might treat MCL patients who initially do not respond to ibrutinib. 

Idelalasib evaluated in patients with relapsed or refractory mantle cell lymphoma patients who had received a median of four prior therapies. 

Overall, nine (18%) patients discontinued therapy due to adverse effects including diarrhea, transaminase elevations, pneumonia, and acute renal failure. 

Thirty-three (84.6%) patients had some reduction in lymph node size, with CR in two (5%) patients, PR in 14 (35%) patients, and stable disease in nineteen (47.5%) patients.

Bortezomib Phase II PINNACLE trial  with 33% response rate and median duration of response of 9 months, with a median time to progression of 6 months .

Lenalidomide Phase II Multicenter Study  with 35% response rate and duration of response was 16 months.

Mantle cell lymphoma is an incurable but increasingly well characterized clinicopathologic entity that unfortunately poses persistent obstacles in achieving durable responses with available treatment regimens. 

Early stage disease is rare, but limited available data suggests that radiation with or without chemotherapy can effectively manage limited, non-bulky distribution. 

For advanced disease can be managed up-front with either observation, R-HyperCVAD, CHOP-like chemotherapy, or less aggressive induction regimens including bendamustine and rituximab. 

Relapsed and refractory disease carries a poorer prognosis, but novel agents including bortezomib, lenalidomide, and ibrutinib have shown promising efficacy and tolerability.

Leave a Reply

Your email address will not be published. Required fields are marked *