throbber
Journal of Clinical Pharmacy and Therapeutics (2008) 33, 219–226
`
`REVIEW ARTICLE
`
`Lenalidomide in the treatment of multiple myeloma:
`a review
`X. Armoiry*  PharmD PhD, G. Aulagner*  PharmD PhD and T. Faconà MD PhD
`*Pharmacy Department, Groupement Hospitalier Est, Hospices Civils de Lyon, Lyon,  Faculty of Pharmacy,
`Henri Poincare´ University, Nancy and àHematology Department, Hoˆpital Claude Huriez, Centre
`Hospitalier Universitaire Re´gional de Lille, Lille, France
`
`S U M M A R Y
`
`I N T R O D U C T I O N
`
`Lenalidomide is an immunomodulatory drug
`derived from thalidomide. It was developed to
`maximize the anti-inflammatory and anti-neo-
`plasic properties of thalidomide and to reduce its
`toxicity. The molecular mechanism of action of
`lenalidomide is unclear, but
`its therapeutic
`activity is mainly due to its well defined anti-
`inflammatory, immunomodulatory, anti-prolifer-
`ative and anti-angiogenic properties. In relapsed
`or refractory multiple myeloma (MM), lenalido-
`mide, combined with standard dose dexametha-
`sone,
`is superior to dexamethasone alone in
`terms of time to progression, response rate and
`overall survival. The most commonly reported
`adverse events include haematological toxicity
`with manageable neutropenia and thrombope-
`nia. Lenalidomide does not trigger the limiting
`toxicities of thalidomide: somnolence, neuropa-
`thy and constipation. Lenalidomide, in combi-
`nation with dexamethasone, is indicated for the
`treatment of MM patients who have received at
`least one prior therapy and is administered orally
`at the dose of 25 mg q.d. for 21 days of 28-day
`cycles. The drug is being investigated for the
`treatment of newly diagnosed MM.
`In this
`review, we summarize the pharmacokinetic,
`pharmacodynamic and clinical
`trial data on
`lenalidomide.
`
`Keywords: immunomodulatory drugs, lenalido-
`mide, myeloma
`
`Received 23 October 2007, Accepted 27 February 2008
`Correspondence: X. Armoiry, Pharmacy Department, Groupe-
`ment Hospitalier Est, Avenue de Doyen Le´pine, 69 500 Bron,
`France. Tel.: +33 472 357 245; fax: +33 472 357 331; e-mail:
`xavier.armoiry@chu-lyon.fr
`
`Multiple myeloma (MM), also called Kahler’s
`disease, is a rare progressive neoplastic disorder of
`unknown aetiology (1, 2) characterized by accu-
`mulation of a plasma cell clone in the bone mar-
`row. The diagnosis is mainly based on plasma cells
`infiltration in the bone marrow (more than 10% of
`nuclear cells), osteolytic lesions and the presence of
`monoclonal (M) immunoglobulin (or fragment) in
`serum or urine (3). MM represents about 10% of
`haematological malignancies and 1% of all cancers.
`The diagnosis of MM is made in about 21 000
`patients ⁄ year in Europe with approximately 16 000
`annual deaths from the disease (4). MM is pri-
`marily a disease of the elderly, with a median age
`at diagnosis of 68 years. Prior to the introduction of
`alkylating agents, the median survival for patients
`with MM was 12–17 months from time of diagnosis
`(5). The combination of melphalan plus prednisone
`(MP) has remained the gold standard therapy for
`decades, although complete remission (CR) was
`rare (<5%) and median survival did not exceed
`3 years (6). In the 1990s, high-dose therapy (HDT)
`with autologous stem cell
`transplantation was
`shown to prolong survival (about 12 months with
`an acceptable mortality rate of 1–2%), when com-
`pared with conventional
`therapy (7). Despite
`intensive therapy, MM remains an incurable
`disease.
`In the last 10 years,
`thalidomide and
`bortezomib have been shown to be effective in the
`treatment of relapsed or refractory MM (8–10) and
`promising in newly diagnosed MM (11–13). More
`recently,
`lenalidomide has been developed and
`evaluated, not only for relapsed or refractory MM
`but also for newly diagnosed MM (14–17). Lena-
`lidomide (CC-5013 trade name REVLIMIDÒ) is the
`leading drug among the immunomodulatory
`
`Ó 2008 Blackwell Publishing Ltd
`
`219
`
`ALVOGEN, Exh. 1041, p. 0001
`
`

`

`lenalidomide
`As an immunomodulatory drug,
`inhibits the secretion of pro-inflammatory cyto-
`kines including TNF-a, interleukin (IL)-1b, IL-6 and
`IL-12 from lipopolysaccharide (LPS)-stimulated
`peripheral blood mononuclear
`cells
`(PBMC)
`(18, 19). It also increases production of the anti-
`inflammatory cytokine IL-10 by LPS-stimulated
`PBMC and consequently inhibits the expression,
`but not the enzymatic activity, of cyclooxygenase-2
`(20). Lenalidomide induces T-cell proliferation and
`IL-2 and interferon (IFN)-c production (19, 21) and
`it augments cytotoxic activity of natural killer cells
`(22).
`Lenalidomide has anti-angiogenic properties as
`it suppresses vascular endothelial growth factor
`and basic fibroblast growth factor production by
`the endothelium and bone marrow stroma, with
`consequent inhibition of angiogenesis (23).
`The activity profile of lenalidomide is similar to
`that of thalidomide but, in vitro, lenalidomide is 50
`to 2000 times more potent in inhibiting cytokines
`production by LPS-stimulated PBMC, such as
`TNF-a, IL-1b or IL-6 (24).
`
`P H A R M A C O K I N E T I C P R O F I L E
`
`Single-dose lenalidomide pharmacokinetics was
`assessed in a phase I, single-blind, placebo-con-
`trolled, ascending study in 19 healthy volunteers
`(25). Lenalidomide was rapidly absorbed following
`oral administration with maximum plasma con-
`centrations (Tmax) occurring between 0Æ6 and 1Æ5 h
`post-dose. Maximal plasma concentration (Cmax)
`and area under the curve (AUC) increased in a
`dose-proportional manner over the single dose
`range of 5–400 mg. Co-administration with food
`did not alter the extent of absorption (AUC) but did
`reduce Cmax by 36%. The pharmacokinetic dispo-
`sition of lenalidomide is linear. In vitro (14C)-le-
`nalidomide
`binding
`to plasma proteins
`is
`approximately 30%. The metabolic profile of lena-
`lidomide in humans has not been studied.
`In
`healthy volunteers, approximately two-thirds of
`lenalidomide is eliminated unchanged through
`urinary excretion. The process exceeds the glo-
`merular filtration rate and is therefore partially or
`entirely active. Half-life of elimination is approxi-
`mately 3 h. In patients with MM, maximum plasma
`concentrations occurred between 0Æ5 and 4Æ0 h
`post-dose both on days 1 and 28. AUC and Cmax
`
`O
`
`NH
`
`O
`
`O
`
`N
`
`O
`
`220 X. Armoiry et al.
`
`Fig. 1. Chemical structure of thalidomide.
`
`NH
`
`O O
`
`N
`
`O
`
`NH2
`
`Fig. 2. Chemical structure of lenalidomide.
`
`compounds derived from thalidomide aimed at
`maximizing its anti-inflammatory and anti-neo-
`plasic properties and improve its tolerability. This
`new drug is a synthetic glutamic acid derivative
`obtained from thalidomide by the removal of an
`oxy group from the phthalyl ring and by the
`addition of an amino group (Figs 1 and 2). This
`review focuses on the efficacy and tolerability of
`lenalidomide in the treatment of MM.
`
`P H A R M A C O D Y N A M I C P R O P E R T I E S
`
`Lenalidomide is a small molecule analogue of
`thalidomide that was originally identified based on
`its ability to potently inhibit tumour necrosis factor
`(TNF)-a production. It is now known that
`the
`therapeutic activity of lenalidomide is likely due to
`multiple mechanisms of action: anti-inflammatory,
`immunomodulatory, anti-proliferative and anti-angio-
`genic (Fig. 3).
`Lenalidomide exerts direct anti-proliferative and
`pro-apoptotic effects on MM cells grown in vitro.
`
`Inhibits stromal
`cell adhesion
`
`Stimulates T cells
`
`Inhibits
`cytokines
`
`IL-6
`TNF
`IL-1
`
`MM cells
`
`Stimulates
`NK cells
`
`Induces
`aptoptosis
`
`Inhibits
`angiogenisis
`
`Fig. 3. Mechanisms of action of lenalidomide in multiple
`myeloma (from Celgene Corporation, France).
`
`Ó 2008 Blackwell Publishing Ltd, Journal of Clinical Pharmacy and Therapeutics, 33, 219–226
`
`ALVOGEN, Exh. 1041, p. 0002
`
`

`

`values increase proportionally with dose following
`single and multiple doses. Exposure (AUC) in MM
`patients is 57% higher than in healthy male vol-
`unteers.
`In a phase I trial in patients with MM (26), the
`mean terminal elimination half-lives were 3Æ1–4Æ2 h
`on both day 1 and day 28. There was little or no
`accumulation of CC-5013. Intersubject variability
`was generally low to moderate for AUC and Cmax,
`with values ranging from 10Æ6% to 51Æ8% and 3%
`to 33% on day 1 and day 28, respectively.
`Multiple myeloma patients with mild renal
`impairment had an AUC 56% greater than those
`with normal renal function. Pharmacokinetic dif-
`ferences due to race, gender, age or hepatic
`impairment have not been studied.
`
`T H E R A P E U T I C T R I A L S
`
`Main studies for the approval in relapsed or
`refractory MM
`
`Phase I and phase II trials. The clinical programme of
`lenalidomide in MM started in April 2000 with the
`initiation of two phase I studies in heavily pre-
`treated patients with relapsed or refractory MM.
`Both trials were designed to identify the maximum
`tolerated dose (MTD) and to evaluate the safety of
`lenalidomide monotherapy in this population.
`Cytopenias were the dose-limiting toxicity in both
`studies and the MTD of lenalidomide was deter-
`mined to be 25 mg ⁄ day for 21 days ⁄ month.
`No significant somnolence, constipation or neu-
`ropathy was observed with continued lenalido-
`mide therapy. However,
`reversible cytopenias
`were observed during the second cycle of treat-
`ment in subjects who received 25 mg ⁄ day. In the
`first study of the 24 evaluable patients treated
`according to this schedule, 29% had an M para-
`protein reduction superior to 50%;
`in 71% of
`patients, the M component decreased to at least
`25% (26). In the second study, 20% of the subjects
`achieved a reduction of M paraprotein superior to
`50% at doses of 25–50 mg ⁄ day (27).
`The results of two phase II trials are also avail-
`able (28, 29).
`The first one was a multicentre, single-arm,
`open-label study conducted to evaluate the efficacy
`and safety of single-agent lenalidomide, adminis-
`tered at a dose of 30 mg q.d. for 21 days every
`
`Lenalidomide in the treatment of multiple myeloma
`
`221
`
`28 days (28-day cycle) in patients (n = 222) with
`relapsed or refractory MM (28). Twenty-five per
`cent of patients achieved partial reference or
`complete response (PR + CR) and 71% achieved
`stable disease or better and the median time
`to progression (TTP) was approximately 6 months.
`The second study was an open-label, random-
`ized trial to evaluate two dose-regimens of lena-
`lidomide for patients (n = 102) with relapsed or
`refractory MM (29). Patients were randomized to
`receive either 30 mg q.d. or 15 mg b.i.d.,
`for
`21 days of every 28-day cycle. With progressive or
`stable disease after two cycles, patients received,
`additionally, 40 mg dexamethasone for 4 days
`every 14 days. Patients receiving 15 mg twice daily
`showed an increased rate of grade 3 ⁄ 4 myelo-
`suppression (41% vs. 13%, P = 0Æ03). The overall
`response (OR) rate was 25% and the median
`overall survival (OS) in the 30 mg once-daily and
`twice-daily groups was 28 and 27 months, respec-
`tively. Dexamethasone was added in 68 patients
`and 29% responded.
`The preliminary results of this study were the
`basis for the once-daily regimen of lenalidomide,
`combined with dexamethasone, used in the sub-
`sequent phase III trials.
`
`Phase III trials. The approval of lenalidomide for
`the treatment of relapsed or refractory MM was
`mainly based on the results of pivotal studies
`(MM-009 and MM-010) initiated in 2003. The FDA
`agreed that
`the best comparator was standard
`dose dexamethasone. The studies were multicen-
`tre, multinational,
`randomized, double-blind,
`placebo-controlled trials to evaluate and compare
`the
`efficacy of
`lenalidomide plus high-dose
`dexamethasone
`(Rev-Dex) vs. dexamethasone
`alone (Dex).
`MM-009 trial was undertaken in North America
`and MM-010 in Europe,
`Israel and Australia.
`A total of 354 and 351 patients were recruited
`respectively.
`Lenalidomide was given at the dose of 25 mg
`q.d. for 21 days of every 28-day cycle and dexa-
`methasone at the dose of 40 mg from days 1 to 4, 9
`to 12 and 17 to 20 every 28-day cycle for four cycles
`then from days 1 to 4 for subsequent cycles.
`Investigational treatment had to be stopped at the
`time of disease progression or in case of unac-
`ceptable adverse events.
`
`Ó 2008 Blackwell Publishing Ltd, Journal of Clinical Pharmacy and Therapeutics, 33, 219–226
`
`ALVOGEN, Exh. 1041, p. 0003
`
`

`

`222 X. Armoiry et al.
`
`The primary endpoint was TTP according to
`Blade et al.’s criteria (30) and secondary endpoint
`included OS, OR rate and safety. A preplanned
`interim analysis of TTP, response rate and safety
`was performed by an Independent Data Monitor-
`ing Committee (IDMC).
`The results presented below relate to analyses of
`data encompassing the whole double-blind period
`(2003 to summer 2005).
`In the MM-009 North American Phase III trial
`(31, 32), the median TTP with Rev-Dex was supe-
`rior to the one in the Dex arm (48Æ1 weeks vs.
`20Æ1 weeks, P < 0Æ01). The hazard ratio for TTP was
`2Æ82 (95% CI, 2Æ14–3Æ7, P < 0Æ001). The OR rate was
`61% in the Rev-Dex arm and 19Æ9% in Dex arm
`including 14Æ1% and 0Æ6% of CR, respectively. OS
`time was longer for patients treated in the Rev-Dex
`arm (29Æ6 vs. 20Æ2 months) with a hazard ratio of
`1Æ52 (95% CI, 1Æ04–2Æ24, P = 0Æ03).
`The MM-010 trial reported very similar results
`with TTP of 48Æ7 weeks in the Rev-Dex arm vs.
`20Æ1 weeks in the Dex arm (33, 34). The hazard ratio
`for TTP was 2Æ85 (95% CI, 2Æ16–3Æ76, P < 0Æ001). OR
`rate was greater with Rev-Dex regimen (60Æ2% vs.
`24% including 15Æ9% and 3Æ4% of CR respectively).
`The OS was also superior for patients in the Rev-
`Dex arm (not estimable as of May 2006 in this arm
`vs. 20Æ6 months in the Dex arm, P < 0Æ01) with a
`hazard ratio of 2Æ10.
`The interim analysis of TTP and response rate
`was performed at the start of 2005. Since the results
`already surpassed the preplanned significance
`threshold, the IDMC decided in early 2005 to lift
`the blind and recommended that all patients in the
`Dex arm should be offered the choice of treatment
`with lenalidomide–dexamethasone combination.
`The impact of prior therapy with thalidomide on
`sensitivity of MM to subsequent lenalidomide was
`evaluated in a prospective subgroup analysis of
`MM-009 and MM-010 (35). In patients who had
`received prior thalidomide, the major response rate
`(CR + PR) was significantly higher in the group
`assigned to receive Rev-Dex than in those who
`received Dex alone (53Æ2% vs. 15Æ2%, P < 0Æ001). In
`this subgroup, the CR rate was also significantly
`higher for the Rev-Dex vs. Dex alone group (8Æ1%
`vs. 1Æ4%, P < 0Æ05).
`treatments was also
`Influence of previous
`assessed (36). Median TTP for patients with one
`prior regimen was 16Æ5 months in Rev-Dex patients
`
`vs. 4Æ6 months in Dex alone patients (HR, 2Æ86). For
`patients with two or more prior regimens, median
`TTP was also significantly higher in Rev-Dex group
`(10Æ2 months vs. 4Æ7 months; HR, 2Æ66).
`There was also a significant difference between
`the patients with one prior regimen vs. two or more
`regimens within the Rev-Dex group (P < 0Æ05).
`
`Ongoing clinical development in MM
`
`Newly diagnosed MM. Patients eligible for HDT:
`A first non-comparative phase II trial assessed the
`efficacy of lenalidomide plus dexamethasone for
`newly diagnosed MM (14). Results showed a 91%
`OR rate including 38% of CR ⁄ near complete
`response (nCR). The 2-year progression-free sur-
`vival rates for patients proceeding to HDT and
`patients remaining on Rev-Dex were 83% and
`59%, respectively; the OS rates were 92% and 90%
`at 2 years and 92% and 85% at 3 years, respec-
`tively. The 3-year OS rate for the whole cohort was
`88% (15). On this encouraging basis, phase III trials
`are being conducted in order to evaluate lenalido-
`mide plus dexamethasone as frontline therapy.
`The Eastern Cooperative Oncology Group
`conducted a
`randomized study
`comparing
`lenalidomide plus standard-dose dexamethasone
`(L-highD: 12 days of dexamethasone per month)
`with lenalidomide plus low-dose dexamethasone
`(L-lowD: 4 days of dexamethasone per month) (16).
`The interim analysis reported a 1 year survival rate
`of 96Æ5% in the R-lowD arm vs. 86% in the R-highD
`arm (17). Major grade 3 or higher toxicities were
`significantly higher in the R-highD arm: thrombo-
`embolism (22Æ1% vs. 6Æ1%), infection ⁄ pneumonia
`(15Æ7% vs. 7Æ5%) and hyperglycaemia (9Æ7% vs.
`6Æ6%).
`Patients not eligible for HDT: With the same
`therapeutic approach taken with bortezomib and
`thalidomide (11–13), lenalidomide is being evalu-
`ated in combination with the MP standard (R-MP)
`for the treatment of newly diagnosed MM in
`patients not eligible for HDT (elderly patients).
`Recently, a phase I ⁄ II trial conducted in 54 patients
`estimated that the MTD was 0Æ18 mg ⁄ kg melphalan
`and 10 mg lenalidomide (37). With these doses,
`81% of patients achieved at least a partial response,
`47Æ6% achieved a very good partial response and
`23Æ8% achieved a CR. A phase III trial comparing
`R-MP to MP (MM-015) is ongoing (38).
`
`Ó 2008 Blackwell Publishing Ltd, Journal of Clinical Pharmacy and Therapeutics, 33, 219–226
`
`ALVOGEN, Exh. 1041, p. 0004
`
`

`

`Maintenance therapy. The use of maintenance ther-
`apy is mainly discussed in younger patients in
`order to improve the quality of response or to
`maintain it. Lenalidomide is being evaluated in two
`phase III placebo-controlled trials conducted by
`two cooperative groups (the Intergroupe Franco-
`phone du Mye´lome and the Cancer and Leukemia
`Group B), to assess its efficacy at prolonging the
`duration of the response after autologous stem cell
`transplantation in patients under the age of 65 (39,
`40). In elderly patients, the MM-015 study com-
`paring R-MP to MP will also investigate the role of
`lenalidomide maintenance (38).
`
`Other lenalidomide-based combinations for relapsed or
`refractory MM. Other lenalidomide-based regimens
`are under investigation for the treatment of relapsed
`or refractory MM such as lenalidomide plus doxo-
`rubicin and dexamethasone (RAD) or lenalidomide
`plus pegylated liposomal doxorubicin–vincristin–
`dexamethasone (R-Dvd) (41). In a phase I ⁄ II trial
`evaluating RAD regimen, 26 patients out of 31
`(84%) achieved a partial response including one
`confirmed CR with an acceptable toxicity profile
`(42). Another phase I ⁄ II
`trial reported an OR
`response rate of 75%, with 29% of CR ⁄ nCR in
`patients treated with R-Dvd regimen (43).
`
`S A F E T Y A N D T O L E R A B I L I T Y
`
`Neutropenia were reported more frequently in the
`Rev-Dex arm than in the Dex arm (39Æ4% vs. 6Æ3%)
`including grade 3 (30% vs. 2Æ9%) and grade 4
`(5Æ4% vs. 0Æ6%) severities (44). Grade 4 febrile
`neutropenia episodes were observed infrequently
`(0Æ6% in the Rev-Dex group compared with 0Æ0%
`in the placebo ⁄ dexamethasone group). In case of
`neutropenia, the physician should consider the use
`of growth factors in patient management. Throm-
`bocytopenia and anaemia were also more frequent
`in the Rev-Dex arm as compared with the pla-
`cebo ⁄ Dex arm (18Æ4% vs. 5Æ7% and 17Æ0% vs. 6Æ3%,
`respectively).
`The non-haematological adverse reactions which
`occurred significantly more frequently in the Rev-
`Dex arm group compared with the Dex arm were
`fatigue (27Æ2% vs. 18%), asthenia (17Æ6% vs.
`10Æ9%), constipation (23Æ5% vs. 8Æ9%), muscle
`cramp (20Æ1% vs. 10Æ6%), diarrhoea (14Æ2% vs.
`7Æ1%) and rash (10Æ2% vs. 3Æ4%).
`
`Lenalidomide in the treatment of multiple myeloma
`
`223
`
`Lenalidomide has almost no sedative effects and
`neuropathic effect is minimal.
`Other adverse events, such as nausea, vertigo,
`dyspnoea, occurred less frequently.
`As with thalidomide, thrombotic or thrombo-
`embolic events, including deep venous thrombosis,
`pulmonary
`embolism, were
`reported more
`frequently in the Rev-Dex arm (11Æ3%) than in the
`Dex arm (3Æ8%) (45). Furthermore, a combined
`review of studies MM-009 and MM-010 showed
`that the incidence of deep venous thrombosis was
`significantly higher among the Rev-Dex patients
`who received erythropoietic agents concomitantly
`(19Æ2% vs. 6Æ8%, P = 0Æ001). Therefore, antithrom-
`botic prophylaxis using low-molecular-weight
`heparin, aspirin or warfarin, should be given to all
`patients receiving lenalidomide plus dexametha-
`sone and low-molecular-weight heparin should be
`recommended for patients with additional throm-
`botic risk-factors.
`In MM-009 and MM-010 phase trials, atrial
`fibrillation occurred more frequently in the Rev-
`Dex arm (4Æ0% vs. 0%).
`
`T E R A T O G E N I C P O T E N T I A L
`
`There is no reported data on foetal exposure to
`lenalidomide in humans.
`in rats
`studies
`Embryo-foetal development
`revealed no teratogenic effects at the highest dose
`of 500 mg ⁄ kg (approximately 600 times the human
`dose of 10 mg based on body surface area). A pre
`and post-natal development study in rats revealed
`few adverse effects on the offspring of female rats
`treated with lenalidomide
`at doses up to
`500 mg ⁄ kg. The male offspring exhibited slightly
`delayed sexual maturation and the female off-
`spring had slightly lower body weight gain during
`gestation when bred to male offspring (25).
`As lenalidomide is chemically related to thalid-
`omide, and thalidomide is one of the most terato-
`genic drugs, there is a strict contra-indication to the
`use of lenalidomide during pregnancy. Therefore,
`lenalidomide is available in Europe under the
`conditions described in a specific risk management
`programme for female patients with childbearing
`potential. Prior
`to the start of
`treatment, a
`medically supervised pregnancy test should be
`performed during the consultation, when lenalid-
`omide is prescribed or in the 3 days prior to the
`
`Ó 2008 Blackwell Publishing Ltd, Journal of Clinical Pharmacy and Therapeutics, 33, 219–226
`
`ALVOGEN, Exh. 1041, p. 0005
`
`

`

`224 X. Armoiry et al.
`
`Table 1. Dose adjustments recommended at the start of the therapy for patients with impaired renal function
`
`Renal function (CLcr)
`
`Mild renal impairment (CLcr ‡ 50 mL ⁄ min)
`Moderate renal impairment (30 £ CLcr <50 mL ⁄ min)
`Severe renal impairment (CLcr < 30 mL ⁄ min, not requiring dialysis)
`End-stage renal disease (CLcr < 30 mL ⁄ min, requiring dialysis)
`
`Dose adjustment
`
`25 mg once daily (full dose)
`10 mg once dailya
`15 mg every other day
`15 mg, thrice a week following
`each dialysis
`
`aThe dose may be escalated to 15 mg once daily after two cycles if patient is not responding to treatment and is tolerating the treatment.
`
`visit to the prescriber once the patient had been
`using effective contraception for at least 4 weeks.
`Then, the pregnancy test should be repeated every
`4 weeks, including 4 weeks after the end of treat-
`ment, except in the case of confirmed tubal sterili-
`zation. These pregnancy tests should be performed
`on the day of the prescribing visit or in the 3 days
`prior to the visit to the prescriber.
`
`D O S A G E A N D A D M I N I S T R A T I O N
`Lenalidomide (REVLIMIDÒ) is available in 5, 10, 15
`and 25 mg capsules. Based on European Union
`approval,
`the recommended starting dose of
`REVLIMIDÒ is 25 mg ⁄ day with water orally
`administered as a single 25 mg capsule on days
`1–21 of repeated 28-day cycles. The recommended
`dose of dexamethasone is 40 mg ⁄ day on days 1–4,
`9–12 and 17–20 of each 28-day cycle for the first
`four cycles of therapy and then 40 mg ⁄ day orally
`on days 1–4 every 28 days. Dosing is modified, if
`necessary, based upon clinical and laboratory
`findings, in particular in the presence of grade 3 or
`4 neutropenia, thrombocytopenia or other grade 3
`or 4 toxicity judged to be related to lenalidomide.
`Because of its urinary excretion, REVLIMIDÒ must
`be used with caution and dose adapted as shown in
`Table 1 for patients with renal impairment.
`
`C O N C L U S I O N
`
`On the basis of MM-009 and MM-010 phase III
`trials, REVLIMIDÒ is approved in the European
`Union since June 2007, for the treatment of MM
`patients who have received at
`least one prior
`therapy, in combination with dexamethasone.
`Because of its possible teratogenicity, lenalido-
`mide must be used according to a restricted
`
`the
`distribution programme, which requires
`participation of physician, pharmacist and patients.
`This new drug will most likely play an important
`role as frontline therapy for patients with MM. The
`drug is being evaluated for the treatment of other
`haematological malignancies and solid tumours.
`
`R E F E R E N C E S
`
`1. Kyle RA, Rajkumar SV (2004) Multiple myeloma.
`New England Journal of Medicine, 351:1860–1873.
`Review. Erratum in: 2005; 352, 1163.
`2. Rajkumar SV, Kyle RA (2005) Multiple myeloma:
`diagnosis and treatment. Mayo Clinic Proceedings, 80,
`1371–1382.
`3. Terriou L, Leleu X, Yakoub-Agha I (2006) Treatment
`of multiple myeloma. Bulletin du Cancer, 93, 101–106.
`4. Eucan Database 1998. Available at: http://www-dep.
`iarc.fr/eucan/eucan.htm (accessed 1 August 2007).
`5. Durie BG, Salmon SE (1982) The current status and
`future perspective of treatment for multiple myelo-
`ma. Clinics in Haematology, 11, 181–210.
`6. Barlogie B, Shaughnessy J, Tricot G et al. (2004)
`Treatment of multiple myeloma. Blood, 103, 20–32.
`7. Attal M, Harousseau JL, Stoppa AM et al. (1996) A
`prospective, randomized trial of autologous bone
`marrow transplantation and chemotherapy in mul-
`tiple myeloma. Intergroupe Franc¸ais du Myelome.
`New England Journal of Medicine, 335, 91–97.
`8. Myeloma Trialists’ Collaborative Group (2001)
`Interferon as therapy for multiple myeloma: an
`individual patient data overview of 24 randomized
`trials and 4012 patients. British Journal Haematology,
`113, 1020–1034.
`9. Richardson PG, Sonneveld P, Schuster MW et al.
`Assessment of Proteasome Inhibition for Extending
`Remissions (APEX) Investigators (2005) Bortezomib
`or high-dose dexamethasone for relapsed multiple
`myeloma. New England Journal of Medicine, 352, 2487–
`2498.
`
`Ó 2008 Blackwell Publishing Ltd, Journal of Clinical Pharmacy and Therapeutics, 33, 219–226
`
`ALVOGEN, Exh. 1041, p. 0006
`
`

`

`10. Richardson PG, Sonneveld P, Schuster M et al. (2007)
`Extended follow-up of a phase 3 trial in relapsed
`multiple myeloma: final time-to-event results of the
`APEX trial. Blood, 110, 3557–3560.
`11. Palumbo A, Bringhen S, Caravita T et al. (2006)
`Oral melphalan and prednisone chemotherapy plus
`thalidomide compared with melphalan and pred-
`nisone alone in elderly patients with multiple
`myeloma: randomised controlled trial. Lancet, 367,
`825–831.
`12. Facon T, Mary JY, Hulin C et al. (2007) Melphalan
`and prednisone plus thalidomide versus melphalan
`and prednisone alone or reduced-intensity autolo-
`gous stem cell transplantation in elderly patients
`with multiple myeloma (IFM 99-06): a randomised
`trial. Lancet, 370, 1209–1218.
`13. Mateos MV, Hernandez JM, Hernandez MT et al.
`(2006) Bortezomib plus melphalan and prednisone
`in elderly untreated patients with multiple myelo-
`ma: results of a multicenter phase 1 ⁄ 2 study. Blood,
`108, 2165–2172.
`14. Rajkumar SV, Hayman SR, Lacy MQ et al. (2005)
`Combination therapy with lenalidomide plus dexa-
`methasone (Rev ⁄ Dex) for newly diagnosed myelo-
`ma. Blood, 106, 4050–4053.
`15. Lacy MQ, Gertz MA, Dispenzieri A et al. (2007)
`Long-term results of response to therapy, time to
`progression, and survival with lenalidomide plus
`dexamethasone in newly diagnosed myeloma. Mayo
`Clinic Proceedings, 82, 1179–1184.
`16. Rajkumar SV, Jacobus S, Callander N, Fonseca R,
`Vesole D, Greipp P. (2006) A randomized phase III trial
`of
`lenalidomide plus high-dose dexamethasone versus
`lenalidomide plus low-dose dexamethasone in newly
`diagnosed multiple myeloma (E4A03): a trial coordinated
`by the Eastern Cooperative Oncology Group. ASH,
`Orlando (Abstract 799).
`17. Rajkumar SV, Jacobus S, Callander N et al. (2007)
`Phase III
`trial of
`lenalidomide plus high-dose
`dexamethasone versus lenalidomide plus low-dose
`dexamethasone in newly diagnosed multiple mye-
`loma (E4A03): a trial coordinated by the Eastern
`Cooperative Oncology Group, 2007 ASCO Annual
`Journal of Clinical
`Meeting Proceedings Part
`I.
`Oncology, 25(18S Suppl.), LBA8025.
`18. Muller GW, Chen R, Huang SY et al. (1999) Amino-
`substituted thaldidomide analogs: potent inhibitors
`of TNF-a production. Bioorganic & Medicinal Chem-
`istry Letters, 9, 1625–1630.
`19. Corral LG, Haslett PA, Muller GW et al. (1999) Dif-
`ferential cytokine modulation and T cell activation
`by two distinct classes of thalidomide analogues that
`are potent inhibitors of TNF- a. Journal of Immuno-
`logy, 163, 380–386.
`
`Lenalidomide in the treatment of multiple myeloma
`
`225
`
`20. Fujita J, Mestre JR, Zeldis JB, Subbaramaiah K,
`Dannenberg AJ (2001) Thalidomide and its ana-
`logues inhibit lipopolysaccharide-mediated induc-
`tion of cyclooxygenase-2. Clinical Cancer Research, 7,
`3349–3355.
`21. Schafer PH, Gandhi AK, Loveland MA et al. (2003)
`Enhancement of cytokine production and AP-1 tran-
`scriptional activity in T cells by thalidomide-related
`immunomodulatory drugs. Journal of Pharmacology
`and Experimental Therapeutics, 305, 1222–1232.
`22. Davies FE, Raje N, Hideshima T et al. (2001) Thalido-
`mide and immunomodulatory derivatives augment
`natural killer cell cytotoxicity in multiple myeloma.
`Blood, 98, 210–216.
`23. Dredge K, Marriott JB, Macdonald CD et al. (2002)
`Novel
`thalidomide analogues display anti-angio-
`genic activity independently of immunomodulatory
`effects. British Journal of Cancer, 87, 1166–1172.
`24. Crane E, List A (2005) Immunomodulatory drugs.
`Cancer Investigation, 23, 625–634.
`25. Revlimid Prescribing Information. Available at: http://
`www.revlimid.com/pdf/REVLIMID_PI.pdf (accessed
`August 1 2007).
`26. Richardson PG, Schlossman RL, Weller E et al. (2002)
`Immunomodulatory drug CC-5013 overcomes drug
`resistance and is well tolerated in patients with
`relapsed multiple myeloma. Blood, 100, 3063–3067.
`27. Zangari M, Tricot G, Zeldis J, Eddlemon P, Saghafi-
`far F, Barlogie B (2001) Results of a phase I study of
`CC-5013 for the treatment of multiple myeloma
`patients who relapse after high dose chemotherapy
`(HDCT) [abstract]. Blood, 98, 775a.
`28. Richardson P, Jagannath S, Hussein M et al. (2005) A
`multicenter, single-arm, open-label study to evaluate
`the efficacy and safety of single-agent lenalidomide
`in patients with relapsed and refractory multiple
`myeloma; preliminary results. Blood, 106, 449a.
`29. Richardson PG, Blood E, Mitsiades CS et al. (2006) A
`randomized phase 2 study of lenalidomide therapy
`for patients with relapsed or relapsed and refractory
`multiple myeloma. Blood, 108, 3458–3464.
`30. Blade J, Samson D, Reece D et al. (1998) Criteria for
`evaluating disease response and progression in
`patients with multiple myeloma treated by high-dose
`therapy and haemopoietic stem cell transplantation.
`Myeloma Subcommittee of the EBMT. European
`Group for Blood and Marrow Transplant. British
`Journal Haematology, 102, 1115–1123.
`31. Weber DM, Chen C, Niesvizky R et al. (2007) Lena-
`lidomide plus dexamethasone for relapsed multiple
`myeloma in North America. New England Journal of
`Medicine, 22, 357.
`32. Weber DM, Chen C, Niesvizky R et al.
`(2006)
`Lenalidomide plus high-dose dexamethasone pro-
`
`Ó 2008 Blackwell Publishing Ltd, Journal of Clinical Pharmacy and Therapeutics, 33, 219–226
`
`ALVOGEN, Exh. 1041, p. 0007
`
`

`

`226 X. Armoiry et al.
`
`vides improved overall survival compared to high-
`dose dexamethasone alone for relapsed or refractory
`multiple myeloma (MM): results of a North Ameri-
`can phase III study (MM-009), 2006 ASCO Annual
`Journal of Clinical
`Meeting Proceedings Part
`I.
`Oncology, 24(18S Suppl.), 7521.
`33. Dimopoulos M, Weber D, Chen C et al. (2005) Eval-
`uating oral lenalidomide and dexamethesone versus
`placebo and dexamethasone
`in patients with
`relapsed or refractory multiple myeloma. Haemoto-
`logica, 90 (Suppl. 2), 160. (Abstract 0402).
`34. Dimopoulos M, Spencer A, Attal M et al. (2007)
`Lenalidomide plus dexamethasone for relapsed or
`refractory multiple myeloma. New England Journal of
`Medicine, 357, 2123–2132.
`35. Wang M, Knight R, Dimopoulos M et al. (2006)
`Comparison of lenalidomide in combination with
`dexamethasone to dexamethasone alone in patients
`who have received prior thalidomide in relapsed or
`Journal of Clinical
`refractory multiple myeloma.
`Oncology, 24, 427s.
`36. Stadtmauer E, Weber D, Dimopolous M et al. (2006)
`Lenalidomide in combination with dexamethasone is more
`effective than dexamethasone at first relapse in relapsed
`multiple myeloma. ASH, Orlando (Abstract 3552).
`37. Palumbo A, Falco P, Corradini P et al.
`(2007)
`Melphalan, prednisone, and lenalidomide treatment
`for newly diagnosed myeloma: a report from the
`GIMEMA Italian multiple myeloma network. Journal
`of Clinical Oncology, 25, 4459–4465.
`
`38. National Institute of Health. Available at: http://
`www.clinicaltrials.gov/ct/show/NCT00405756?or
`der=1 (accessed 1 August 2007).
`39. National Institute of Health. Available at: http://
`clinicaltrials.gov/ct/show/NCT00430365?order=21
`(accessed 1 August 2007).
`40. National Institute of Health. Available at http://
`clinicaltrials.gov/ct/show/NCT00114101?order=2
`(accessed 1 August 2007).
`41. Richardson PG, Mitsiades C, Schlossman R, Munshi
`N, Anderson K (2007) New drugs for myeloma.
`Oncologist, 12, 664–689.
`42. Knop S, Gerecke C, Topp M et al. (2006) Lenalidomide
`

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