throbber
Critical Reviews in Oncology/Hematology 58 (2006) 257–265
`
`Treatment of mantle cell lymphoma:
`Current approach and future directions
`∗
`
`Joshua Brody, Ranjana Advani
`
`Department of Medicine, Division of Oncology, Stanford University Medical Center, Clinical Cancer Center,
`875 Blake Wilbur Drive, Stanford, CA 94305, USA
`
`Accepted 5 October 2005
`
`Contents
`
`1. Diagnosis. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
`2.
`Prognosis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
`3. Treatment approaches . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
`3.1. Chemotherapy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
`3.2. Rituximab . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
`3.3. Autologous stem cell transplant. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
`3.4. Role of purging . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
`3.5. Allogeneic transplantion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
`3.6. Radioimmunotherapy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
`3.7. Other immunotherapies . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
`3.8. Newer agents . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
`Summary and future directions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
`Reviewers . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
`References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
`Biographies . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
`
`4.
`
`258
`258
`258
`259
`259
`260
`261
`261
`262
`262
`262
`262
`263
`263
`265
`
`Abstract
`
`Although mantle cell lymphoma has been described as “moderately aggressive” it has become clear that it carries a worse long-term prognosis
`than other subtypes of non-Hodgkin’s lymphoma. In recent years, this has prompted numerous clinical trials of novel and more aggressive
`therapies in hopes of impacting these poor outcomes. These include more intensive combination chemotherapy regimens, monoclonal antibody
`therapy in conjunction with other treatments or conjugated to radioactive isotopes, high-dose chemotherapy followed by autologous or
`allogeneic stem cell transplantation, and newer targeted therapies based on increasing understanding of the molecular pathways of this
`malignancy.
`© 2005 Elsevier Ireland Ltd. All rights reserved.
`
`Keywords: Mantle cell lymphoma; Clinical trials; Non-Hodgkin’s lymphoma
`
`Mantle cell lymphoma (MCL) is a distinct non-Hodgkin’s
`lymphoma (NHL) subtype first put forward by an interna-
`tional consensus panel [1] in 1992 and then incorporated into
`
`∗
`
`Corresponding author. Tel.: +1 650 723 1479; fax: +1 650 725 9113.
`E-mail addresses: josho@stanford.edu (J. Brody),
`radvani@stanford.edu (R. Advani).
`
`the REAL-WHO classification system in 1994 [2]. It com-
`prises a group of subtypes including those previously classi-
`fied as centrocytic, lymphocytic or diffuse small-cleaved cell
`lymphoma. This subtype represents approximately 4–6% of
`all non-Hodgkin lymphomas [3,4] with an incidence of 2–3
`per 100,000 years and with a median age at diagnosis of 63
`years. Diagnosis is based on histologic, immunophenotypic,
`
`1040-8428/$ – see front matter © 2005 Elsevier Ireland Ltd. All rights reserved.
`doi:10.1016/j.critrevonc.2005.10.001
`
`IPR2018-00685
`Celgene Ex. 2046, Page 1
`
`

`

`258
`
`J. Brody, R. Advani / Critical Reviews in Oncology/Hematology 58 (2006) 257–265
`
`cytogenetic and molecular criteria, most importantly the char-
`acteristic t(11;14) (q13;q32) translocation which places the
`immunoglobulin heavy chain locus upstream of the BCL1
`gene causing overexpression of its gene product—cyclin D1.
`In a series of 81 patients diagnosed from 1988 to 1990 MCL
`carried the worst 5-year failure free survival rate of any of the
`major subtypes (∼11%) and median overall survival of less
`than 3 years [5].
`Because of this poor prognosis there has been an ongo-
`ing search for improved treatments. In contrast to the more
`common aggressive lymphomas (e.g. diffuse large B-cell
`lymphoma (DLBCL)) there is not one regimen that has
`demonstrated superiority or equivalence compared to mul-
`tiple alternatives, hence there is little agreement amongst
`clinicians regarding appropriate first-line therapy. The pur-
`pose of this review is to consider the rationale and available
`data for commonly used treatment regimens and to survey
`promising future therapies currently being developed and
`tested.
`
`1. Diagnosis
`
`Selection of appropriate treatment regimens for patients
`with MCL and the pursuit of continually better regimens
`relies upon uniformity of diagnostic criteria for this entity.
`Clinically, MCL has a male predominance and a tendency for
`extra-nodal involvement including the bone marrow, spleen
`and GI tract (particularly the colon). The histologic pattern
`may be diffuse, nodular, mantle-zone, or a combination of
`the three with some reports suggesting a better prognosis
`for those with a mantle-zone pattern [6]. The malignant cell
`type of classic MCL is composed of small to medium sized
`lymphocytes with irregular nuclei and condensed chromatin,
`though there also exists a broad spectrum of morphologic fea-
`tures ranging from small cell to blastoid types and these may
`reflect distinct biologic characteristics. Patients can present
`de novo or during the course of their disease with a blastoid
`variant composed of medium size rounded nuclei with dis-
`persed chromatin, scant cytoplasm and a high mitotic index,
`resembling lymphoblasts. Interestingly, one series found that
`although this “transformation” was found in 32% of patients
`during the course of disease, 70% of autopsy specimens
`revealed some site of blastoid disease [7].
`The immunophenotype of MCL corresponds to mature,
`na¨ıve pre-germinal center B cells with expression of CD19,
`CD20, CD22, CD79A, IgM and/or IgD. They are usually
`−
`−
`CD5+ and CD43+, but CD10
`and CD23
`. Though there
`are exceptions, this basic schema can be helpful in distin-
`guishing between the common small B-cell NHL subtypes
`(see Table 1).
`The most distinctive aspects of MCL are its numerous
`genetic aberrations that are important both in its diagnosis and
`in its pathophysiology. The most pathognomonic of these is
`t(11;14) (q13;q32) translocation. Other well-described muta-
`tions include loss of the ataxia telangiectasia mutated (ATM)
`
`Table 1
`Immunophenotype of small B-cell NHL subtypes [73]
`
`SLL/CLL
`
`MCL
`
`Follicular
`
`MZL
`
`CD20
`CD5
`CD23
`CD43
`CD10
`
`+
`+
`+
`−
`+
`+
`−
`−
`+
`+
`+
`+
`−
`−
`−
`Abbreviations: SLL/CLL, small lymphocytic lymphoma/chronic lympho-
`cytic leukemia; MZL, marginal zone lymphoma.
`
`+
`−
`−
`−
`+
`
`gene (frequently through 11q deletion), deletions of 9p21
`and 17p13 (thought to be important for the loss P16 and
`P53, respectively) and 8q22–24 amplification (thought to be
`important for its consequent over-expression of c-myc) [8].
`The t(11;14) translocation juxtaposes the BCL1 gene to the
`B-cell immunoglobulin transcription enhancer and results in
`the over-expression of cyclin D1 and cell-cycle dysregula-
`tion. In approximately 50% of cases the translocation occurs
`in one identified region called the major translocation cluster.
`In most of these cases PCR can be used to diagnose and (more
`importantly) to follow response to treatment at the molecu-
`lar level. Generally fluorescence in situ hybridization (FISH)
`assessment for the translocation is the gold standard when
`definitive genetic testing is required. This technique is 95%
`sensitive for the specific translocation and can be performed
`on paraffin fixed specimens.
`
`2. Prognosis
`
`There are numerous clinical and pathologic parameters
`of negative prognostic significance including: age, poor per-
`formance status [9], splenomegaly, nodal (as opposed to
`non-nodal leukemic) disease [10], anemia, non-mantle zone
`histology [6], blastoid morphology, high mitotic index [11],
`topoisomerase II␣ [12], cyclin D1 [13] and c-myc [14] over-
`expression. More recently, it has been shown that specifically
`VH3-21 gene rearrangement (compared to other gene rear-
`rangements) correlates with an improved prognosis [15].
`Also, RNA expression profiling, has identified gene sets,
`which can prognosticate MCL patients more accurately than
`clinical parameters (such as morphology) [16]. In the lat-
`ter study, genes associated with a high proliferation index
`were associated with aggressive disease and a shorter sur-
`vival. In this study, patients could be stratified into subsets
`with median survival differences of greater than 5 years. Cer-
`tainly some of these parameters will be helpful in the future to
`guide appropriate therapy for each patient; however, there has
`been no evidence so far that higher risk groups have greater
`benefit from more aggressive treatment regimens.
`
`3. Treatment approaches
`
`Considering the poor prognosis of all current therapies
`there is currently no standard treatment for the disease. Treat-
`
`IPR2018-00685
`Celgene Ex. 2046, Page 2
`
`

`

`J. Brody, R. Advani / Critical Reviews in Oncology/Hematology 58 (2006) 257–265
`
`259
`
`Table 2
`Summary of mantle cell lymphoma treatment options
`
`Treatment
`
`CHOP [28]
`R-CHOP [28,29]
`
`HCVAD (>65 years) [20]
`R-HCVAD [31]
`HCVAD-ASCT [21]
`◦
`
`ASCT [38] 1
`therapy
`◦
`therapy
`and 2
`ASCT (GELA)[37] 1
`◦
`therapy
`ASCT–R-HDS [46] 1
`◦
`◦
`and 2
`ASCT (EORTC) [40] 1
`◦
`Bexxar®-ASCT [58] 2
`therapy
`◦
`therapy
`Allo BMT [49] 2
`◦
`NMA-allo [52,52] 2
`therapy
`
`◦
`
`therapy
`
`N
`
`60
`62
`40
`25
`97
`45
`
`62
`24
`28
`195
`16
`16
`18
`33
`
`ORR (%)
`
`CR (%)
`
`PFS/EFSa
`
`75
`94
`96
`92
`100
`94
`
`98
`100
`100
`88
`100
`100
`100
`85
`
`7
`34
`48
`68
`87
`38
`
`81
`79
`100
`67
`91
`86
`94
`75
`
`19 m
`20 m
`16.6 m
`15 m
`64%a
`◦
`: 72%a
`1
`◦
`: 17%a
`2
`39 m
`55%a
`79%a
`33%
`61%
`55%
`82%a
`60%a
`
`OS
`
`82%
`84%
`
`92%
`82%
`◦
`1
`: 92 %
`◦
`2
`: 25 %
`83%
`68%
`89%
`50 %
`93 %
`55%
`86%
`65%
`
`Median follow-up
`
`18 m
`18 m
`25 m
`17 m
`3 y
`3 y
`
`3 y
`3 y
`35 m
`5 y
`3 y
`3 y
`26 m
`24.6 m
`
`CHOP: cyclophosphamide, adriamycin, vincristine, prednisone; R: rituximab; HCVAD: high-dose cytarabine, cyclophosphamide, vincristine, adriamycin,
`◦
`◦
`dexamethasone; 1
`therapy: initial treatment; 2
`therapy: treatment of relapsed or refractory disease; HDS high-dose cyclophosphamide, high-dose cytarabine,
`high-dose melphalan and high-dose mitoxantrone plus melphalan; NMA: non-myeloablative.
`a For those timepoints measuring EFS where events are usually defined as progression or death from any cause.
`
`ment options are varied and in general the more aggressive
`approaches improve progression free survival (PFS) how-
`ever, no therapy to date is curative. Various approaches are
`summarized in Table 2.
`
`3.1. Chemotherapy
`
`Though available data have not suggested an optimal first
`line treatment for MCL, the standard of care has developed
`from the understanding that the clinical course of MCL is
`aggressive; treatments have been similar to those for other
`aggressive lymphomas. An anthracycline-based approach
`has been standard although most randomized clinical stud-
`ies have not proven a survival advantage attributable to the
`inclusion of anthracycline [17–19].
`Before the introduction of the monoclonal antibody rit-
`uximab, the most commonly used first line regimen was
`cyclophosphamide, adriamycin, vincristine and prednisone
`(CHOP), which has shown complete response (CR) rates
`ranging 20–80%, median failure free survival (FFS) of 10–16
`months, and median overall survival (OS) of 3 years. Other
`approaches have used dose intensified, CHOP-like regimens.
`Several trials at the MD Anderson Cancer Center have uti-
`lized the intensive leukemic regimen hyper-CVAD (a dose
`intense, hyper-fractionated CHOP-like combination in com-
`bination with high-dose methotrexate (MTX) and cytosine
`arabinoside (ara-C)) both as primary and as salvage therapy.
`The number of cycles of therapy has varied from four to
`eight depending on whether or not stem cell transplant was
`included as primary treatment. In a study of primary ther-
`apy for 25 patients over age 65 years, overall response rates
`(ORR) of 92%, CR rates of 68% and median duration FFS of
`15 months were reported [20]. Another study of 45 patients
`with advanced stage MCL (50% previously treated) received
`four cycles of hyper-CVAD and patients that achieved a CR
`
`went on to autologous stem cell transplant (ASCT) or allo-
`geneic transplant. The ORR was 93.5% with 38% of patients
`achieving a CR [21]. Previously untreated patients had a 3-
`year OS and event free survival (EFS) of 92% and 72%,
`respectively, significantly higher than CHOP-treated histori-
`cal controls with 3-year OS and EFS rates of 25% and 17%.
`Although it is difficult to dissect the impact of hyper-CVAD
`from that of the preparative regimen for ASCT (cyclophos-
`phamide and total body irradiation) it does seem that this
`intensified regimen is at least as effective for primary or sal-
`vage therapy as its predecessor. It must be emphasized these
`results represent single-institution data and have not been
`validated by other centers or in a randomized study.
`Another approach has been to incorporate regimens used
`in salvage therapy of other lymphoma subtypes as part of a
`primary treatment regimen for mantle cell. In a phase II, trial
`of 28 patients with aggressive MCL, four cycles of CHOP
`induced CR in only 7%. Remarkably, for those patients with
`a partial response, the addition of DHAP to CHOP in a
`sequential fashion induced a complete response in 84% of
`the remaining patients compared to CHOP alone [22].
`Despite the lack of randomized studies evaluating the dif-
`ferent treatment regimens for MCL, it appears that newer,
`more intensive approaches may result in superior ORR com-
`pared to CHOP or CHOP-like therapy. The superiority of
`these regimens may be due to the use of high-dose AraC,
`which is therefore being tested in ongoing clinical trials.
`Whether the improved remission rates translate into
`improved survival is unclear as the median follow-up of all
`these studies in short (<3 years). Patients should be enrolled
`on clinical trials in order to evaluate these new approaches.
`
`3.2. Rituximab
`
`Because MCL is a B-cell malignancy that expresses CD20,
`the anti-CD20 antibody (Ab), rituximab, has been studied
`
`IPR2018-00685
`Celgene Ex. 2046, Page 3
`
`

`

`260
`
`J. Brody, R. Advani / Critical Reviews in Oncology/Hematology 58 (2006) 257–265
`
`as a single agent in mantle cell lymphoma with response
`rates of 20–40% [23–26]. As in other lymphoma subtypes,
`it has also been studied as longer term maintenance ther-
`apy. A randomized trial followed 61 MCL patients who did
`not progress during rituximab induction therapy assigned to
`either maintenance (every 2 months for four treatments) rit-
`uximab or observation [27]. In this trial, maintenance therapy
`showed a statistically non-significant trend towards increased
`EFS of 12 months versus 6 months. More commonly, rit-
`uximab has been studied in combination with chemother-
`apy such as CHOP (i.e. R-CHOP). A prospective study by
`the German Lymphoma Study Group (GLSG) randomized
`untreated patients with advanced disease to conventional
`CHOP or R-CHOP. Responding patients received six cycles
`of CHOP or R-CHOP followed by a second randomization
`to ␣-interferon maintenance or a myeloablative consolida-
`tion followed by ASCT (depending on the patient’s age). Of
`the 122 patients evaluated prior to transplant, R-CHOP was
`resulted in a significantly superior ORR and CR rates com-
`pared to CHOP alone: 94% versus 75% (p = 0.005) and 34%
`versus 7% (p = 0.00024) [28]. Although the addition of ritux-
`imab increased time to treatment failure (TTF) from 14 to 21
`months, median PFS and OS were not statistically improved.
`The lack of an effect on OS is possibly due to the confounding
`effect of the second randomization.
`Similar results were achieved in an earlier phase II study
`in which R-CHOP yielded ORR and CR rates of 96% and
`48% with a median PFS of 16.6 months [29]. However, in
`the latter study patients obtaining a molecular CR did not
`have significantly improved PFS.
`The GLSG have also recently updated preliminary results
`of a prospective randomized trial of rituximab in combination
`with fludarabine, cyclophosphamide, mitoxantrone (FCM)
`compared to FCM alone in patients with relapsed or refrac-
`tory MCL. Of 50 patients randomized, R-FCM achieved
`higher CR and overall median survival times: 29% versus
`0% (p = 0.004) and 2.5 years versus 0.9 years (p = 0.031)
`[30]. After the superiority of the rituximab arm was demon-
`strated, an additional 45 patients were assigned to combined
`immunotherapy and confirmed these promising results.
`Rituximab has also been studied in phase II trials with
`more aggressive regimens such as hyper-CVAD. In 56
`untreated patients, R-hyper-CVAD for at
`least 6 cycles
`yielded a CR rate of 90% and 2-year FFS and OS were 72%
`and 90% (median follow-up of 14 months). Subset analy-
`sis revealed that patients younger than 65 years treated with
`R-hyper-CVAD without ASCT did as well historical con-
`trols receiving the same hyper-CVAD regimen with ASCT.
`In a recent update of this trial on 97 patients the 3-year FFS
`and OS were 64% and 82%, respectively [31]. The toxicity
`of this regimen, is of concern as five deaths were reported
`during treatment and four occurrences of myelodysplasia or
`leukemia developing in patients during CR. More recently, a
`modified hyper-CVAD regimen that added rituximab induc-
`tion and maintenance therapy but removed MTX and ara-C
`was shown to have comparable efficacy as primary therapy
`
`with ORR of 85%, CR of 70% and median progression free
`survival (PFS) and OS not reached after 22.5 months [32].
`While the results of this modified approach compare favor-
`ably with the original regimen from MD Anderson, longer
`follow-up along with validation by other centers will be
`required. Taken together, these data suggest that the addi-
`tion of rituximab to standard induction regimens appears to
`result in improved response rates.
`Two studies have suggested that post-transplant rituximab
`increases the clinical and molecular response rate of MCL
`patients receiving ASCT [33,34]. In one study of advanced
`stage MCL all patients who received post-transplant ritux-
`imab were alive without clinical or molecular relapse at 239
`days post-transplant. The treatment was well tolerated and
`encouraging albeit longer follow-up is needed.
`
`3.3. Autologous stem cell transplant
`
`Myeloablative dose chemotherapy followed by stem cell
`transplantation can improve survival in other subtypes of
`lymphoma. This treatment has also been applied to MCL
`with mixed results [35–37]. Most studies have utilized this
`approach in patients with relapsed or refractory disease.
`Recently, the European MCL Network reported the results
`of the first prospective randomized trial comparing myeloab-
`lative radio-chemotherapy versus ␣-interferon maintenance
`therapy in 122 patients that achieved at least a PR after a
`CHOP-like induction regimen [38]. ASCT consisted of a
`mobilization regimen of dexamethasone, BCNU, etoposide,
`ara-C and melphalan (dexa-BEAM) followed by consol-
`idative therapy with total body irradiation (TBI) and high-
`dose cyclophosphamide (CTX). Compared to the IFN-␣
`arm, patients in the ASCT arm experienced a significantly
`longer PFS with a median of 39 months versus 17 months
`(p = 0.0108), but no difference in 3-year OS: 83% versus 77%
`(p = 0.18).
`Similar outcomes have been described in other mature
`phase II studies; with a median follow-up of four years,
`a trial of 25 patients were treated with rituximab plus
`induction chemotherapy followed by high-dose chemother-
`apy ± radiotherapy and ASCT [39]. Induction allowed all
`patients to achieve either CR (36%) or PR (64%) and 3 year
`OS is 80%.
`Data from other trials have also suggested that ASCT is
`of greater benefit earlier in the course of disease. A retro-
`spective analysis of the European Blood and Bone Marrow
`Transplant registries of 195 MCL patients treated with ASCT
`reported OS at 2- and 5-year as 76% and 50% with PFS as
`55% and 33%, respectively [40]. Patients who were trans-
`planted in first CR were 33% less likely to die from MCL
`than patients with chemosensitive disease transplanted later
`in their course. Results were similar in a retrospective analysis
`of 69 patients at Stanford and City of Hope who underwent
`ASCT [41]. Patients who were in first CR at the time of
`transplant had 3- and 5-year OS/DFS rates of 93%/74% and
`77%/50%, respectively. In comparison, the OS/DFS rates at
`
`IPR2018-00685
`Celgene Ex. 2046, Page 4
`
`

`

`J. Brody, R. Advani / Critical Reviews in Oncology/Hematology 58 (2006) 257–265
`
`261
`
`3 and 5 years for patients who were not in first remission
`at the time of transplant were 64%/51% and 39%/21%. The
`median time to relapse in the group transplanted in first CR
`was 32 months compared to 10.5 months.
`As described above, hyper-CVAD remission induction for
`MCL has yielded 3 year OS and EFS rates were 92% and
`72% [21]; these compare favorably with the randomized trial
`described using CHOP or CHOP-like regimens [38]. A ret-
`rospective analysis by Conde et al. evaluated the induction
`regimens used for MCL [42,43]. An international database
`of 119 patients with MCL who had received ASCT between
`1988 and 2002 was evaluated. The induction regimens were
`primarily hyper-CVAD and CHOP-like therapy and the esti-
`mated 10-year OS and DFS were impressive at 50% and 32%,
`respectively. Patients receiving hyper-CVAD had a 4-year
`DFS of 68% compared to 33% in patients treated with other
`regimens. It should be noted that none of these regimens
`employed rituximab along with the induction chemother-
`apy. Though there were differences in baseline characteristics
`between these two groups, these data suggest the need to
`evaluate the role of these newer therapies in prospective ran-
`domized trials.
`
`3.4. Role of purging
`
`As in other B-cell malignancies, the role of “purging” of
`the mobilized autologous stem cell product prior to trans-
`plant has been examined in MCL. One such approach is
`the use of ex vivo treatment of stem cell products with anti-
`CD20 Ab and complement-mediated lysis. A study assessing
`for minimal residual disease (MRD) at the time of trans-
`plant using PCR amplification of the Bcl-1/IgH translocation
`product showed ex vivo purging to be effective in only 2
`of 19 patients [44]. Alternatively, the use of in vivo purging
`using rituximab has been more promising with evidence of
`clinical and molecular remissions. The first of these studies
`demonstrated that stem cell products collected after high-
`dose cyclophosphamide and cytarabine still had a high rate
`of MRD versus a parallel cohort that received rituximab after
`high-dose therapy (60% versus 7%; p = 0.007) [45]. The sig-
`nificance of this study for MCL treatment is limited by its
`inclusion of non-MCL sub-types of NHL. A follow-up study
`treated 28 MCL patients with cisplatin or doxorubicin-based
`debulking chemotherapy followed by high-dose sequential
`therapy with rituximab and ASCT [46]. The 54-month OS
`and EFS rates were 89% and 79%, respectively as com-
`pared to historical controls with 42% and 18%. Again, after
`cyclophosphamide, cytarabine and rituximab mobilization,
`19 out of 19 evaluable patients had stem cell products that
`were without MRD. Adding cytarabine with rituximab in vivo
`purging to a CHOP-like regimen compared to the latter regi-
`men alone, showed benefit both in pre-transplant CR rate and
`post-transplant PCR negativity [47].
`Cumulatively these data suggest that in addition to its
`above noted uses in remission induction and salvage therapy,
`rituximab also may have an important pre-transplant role both
`
`as part of conditioning regimens and for in vivo. Again, none
`of these studies have a long enough follow-up to assess the
`impact on overall survival.
`
`3.5. Allogeneic transplantion
`
`Although ASCT has demonstrated prolonged survival for
`patients with MCL, there has been no evidence of cure with
`these modalities so more aggressive approaches are still being
`evaluated. As allogeneic transplantation has demonstrated
`increased effectiveness in other lymphoma sub-types because
`of a graft-versus-lymphoma (GVL) effect [48], this approach
`has been considered for MCL patients refractory to other ther-
`apies (or occasionally those in first CR). Along with degree
`and duration of clinical response, evaluation for evidence of
`this GVL effect has been an important outcome in such trials.
`In an early study, 16 patients underwent allogeneic trans-
`plant after myeloablative regimens of cyclophosphamide plus
`TBI (11 patients) or BEAM (3 patients) or a non-ablative
`preparative regimen consisting of cisplatin, cytarabine and
`fludarabine [49]. OS and failure-from-progression (FFP) at
`3 years were both 55% (28–83%) including a high treatment
`related mortality of 38%. The suggestion of a GVL effect
`was evident in several patients who converted to a molec-
`ular CR months after the completion of chemotherapy and
`coincident with the development of graft-versus-host dis-
`ease (GVHD). Another trial using a myeloablative regimen
`in 20 MCL patients has reported nine patients alive and dis-
`ease free 1–9 years post-transplant [50]. This and other [51]
`reports of long-term DFS raise the question as to whether this
`approach has curative potential for some patients.
`Because MCL occurs more in older patients who are not
`candidates for myeloablative allogeneic transplants, there has
`been consideration of non-myeloablative regimens followed
`by allogeneic transplant (NMA-allo). Early results have been
`mixed. A recent trial investigated non-myeloablative trans-
`plant in 18 patients with MCL who had failed multiple prior
`chemotherapies including 28% who had failed prior ASCT.
`CR was achieved in 17 of 18 patients and with a median
`follow-up of 26 months the estimated 3-year survival rate and
`current PFS was 85.5% and 82%, respectively [52]. Another
`study of 33 MCL patients using a non-myeloablative con-
`ditioning regimen of fludarabine and 2 Gy TBI followed by
`related or unrelated allogeneic transplantation demonstrated
`2-year OS and DFS of 65% and 60% with a non-relapse mor-
`tality of 24% [53]. Still, the outcomes for this approach have
`been mixed as a larger trial of non-myeloablative chemother-
`apy followed by allogeneic transplant included 22 patients
`with MCL and yielded 2-year OS and PFS of 12% and 0%
`[54].
`These data plus other recent retrospective analyses [55,56]
`suggest that myeloablative conditioning or, in older patients,
`non-myeloablative conditioning, followed by allogeneic stem
`cell transplantation is feasible for MCL patients in first CR
`and after salvage therapy. The high treatment related mor-
`tality rates are consistent with results from other lymphoma
`
`IPR2018-00685
`Celgene Ex. 2046, Page 5
`
`

`

`262
`
`J. Brody, R. Advani / Critical Reviews in Oncology/Hematology 58 (2006) 257–265
`
`sub-types and suggest use of this modality in those patients
`at highest risk for rapid disease recurrence and progression.
`The reports of clinical responses correlating with GVHD
`and remote from chemotherapy suggest a GVL effect, which
`should prompt investigation into other immunotherapeutic
`approaches.
`
`3.6. Radioimmunotherapy
`
`Though radiation therapy (RT) has been described to be
`effective in MCL [57], external beam RT has little role in
`MCL because of the rarity of limited stage disease. Because
`immunotherapy with rituximab has shown additive effect
`with chemotherapy, radio-immunotherapy is an attractive
`therapeutic option for patients with MCL. The two clini-
`cally available radio-immunoconjugates are iodine 131I tosi-
`tumomab (Bexxar®) and yttrium 90Y ibritumomab tiuxetan
`(Zevalin®).
`A recent study of Bexxar in 16 patients with relapsed
`MCL utilized an 131I dose calculated to deliver 20–25 Gy to
`vital organs [58]. The treatment was followed 10 days later
`by high-dose etoposide, cyclophosphamide and ASCT. The
`3-year OS and PFS were estimated at 93% and 61%. Subse-
`quently, in a study of 15 patients with a median of three prior
`therapies, patients were treated with Zevalin at 0.3–0.4 mCi
`90Y kg [59]. Treatment was well tolerated and the ORR was
`33% with CR rate of 20%. The median PFS in this group was
`only 4.9 months (and only 5.7 months in responding patients).
`The role of radio-immunotherapy in the upfront manage-
`ment of MCL is also being assessed. A strategy employed
`by the Eastern Cooperative Oncology Group was to use R-
`CHOP for four cycles to induce remission (in an attempt to get
`systemic control and marrow clearing). Responding patients
`who met standard criteria for RIT administration were treated
`with a single dose of Zevalin. This study has just completed
`accrual. Another recent trial treated 24 newly diagnosed MCL
`patients who were ineligible for ASCT with Bexxar followed
`by CHOP 12 weeks later. After Bexxar, the ORR and CR rate
`were 83% and 42%. Nine of the 15 patients who have com-
`pleted their therapy remain in remission after 2–36 months of
`follow-up and 2 patients who progressed were successfully
`able to receive ASCT [60]. Longer follow-up will be required
`to see if these novel strategies improve outcomes.
`
`3.7. Other immunotherapies
`
`More recently, idiotype vaccines that deliver a recom-
`binant protein derived from the surface immunglobulin of
`an individual patient’s MCL have been investigated with
`promising early results [61]. These trials have advanced to
`take advantage of lessons learned from recent advances in
`melanoma immunotherapy [62] such as treating patients with
`minimal disease burden and after lymphodepletion (to reduce
`regulatory T cell interference). A recent example used idio-
`type vaccinations 3 months after ASCT in eight MCL patients
`[63]. Six of these patients were in CR after ASCT and four
`
`remain in CR 14–38 months later. CD4+ T cell and humoral
`responses against the individual’s idiotype were seen in most
`patients.
`
`3.8. Newer agents
`
`Bortezomib is a potent and specific proteasome inhibitor
`that was FDA approved in 2003 for the treatment of relapsed
`multiple myeloma. There have been several smaller studies
`of this agent, but the largest so far (PINNACLE study) has
`yielded initial data on 48 of the 102 enrolled patients. All
`patients had failed one or two prior regimens and response
`rate was 40% [64]. Though it is too early to draw conclusions
`regarding the significance of these data, they are encouraging
`considering that these patients have been treated with aggres-
`sive prior regimens.
`Thalidomide in combination with rituximab was evaluated
`in relapsed/refractory MCL patients with impressive anti-
`tumor activity recently reported. In one study, 16 patients
`with relapsed/refractory MCL were treated with rituximab
`at 375 mg/m2 for four weekly doses concomitantly with
`thalidomide. Eighty percent (13/16) experienced an objective
`response with five achieving a CR (31%) [65]. The median
`PFS was 20.4 months (95% CI, 17.3–23.6), and estimated 3-
`year survival

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket