throbber
Cancer Therapy: Clinical
`
`Lenalidomide and Rituximab in Waldenstrom’s Macroglobulinemia
`
`Steven P. Treon,1,3 Jacob D. Soumerai,1Andrew R. Branagan,1 Zachary R. Hunter,1ChristopherJ. Patterson,1
`Leukothea Ioakimidis,1Luis Chu,4 Paul Musto,5 Ari D. Baron,6 Johannes C. Nunnink,7
`Joseph J. Kash,8 Terenig O. Terjanian,9 Paul M. Hyman,10 Elena L. Nawfel,11
`David J. Sharon,12 Nikhil C. Munshi,2,3 and Kenneth C. Anderson2,3
`
`Abstract Purpose: Thalidomide and its more potent immunomodulatory derivative lenalidomide enhance
`rituximab-mediated antibody-dependent cell-mediated cytotoxicity. We therefore evaluated
`lenalidomide and rituximab in symptomatic Waldenstrom’s macroglobulinemia (WM) patients
`naive to either agent.
`Experimental Design: Intended therapy consisted of 48 weeks of lenalidomide (25 mg/d for
`3 weeks and then 1week off) along with rituximab (375 mg/m2/wk) dosed on weeks 2 to 5 and
`13 to 16. Sixteen patients were enrolled, 12 of whom were previously untreated.
`Results: Unexpectedly, we observed an acute decrease in hematocrit in 13 of 16 patients
`(median hematocrit decrease, 4.8%), which was attributable to lenalidomide patients and which
`led to cessation of further enrollment on this study. Lenalidomide-related anemia was observed
`even at doses as low as 5 mg/d and occurred in the absence of hemolysis or other cytopenias.
`The overall response and major response (<50% decrease in serum IgM) rates were 50% and
`25%, respectively, on an intent-to-treat basis. With a median follow-up of 31.3 months, 4 of
`8 responding patients have progressed with a median time to progression of 18.9 months.
`Conclusion: Lenalidomide produces unexpected but clinically significant acute anemia in
`patients with WM. In comparison with our previous study with thalidomide and rituximab in an
`analogous patient population, the responses achieved in WM patients with lenalidomide and
`rituximab appear less favorable.
`
`Waldenstrom’s macroglobulinemia (WM) is a B-cell disorder
`characterized primarily by bone marrow infiltration with
`lymphoplasmacytic cells along with demonstration of an IgM
`monoclonal gammopathy (1). This condition is considered to
`
`Authors’ Affiliations: 1Bing Center for Waldenstrom’s Macroglobulinemia and
`2Jerome Lipper Multiple Myeloma Center, Dana-Farber Cancer Institute; 3Harvard
`Medical School, Boston, Massachusetts; 4Florida Cancer Specialists, Sarasota,
`Florida; 5Commonwealth Hematology Oncology, Quincy, Massachusetts; 6Pacific
`Hematology Oncology Associates, San Francisco, California; 7University of
`Vermont Cancer Center, Burlington,Vermont; 8Edward Hospital and Health Services
`Cancer Center, Naperville, Illinois; 9Sanford R. Nalitt Institute for Cancer and Related
`Blood Disease, Staten Island, New York; 10Hematology Oncology Associates of
`Western Suffolk PC, Bayshore, New York; 11Harold Alfond Center for Cancer Care,
`Augusta, Maine; and 12Monmouth Medical Center’, Long Branch, NewJersey
`Received 4/4/08; revised 7/10/08; accepted 7/27/08.
`Grant support: Bing Fund for Waldenstrom’s macroglobulinemia, Research Fund
`for Waldenstro« m’s at the Dana-Farber Cancer Institute, and NIH Development
`Award K23CA087977-03 (S.P. Treon).
`The costs of publication of this article were defrayed in part by the payment of page
`charges. This article must therefore be hereby marked advertisement in accordance
`with 18 U.S.C. Section 1734 solely to indicate this fact.
`Note: S.P. Treon, A.R. Branagan, Z.R. Hunter, N.C. Munshi, and K.C. Anderson
`designed and wrote the study. J.D. Soumerai, C.J. Patterson, and L. Ioakimidis
`oversaw the data collection. S.P. Treon, L. Chu, P. Musto, A.D. Baron, J.C. Nunnink,
`J.J. Kash,T.O. Terjanian, P.M. Hyman, E.L. Nawfel, and D.J. Sharon treated the study
`patients and provided study data.
`Requests for reprints: Steven P. Treon, Bing Center for Waldenstrom’s
`Macroglobulinemia, Dana-Farber Cancer Institute, M547, 44 Binney Street, Boston,
`MA 02115. Phone: 617-632-2681; Fax : 617-632-4862; E-mail: steven___treon@
`dfci.harvard.edu.
`F 2009 American Association for Cancer Research.
`doi:10.1158/1078-0432.CCR-08-0862
`
`be lymphoplasmacytic lymphoma as defined by the REAL and
`WHO classification systems (2, 3). Despite advances in therapy,
`WM remains incurable. As such, novel therapeutic agents are
`needed for the treatment of WM.
`One class of therapeutics that has been successfully used in
`patients with WM are monoclonal antibodies. Both rituximab
`and alemtuzumab have been evaluated in WM as single agents
`with major response rates of 30% to 40%, whereas the
`combination of rituximab with chemotherapy has resulted in
`response rates of 70% to 90% (4). With the attainment of
`higher response rates with chemo-antibody therapy, consider-
`ably more short-term and long-term toxicities have been
`reported (4, 5). In an effort to augment monoclonal antibody
`responses in WM patients while averting short-term and long-
`term chemotherapy-induced toxicities, we have sought
`the
`development of immunomodulatory agents for combination
`with rituximab. Thalidomide and its more potent immuno-
`modulatory derivative lenalidomide augment antibody-
`dependent cell-mediated cytotoxicity (5). Moreover,
`these
`agents also lead to expansion of natural killer cells, which
`serve as important effector cells for rituximab activity in
`patients with indolent non-Hodgkin’s lymphoma (6 – 9). As a
`follow-up to these findings, we recently performed a clinical
`trial exploring the combined use of thalidomide and rituximab
`in patients with WM who were naive to either agent (10). The
`results of
`this study demonstrated a higher (70%) major
`(z50% decrease in IgM) response rate than observed previously
`with either thalidomide (20-25%) or rituximab (30-40%)
`alone as upfront therapy in WM patients. Responses in this
`
`www.aacrjournals.org
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`355
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`Clin Cancer Res 2009;15(1) January 1, 2009
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`

`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`IPR2018-00685
`Celgene Ex. 2017, Page 1
`
`

`

`Cancer Therapy: Clinical
`
`Translational Relevance
`
`The intent of this study was to examine the potential for
`the immunomodulator lenalidomide to augment rituximab
`activity in patients with WM. The study showed a unique
`idiopathy for this drug in this patient population, which
`consisted of acute anemia and which persisted despite
`dose reduction and resulted in numerous hospitalizations
`for patients due to anemia complications. This aggravated
`anemia was seen even with dose reduction.We believe that
`this study is very important to alerting clinicians to this nov-
`el and unexpected toxicity, which appears idiopathic to
`WM patients. The mechanism for this finding remains to
`be delineated. The findings of this study are particularly im-
`portant given the relatedness of WM to multiple myeloma
`and the interchange of therapeutics among these disease
`entities by clinicians. Moreover, in comparison with our
`previous study with thalidomide and rituximab in an analo-
`gous patient population, the responses achieved in WM
`patients with lenalidomide and rituximab appear less favor-
`able.We believe that the results of this study will be impor-
`tant not only to clinicians caring for patients with WM but
`also to investigators examining the role of lenalidomide in
`related low-grade B-cell malignancies.
`
`study were durable, with time to progression (TTP) of z38
`months observed among responders. The response rate and TTP
`for combined thalidomide and rituximab therapy was compa-
`rable with that reported with combinations of cytotoxic agents
`or nucleoside analogues and rituximab (4). However, we
`observed a high incidence (44%) of reversible grade z2
`peripheral neuropathy. A higher incidence of peripheral
`neuropathies has also been observed by us in WM patients
`treated with bortezomib in comparison with patients with
`other B-cell malignancies, suggesting a constitutive predisposi-
`tion to neuropathy (11). As such, given the greater immuno-
`modulatory activity as well as diminished neuropathic
`potential for lenalidomide relative to thalidomide, we per-
`formed this phase II study of lenalidomide and rituximab in
`WM patients naive to either agent and reported herein the
`toxicities and activity of this novel combination.
`
`Patients and Methods
`
`Patients with a clinicopathologic diagnosis of WM requiring therapy
`based on the consensus recommendations of the Second International
`Workshop on Waldenstrom’s Macroglobulinemia (12), and who were
`naive to rituximab and lenalidomide, and with CD20+ tumor cells as
`determined by previous bone marrow immunohistochemistry or flow
`cytometry were eligible for this study. To meet eligibility, patients had
`to show a monoclonal IgM protein, minimum IgM level z2 times the
`upper limit of normal, baseline platelet count z25,000/AL, absolute
`neutrophil count z500/AL, serum creatinine <2.5 mg/dL (unless
`nephropathy was attributable to their WM), serum total bilirubin and
`SGOT <2.5 times the upper limit of normal, and Eastern Cooperative
`Oncology Group performance status 0 to 2. No chemotherapy, steroid
`therapy, or radiation therapy within 30 days of study entry was
`permitted. Patients who were pregnant or lactating, had serious
`comorbid disease, had any uncontrolled bacterial, fungal, or viral
`infection, or an active second malignancy were not eligible. All men and
`
`women of reproductive potential were required to agree to use an
`acceptable method of birth control before, during treatment, and for
`6 months after completion of study treatment.
`All patients provided informed written consent and the institutional
`review board approved the protocol. Intended therapy consisted of
`lenalidomide administered at a starting dose of 25 mg/d by mouth for
`3 weeks and then 1 week off on a syncopated schedule as part of a
`4-week cycle. Patients could receive up to 48 weeks of therapy with
`lenalidomide. Dose de-escalation was permitted to 15 mg/d on days of
`treatment. Following enrollment of the first 11 patients, the protocol
`was amended to initiate patients on a start dose of 20 mg/d on the same
`syncopated schedule as before, with dose de-escalation permitted to
`10 mg/d on days of treatment. Following the enrollment of one patient
`at the start dose of 20 mg/d, the protocol was again amended to initiate
`patients at a start dose of 15 mg/d, on the same syncopated schedule as
`before, with dose de-escalation permitted to 5 mg/d on days of
`treatment.
`Rituximab was administered at 375 mg/m2/wk during weeks 2 to 5
`and 13 to 16 for a total of 8 infusions. Patients who did not tolerate the
`first cycle (4 infusions) of rituximab therapy were removed from the
`study and not replaced. Sixteen patients were enrolled in this study,
`which used a Simon two-stage design. Sample size was based on the
`assumption that the expected response rate would be at least 50%.
`Therefore, to have a 95% confidence interval of f20%, a sample size of
`25 was required.
`Response determination. A baseline evaluation was obtained for
`enrollment within 30 days before initiation of
`therapy. Patients
`underwent re-staging studies every 3 months while on therapy and
`thereafter every 3 months until progression of disease. As part of their
`response evaluation, all patients underwent history and physical exam,
`laboratory studies consisting of a complete blood count and
`differential, serum IgM levels, h2-microglobulin levels, and bone
`marrow biopsy and aspiration. Response determinations were made
`using modified consensus panel criteria from the Third International
`Workshop on Waldenstrom’s Macroglobulinemia, and response rates
`determined on an evaluable basis (13). A complete response was
`defined as having resolution of all symptoms, normalization of serum
`IgM levels with complete disappearance of
`IgM paraprotein by
`immunofixation, and resolution of any adenopathy or splenomegaly.
`Patients achieving a major response and a minor response were defined
`as achieving z50% and z25% reduction in serum IgM levels,
`respectively. Patients with stable disease were defined as having <25%
`change in serum IgM levels in the absence of new or increasing
`adenopathy or splenomegaly and/or other progressive signs or
`symptoms of WM. Progressive disease was defined as occurring when
`a >25% increase in serum IgM level occurred from the lowest attained
`response value or progression of clinically significant disease-related
`symptom(s). For patients undergoing plasmapheresis, serum IgM levels
`were used for purposes of assessing response only after steady-state
`attainment (z5 weeks from last plasmapheresis). TTP was calculated
`from the start of therapy using the Kaplan-Meier method.
`Analysis of peripheral blood effector cells. Serial changes in the
`absolute levels of peripheral blood effector cells following lenalidomide
`monotherapy during the first week (and before introduction of
`rituximab to prevent effector cell alterations) were done as described
`previously (10).
`Statistical analysis. Comparison of pretreatment and post-
`treatment variables was done using a two-tailed Student’s t test on
`Microsoft Excel software. P V 0.05 was deemed to be significant for the
`above studies.
`
`Results
`
`Patient and disease characteristics. The clinical features of
`the 16 patients enrolled in this study are summarized in
`Table 1. Of
`the 16 patients enrolled on study, 12 were
`previously untreated. Of the 4 previously treated patients, all
`
`Clin Cancer Res 2009;15(1) January 1, 2009
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`www.aacrjournals.org
`
`356
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`

`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`IPR2018-00685
`Celgene Ex. 2017, Page 2
`
`

`

`Table 1. Baseline characteristics for all 16 patients
`enrolled on study
`
`Gender
`Untreated
`Age (y)
`Prior therapies
`Bone marrow involvement (%)
`Serum IgM (mg/dL)
`h2-Microglobulin (mg/L)
`Hematocrit (%)
`Platelets (k/AL)
`Leukocytes (k/AL)
`
`Median (range)
`
`12 male/4 female
`12/16 (75%)
`65 (49-85)
`0 (0-2)
`37.5 (5-90)
`4,000 (1,180-7,130)
`3.3 (1.8-6)
`32.1 (24-36.6)
`282.5 (78-568)
`6.05 (3-22)
`
`had relapsed to their prior therapy. The median cumulative
`dose of lenalidomide administered for the intended 48-week
`treatment period among all enrolled patients was 455 mg
`(range, 140-5,670), and the median number of weeks on
`lenalidomide therapy was 15.8 (range, 1.1-48). The median
`number of cycles of rituximab among all enrolled patients was
`8 (range, 3-8). Twelve patients were evaluable for response.
`Four patients were unevaluable for response due to their
`premature withdrawal resulting from adverse events associated
`with lenalidomide (n = 3) and rituximab (n = 1).
`Clinical response to therapy. The individual changes in
`serum IgM levels at best response for all evaluable patients
`are shown in Fig. 1. Median serum IgM levels for all evaluable
`patients declined from 2,980 mg/dL (range, 1,180-7,130) to
`1,775 mg/dL (range, 83-4,220) at best response (P = 0.015).
`Pre-therapy, 6 of 12 (50%) showed a serum IgM level z3,000
`mg/dL; at best response, only 3 of 12 (25%) had an IgM level
`z3,000 mg/dL. Overall, 8 of 16 patients enrolled in this study
`showed at least a minor response as their best response. Of
`these patients, 4 of 16 (25%) achieved a major response and
`4 of 12 (25%) achieved a minor response. There were no
`complete responders. Among responding patients, the median
`time to best response was 11.8 months (range, 6-26.4), and the
`median time for a 25% reduction in serum IgM among
`responders was 3.6 months (range, 1.1-7.3). Among major
`responders, the median time to achieving a 50% reduction in
`serum IgM was 4.6 months (range, 2-13). Two patients
`displaying bulky adenopathy (n = 2) and/or splenomegaly
`(n = 1) had complete resolution of their extramedullary disease
`
`Waldenstrom’s Macroglobulinemia
`
`these disease sites following
`after experiencing pain at
`administration of lenalidomide and rituximab.
`Time to progression. The median TTP for all evaluable
`patients was 17.1 months (range, 2-34.3) (Fig. 2). With a
`median follow-up of 31.3 months, 4 of the 8 responding
`patients have progressed. The median TTP for all responding
`patients was 18.9 months (range, 11.4-34.3; Fig. 2).
`Changes in hematologic variables. Pre-therapy, 2 (16.6%)
`and 1 (8.3%) of the 12 evaluable patients showed hematocrit
`V30% and platelet count V100,000/AL, respectively. Following
`therapy, at best response, 1 (8.3%) and none of
`the 12
`evaluable patients showed hematocrit V30% and platelet count
`V100,000/AL, respectively. A significant increase in the median
`hematocrit at best response was noted for the 12 evaluable
`patients from 32.1% (range, 24-36.6%) to 36% (range, 29-
`40.8%) following therapy (P = 0.037). The observed increase in
`median hematocrit with therapy followed cessation and/or dose
`modification of lenalidomide. Pre- and post-therapy, median
`platelet counts remained unaffected by therapy (P = 0.22).
`Toxicities. Premature discontinuation of lenalidomide ther-
`apy occurred in 14 of 16 (88%) patients despite dose reduction
`in 13 of
`these patients and led to cessation of
`further
`enrollment on this study. Discontinuation of lenalidomide
`was based on aggravated anemia or anemia-related complica-
`tions for 13 patients, and/or concurrent myelosuppression
`(neutropenia, n = 1; thrombocytopenia, n = 1), as well as
`development of lenalidomide-related palpitations in 1 patient.
`Acute decreases in hematocrit were observed during first
`2 weeks of lenalidomide therapy in 13 of 16 (81%) patients
`(Fig. 3). Within the first 2 weeks of therapy, the median
`hematocrit declined from 32.3% (range, 26.4-36.6%) to 27.1%
`(range, 24-32.4%). This decline was observed for most patients
`even after 1 week of being on lenalidomide alone and before
`receiving the first rituximab infusion. The median decrease in
`absolute hematocrit values at lowest point in the 2 weeks
`following start of lenalidomide was similar for all three start
`doses examined: -3.15% (25 mg; n = 11), -5.4% (20 mg; n = 1),
`and -3.75% (15 mg; n = 4). The decrease in hematocrit
`observed following initiation of
`lenalidomide therapy was
`deemed to be a factor, at least in part, for the hospitalization of
`4 patients due to congestive heart failure (n = 2), arrhythmia
`(n = 1), and syncope (n = 1). Examination of lactic dehydro-
`genase and reticulocyte counts post-lenalidomide in 4 patients
`who became anemic were unrevealing for hemolysis, and
`a bone marrow biopsy and aspiration done in 2 patients
`
`Fig. 1. Individual changes (%) in serum IgM
`levels at best response following treatment
`with lenalidomide and rituximab.
`
`www.aacrjournals.org
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`357
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`Clin Cancer Res 2009;15(1) January 1, 2009
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`

`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`IPR2018-00685
`Celgene Ex. 2017, Page 3
`
`

`

`Cancer Therapy: Clinical
`
`Fig. 2. TTP for (A) all evaluable patients and (B) those who responded to
`lenalidomide and rituximab. Open circles, patients who had not progressed at last
`follow-up.
`
`showed normal or mildly decreased erythroid elements. No
`overt blood loss was seen. Erythropoietin administration was
`required for 10 patients along with transfusional support
`consisting of packed RBC and platelets for 7 and 1 patients,
`respectively, as a consequence of lenalidomide. Although no
`dose reduction in rituximab was permitted on this study,
`premature discontinuation for this treatment occurred in 3
`patients for rituximab-induced symptomatic hyperviscosity
`(n = 2) or anaphylaxis (n = 1). A complete list of all grade
`z2 toxicities and the treatment (lenalidomide, rituximab, or
`both) to which they are possibly, probably, or definitely
`attributed to appears in Table 2.
`Paradoxical
`increases in serum IgM levels. Abrupt and
`paradoxical increases in serum IgM levels have been reported
`with the use of rituximab in patients with WM and can
`aggravate hyperviscosity and contribute to hyperviscosity-
`related symptoms (14, 15). For this reason, plasmapheresis
`was strongly encouraged for patients who had a pre-therapy
`serum viscosity of z3.5 CP. Six patients underwent pre-therapy
`plasmapheresis. During the first four infusions of rituximab, an
`increase in serum IgM above baseline of z25% was observed in
`12 of 16 (75%) patients and prompted the use of plasmaphe-
`
`resis in 7 of 16 (44%) patients. This paradoxical spike in serum
`IgM levels was not observed for the 10 patients who received
`the second 4-week course of rituximab, and none required
`plasmapheresis just before or during the second course of
`rituximab treatment. No spike in IgM levels was observed
`during the period patients were on lenalidomide monotherapy
`(during the first week) and only occurred following initiation
`of rituximab therapy.
`Effect of lenalidomide on immune effector cell levels. As part
`of this study, we assessed the effect of lenalidomide as an
`immunomodulating agent by determining changes in absolute
`total T cells (CD3+), natural killer cells
`median levels of
`(CD16+CD56+), helper T cells (CD4+), cytotoxic T cells (CD8+),
`as well as monocytes following 7 days of
`lenalidomide
`monotherapy and before the first rituximab infusion. The
`baseline and day 7 levels for these effector cells are provided in
`Table 3. No significant change in the level of total, CD4 and
`CD8 lymphocyte, natural killer cell, and monocyte peripheral
`blood levels was observed. In addition, no association with
`changes in any effector cell population and response was
`observed.
`
`Discussion
`
`Despite robust expression of CD20 in WM, responses to
`rituximab monotherapy are seen in less than half of treated
`patients, paralleling the experiences reported in other indolent
`non-Hodgkin’s lymphoma (4). Tumor-related variables includ-
`ing CD20 antigen loss, complement resistance antigen expres-
`sion, and tumor burden have been addressed previously by us
`and others and do not account for the heterogeneous response
`observed in WM patients treated with rituximab (16 – 18).
`The possibility that patient-related differences might account
`for the heterogeneity in rituximab response in WM patients
`was suggested by our previous studies, showing a high degree
`of dependence for its activity on polymorphisms present
`within the rituximab binding domain of FcgRIIIA (CD16;
`ref. 19). These studies also implicated antibody-dependent
`
`Fig. 3. Pre-therapy and lowest post-therapy hematocrit for 13 WM patients
`who showed aggravated anemia within a 2-week period of initiation of
`lenalidomide therapy.
`
`Clin Cancer Res 2009;15(1) January 1, 2009
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`www.aacrjournals.org
`
`358
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`

`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`IPR2018-00685
`Celgene Ex. 2017, Page 4
`
`

`

`Table 2. Toxicities experienced by the 16 patients on study deemed to be possibly, probably, or definitely
`related to either lenalidomide, rituximab, or both
`
`Toxicity
`
`Grade 2, n (%)
`
`Grade 3, n (%)
`
`Grade 4, n (%)
`
`Attribution
`
`Waldenstrom’s Macroglobulinemia
`
`Allergic reaction
`Anemia
`Arrhythmia
`Back pain
`Chest pain
`Diaphoresis
`Fatigue
`Headache
`Infection
`Leukopenia
`Liver function abnormalities
`Nausea
`Neutropenia
`Peripheral neuropathy
`Pleural effusion
`Pruritus
`Pulmonitis
`Thrombocytopenia
`Tinnitus
`Urinary urgency
`Visual impairment
`
`Abbreviation: NA, not applicable.
`
`1 (6.2)
`8 (50.0)
`2 (12.5)
`1 (6.2)
`NA
`1 (6.2)
`4 (25.0)
`2 (12.5)
`2 (12.5)
`6 (37.5)
`2 (12.5)
`1 (6.2)
`3 (18.7)
`1 (6.2)
`NA
`1 (6.2)
`NA
`1 (6.2)
`2 (12.5)
`1 (6.2)
`1 (6.2)
`
`NA
`1 (6.2)
`1 (6.2)
`NA
`1 (6.2)
`NA
`NA
`NA
`NA
`NA
`NA
`NA
`5 (31.2)
`NA
`1 (6.2)
`NA
`1 (6.2)
`1 (6.2)
`NA
`NA
`NA
`
`NA
`NA
`NA
`NA
`NA
`NA
`NA
`NA
`NA
`NA
`NA
`NA
`NA
`NA
`NA
`NA
`NA
`NA
`NA
`NA
`NA
`
`Rituximab
`Lenalidomide
`Lenalidomide
`Both
`Rituximab
`Lenalidomide
`Lenalidomide
`Rituximab
`Both
`Both
`Both
`Lenalidomide
`Lenalidomide
`Lenalidomide
`Rituximab
`Lenalidomide
`Rituximab
`Lenalidomide
`Lenalidomide
`Lenalidomide
`Rituximab
`
`cell-mediated cytotoxicity function, particularly mediated by
`FcgRIIIA-expressing natural killer cells, as being essential to
`rituximab response in WM patients. We therefore focused our
`efforts on enhancing rituximab efficacy by use of agents, which
`could augment natural killer cell levels as well as antibody-
`dependent cell-mediated cytotoxicity activity. Our previous
`findings showed that thalidomide could enhance circulating
`natural killer cells in MM patients, whereas thalidomide and
`more so its analogue lenalidomide enhanced rituximab-
`mediated antibody-dependent cell-mediated cytotoxicity activ-
`ity (6, 20). As such, we initiated clinical trials first examining
`the activity of thalidomide and thereafter lenalidomide in
`combination with rituximab.
`The results of the present study examining lenalidomide in
`combination with rituximab showed overall and major
`response rates of 50% and 25%, respectively, on an intent-to-
`treat basis. No complete responses were observed. The median
`TTP for responding patients was 18.9 months. Disappointingly,
`
`however, the overall and major response rates as well as the TTP
`observed with lenalidomide and rituximab appeared inferior to
`the results attained in our recent study with thalidomide and
`rituximab in a highly comparable patient population, wherein
`the overall and major response rates were 72% and 64%,
`respectively, and TTP for responding patients was 38.7 months.
`This finding is unexpected, particularly in context of previous
`findings by us and others showing greater immunostimulatory
`properties including enhanced antibody-dependent cell-medi-
`ated cytotoxicity activity for lenalidomide versus thalidomide
`(6, 20). Potential reasons for this discrepancy in activity for
`lenalidomide versus thalidomide could include higher rate
`of premature discontinuation of therapy due to intolerance
`(88% versus 44%) as well as greater nonimmunomodulatory
`mechanisms of action for thalidomide such as phosphodies-
`terase-4 inhibition. Impressive, however, was the observation of
`complete nodal responses in 2 patients with bulky adenopathy,
`which included 1 patient with marked (22 cm) splenomegaly.
`
`Table 3. Changes in peripheral blood effector cell
`patients with WM
`
`levels following 7 days of lenalidomide therapy in 16
`
`Peripheral blood effector cells
`
`Baseline level
`
`Post-7 days lenalidomide
`
`Lymphocytes
`Monocytes
`CD3+
`CD4+
`CD8+
`CD4/CD8 ratio
`CD16+/CD56+
`
`1,185 (740-7,260)
`630 (110-1,320)
`827 (310-2,066)
`536 (140-1603)
`273 (131-856)
`1.68 (0.46-6.55)
`208 (37-392)
`
`770 (400-12,560)
`520 (10-1,900)
`436 (284-6,028)
`273 (55-7,132)
`180 (64-1,632)
`1.87 (0.39-9.71)
`259 (4-1,005)
`
`P
`
`0.87
`0.62
`0.89
`0.76
`0.80
`0.94
`0.51
`
`NOTE: Levels were determined before administration of first rituximab infusion. Median (range) for absolute circulating levels of total
`lymphocytes, monocytes, CD3+, CD4+, CD8+, and CD16+/56+ cells.
`
`www.aacrjournals.org
`
`
`
`
`
`
`
`
`
`
`
`
`
`359
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`Clin Cancer Res 2009;15(1) January 1, 2009
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`

`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`IPR2018-00685
`Celgene Ex. 2017, Page 5
`
`

`

`Cancer Therapy: Clinical
`
`This finding may hold particular relevance for the investigation
`of lenalidomide and rituximab in patients with other types of
`lymphomas.
`An important recognition in this study was the unexpected
`anemia produced by lenalidomide in patients with WM,
`particularly the acuity in which such decreases in hematocrit
`occurred. The mechanism for this finding remains to be
`clarified but suggests that
`lenalidomide may aggravate an
`underlying constitutional genetic defect perhaps central to the
`pathogenesis of WM itself given the uniqueness of
`this
`toxicity in WM patients. The immunomodulatory derivatives
`of
`thalidomide including lenalidomide and pomalidomide
`are known to dampen both erythroid maturation and/or
`erythroid colony formation in part through down-regulation
`of GATA-1, GATA-2, EKLF, and Gfi-1b (21, 22). Studies
`delineating the relative expression levels of these and other
`red cell transcription factors in stem cells of WM patients
`may help elucidate this idiopathic finding of lenalidomide-
`related anemia in this patient population. Unfortunately,
`despite aggressive dose reduction in lenalidomide, many
`patients did not tolerate lenalidomide because of faltering
`hematocrits at the doses and schedule examined in this study.
`Future trials using lower start doses of
`lenalidomide or
`alternative schedules could be considered in WM patients,
`although in view of the inferior outcomes observed in this
`trial with lenalidomide versus thalidomide with rituximab,
`other combinations should be considered, such as with
`bortezomib.
`
`Lastly, this study underscores the importance of conducting
`clinical trials in patients with various malignancies, because
`unexpected toxicities can often occur even among patients
`considered to have similar diseases. Although lenalidomide is
`known to produce late neutropenia and thrombocytopenia in
`patients with multiple myeloma at the same dose levels and
`schedule used in this study, WM patients experienced unex-
`pected aggravation of anemia as their drug-limiting toxicity.
`Similarly, we and others have reported unexpected or exagger-
`ated toxicities in WM patients with agents commonly employed
`in other related B-cell disorders such as the paradoxical IgM
`spike seen with rituximab or the higher incidence of peripheral
`neuropathy with bortezomib.
`In summary, the results of this study show that lenalidomide
`at conventional dosing and schedule can produce unexpected
`and acute declines in hematocrit in patients with WM, which
`are clinically significant and can result in hospitalizations.
`Moreover, in comparison with thalidomide and rituximab, the
`combination of lenalidomide and rituximab produced inferior
`outcomes in terms of both response rates and TTP in an
`analogous patient population.
`
`Disclosure of Potential Conflicts of Interest
`
`S.P. Treon, N.C. Munshi, and K.C. Anderson are members of the speakers’ bureau
`of and have received research support from Celgene, Inc. S.P. Treon is also a
`member of the speakers’ bureau of and has received research support from
`Genentech, Inc.
`
`References
`1. Owen RG, Treon SP, Al-Katib A, et al. Clinicopatho-
`logical definition of Waldenstro« m’s macroglobuline-
`mia: Consensus Panel Recommendations from the
`Second International Workshop on Waldenstro« m’s
`Macroglobulinemia. Semin Oncol 2003;30:110 ^ 5.
`2. Harris NL, Jaffe ES, Stein H. A revised European-
`American classification of lymphoid neoplasms: a pro-
`posal from the International Lymphoma Study Group.
`Blood 1994;84:1361 ^ 92.
`3. Harris NL, Jaffe ES, Diebold J, et al.World Health Or-
`ganization classification of neoplastic diseases of the
`hematopoietic and lymphoid tissues: Report of the
`Clinical Advisory Committee Meeting, Airlie House,
`Virginia, November 1997. J Clin Oncol 1999;17:
`3835 ^ 49.
`4. Treon SP, Gertz MA, Dimopoulos M, et al. Update on
`treatment recommendations from the Third Interna-
`tional Workshop on Waldenstrom’s Macroglobuline-
`mia. Blood 2006;107:3442 ^ 6.
`5. Leleu X, Soumeria JD, Hunter ZR, et al. Increased in-
`cidence of transformation and myelodysplasia/acute
`leukemia in patients with Waldenstrom’s macroglobu-
`linemia treated with nucleoside analogues. J Clin
`Oncol 2008. In press.
`6. Davies FE, Raje N, Hideshima T, et al. Lenalidomide
`and immunomodulatory derivatives augment natural
`killer cell cytotoxicity in multiple myeloma. Blood
`2001;98:210 ^ 6.
`7. Janakiraman N, McLaughlin P,White CA, et al. Ritux-
`imab: correlation between effector cells and clinical
`activity in NHL. Blood 1998;92:337a.
`
`8. Gluck WL, Hurst D, Yuen A, et al. Phase I studies
`of interleukin (IL)-2 and rituximab in B-cell non-
`Hodgkin’s lymphoma: IL-2 mediated natural killer cell
`expansion: correlations with clinical response. Clin
`Cancer Res 2004;10:2253 ^ 64.
`9. Khan KD, Emmanouilides C, Benson DM, et al. A
`phase 2 study of rituximab in combination interleu-
`kin-2 for rituximab-refractory indolent non-Hodgkin’s
`lymphoma. Clin Cancer Res 2006;12:7046 ^ 53.
`10. Treon

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