throbber
R E V I E W
`
`A review of lenalidomide in combination
`with dexamethasone for the treatment
`of multiple myeloma
`
`Teru Hideshima1
`Noopur Raje1,2
`Paul G Richardson1
`Kenneth C Anderson1
`1Jerome Lipper Multiple Myeloma
`Center, Dana-Farber Cancer Institute;
`2Massachusetts General Hospital
`Cancer Center, Harvard Medical
`School, Boston, MA, USA
`
`Correspondence: Teru Hideshima
`Dana-Farber Cancer Institute, Mayer 549,
`44 Binney Street, Boston, MA 02115, USA
`Tel +1 617 632 2144
`Fax +1 617 632 2140
`Email teru_hideshima@dfci.harvard.edu
`
`Abstract: Lenalidomide (also known as Revlimid®, CC-5013) is an immunomodulatory
`derivative of thalidomide and has more potent anti-tumor and anti-infl ammatory effects than
`thalidomide. The molecular mechanisms of anti-tumor activity of lenalidomide have been
`extensively studied in multiple myeloma (MM) both preclinical models and in clinical trials.
`Lenalidomide: directly triggers growth arrest and/or apoptosis of drug resistant MM cells;
`inhibits binding of MM cells to bone marrow (BM) extracellular matrix proteins and stromal
`cells; modulates cytokine secretion and inhibits angiogenesis in the BM milieu; and augments
`host anti-tumor immunity. Lenalidomide achieved responses in patients with relapsed refractory
`MM. Moreover, lenalidomide with dexamethasone (Dex) demonstrates more potent anti-MM
`activities than Dex both in vitro and in randomized phase III clinical trials. Specifi cally, the
`combination improved overall and extent of response, as well as prolonged time to progression
`and overall survival, resulting in FDA approval of lenalidomide with Dex for therapy MM
`relapsing after prior therapy.
`Keywords: lenalidomide, dexamethasone, multiple myeloma
`
`Introduction
`Multiple myeloma (MM) is a B cell malignancy characterized by excess monotypic
`plasma cells in the BM in association with monoclonal protein in serum and/or urine,
`decreased normal immunoglobulin (Ig) levels, and lytic bone disease. The 2006
`estimate of multiple myeloma incidence in the United States is 16,570 cases, with an
`estimated number of 11,300 deaths. Conventional therapies with alkylating agents,
`anthracyclines, and corticosteroids can extend patient survival to a median of 3–4
`years (Gregory et al 1992; Group 1998), and high dose therapy followed by autolo-
`gous transplantation can modestly prolong median survival to 4–5 years (Fermand
`et al 1998; Lenhoff et al 2000). Attempts to improve autografting include repeated
`use of high dose therapies (Desikan et al 2000; Attal et al 2003), as well as immune
`strategies to treat minimal residual disease post-transplant (Massaia et al 1999) can
`improve outcome in some studies, few, if any, patients are cured. MM remains incur-
`able due to the development of tumor cell resistance to all therapies, highlighting the
`urgent need for novel treatment strategies.
`Thalidomide (Thal) has shown to be useful in various diseases including MM;
`however, it is a potent teratogen and causes side effects including peripheral neuropathy
`(Tseng et al 1996). Attempts were therefore made to develop Thal analogs which are
`more potent and have less adverse effects: lenalidomide (C13H13N3O3, MW = 259.26) is
`one such analog belonging to the class ofi mmunomodulatory drugs (IMiDs) developed
`by the drug discovery program.
`
`Therapeutics and Clinical Risk Management 2008:4(1) 129–136
`© 2008 Dove Medical Press Limited. All rights reserved
`
`129
`
`IPR2018-01714
`Celgene Ex. 2001, Page 1
`
`

`

`Hideshima et al
`
`Preclinical studies of lenalidomide
`Overview (Figure 1)
`The interaction of multiple myeloma (MM) cells with bone
`marrow (BM) extracellular matrix (ECM) proteins and BM
`accessory cells, BM stromal cells (BMSCs), osteoblasts,
`osteoclasts, endothelial cells, as well as other factors in
`the BM milieu (ie, cytokines, angiogenesis) plays a crucial
`role in MM pathogenesis and drug resistance (Damiano
`et al 1999; Akiyama et al 2002; Hideshima and Anderson
`2002; Hideshima et al 2003, 2004, 2006; Chauhan et al
`2004). These accessory cells not only physically interact
`with MM cells, but also secrete growth and/or anti-
`apoptotic factors such as interleukin (IL)-6, insulin-like
`growth factor (IGF)-1, vascular endothelial growth factor
`(VEGF), and tumor necrosis factor (TNF)-α (Akiyama
`et al 2002; Chauhan et al 1996, 2004, 2005; Catley et al
`2004; Hideshima et al 2006). Delineation of the mecha-
`nisms of BM stromal cell (SC)-mediated MM cell prolif-
`eration, survival, drug resistance, and migration therefore
`provides the framework for identifi cation and validation
`of novel therapeutic targets.
`
`Within the BM microenvironment, several proliferative/
`antiapoptotic signaling cascades are activated in MM cells:
`phosphatidylinositol-3 kinase (PI3K)/Akt (also known as
`protein kinase B); I κ B kinase (IKK)/nuclear factor κ-B
`(NFκB); Ras/Raf/mitogen-activated protein kinase (MAPK)
`kinase (MEK)/extracellular signal-related kinase (ERK); and
`Janus kinase (JAK) 2/signal transducers and activators of
`transcription (STAT)-3 (Figure 1, Table 1). These signaling
`cascades mediate: cytoplasmic sequestration of many tran-
`scription factors; upregulation of cyclin D and anti-apoptotic
`Bcl-2 family members; as well as augmentation of telom-
`erase activity (Hideshima et al 2001a; Akiyama et al 2002).
`Importantly, these molecular events are triggered by both
`MM cell adherence to BMSCs and by cytokines secreted
`from BMSCs (Dankbar et al 2000; Hideshima et al 2004;
`Mitsiades et al 2004). Cytokines secreted from MM cells
`and BMSCs and other cells may in turn further augment
`cytokine secretion.
`Novel biologically based agents target not only the
`MM cell, but also MM cell–host interactions, cytokines,
`and their sequelae in the BM milieu. Thalidomide and
`
`Figure 1 Potential mechanisms of action of anti-MM activity of lenalidomide. Lenalidomide: directly induces tumor cell apoptosis and/or growth arrest (A); enhances NK
`and/or NK cell activity via activation of CD28/NF-AT2 pathway (B); inhibits MM cell adhesion to host microenvironment (C); inhibits angiogenesis (D); inhibits osteoclasto-
`genesis (E); as well as inhibits cytokine secretion (F).
`
`130
`
`Therapeutics and Clinical Risk Management 2008:4(1)
`
`IPR2018-01714
`Celgene Ex. 2001, Page 2
`
`

`

`its immunomodulatory derivative (IMiD) lenalidomide
`(Revlimid®; Celgene Corp., Summit, NJ, USA) are examples
`of such agents targeting the tumor cell in its BM milieu
`which can achieve responses even in refractory relapsed MM.
`Lenalidomide may inhibit MM cell growth by several differ-
`ent mechanisms (Figure 1). First, lenalidomide has a direct
`effect on MM cells to induce G1 growth arrest or apoptosis
`even of drug resistant cells (Hideshima et al 2000; Mitsiades
`et al 2002). Second, lenalidomide inhibits adhesion of MM
`cells to BMSCs, and thereby can overcome cell adhesion
`mediated drug resistance (CAM-DR); third, lenalidomide
`inhibits bioactivity and/or secretion in MM cells and/or BM
`stromal cells of cytokines [eg, interleukin (IL)-6, IL-1β,
`IL-10, and tumor necrosis factor (TNF)α] which augment
`MM cell growth, survival, drug resistance, migration, and
`expression of adhesion molecules. Importantly, lenalidomide
`is several thousand fold more potent than Thal at inhibiting
`TNFα/IL-1β secretion from mononuclear cells stimulated
`with lipopolysaccharide (LPS) in vitro (Corral et al 1999;
`Muller et al 1999). Fourth, vascular endothelial growth fac-
`tor (VEGF) and basic fi broblast growth factor (bFGF) are
`secreted by MM cells and/or BMSCs, and lenalidomide may
`inhibit activity of VEGF, bFGF, and angiogenesis in MM.
`Lenalidomide also acts against MM through immunomodu-
`latory effects such as augmentation of activity of cytotoxic
`T-cells and natural killer (NK) cells, associated with secretion
`of IL-2 and interferon-γ (Davies et al 2001; LeBlanc et al
`2004; Hayashi et al 2005).
`Bone destruction is a hallmark of MM, with 70%–80% of
`patients manifesting bone involvement. Recently, Anderson
`et al demonstrated that an IMiD CC-4047 (Actimid®; Celgene
`Corp., Summit, NJ, USA) inhibits osteoclastgenesis via
`downregulation of transcription factor PU.1 (Anderson et al
`2006). Lenalidomide also has inhibitory effect on osteoclas-
`togenesis (Terpos et al 2007).
`
`Table 1 Selected ongoing clinical trials of lenalidomide based
`combination treatment in multiple myeloma
`Agent
`Phase
`Perifoine + Dex
`I
`Hepatitis B vaccine
`I
`Doxorubicin + Dex
`I/II
`Bortezomib + Dex
`I/II
`Bortezomib + Dex
`II
`Bevacizumab + Dex
`II
`Clarithromycin + Dex
`II
`Dex
`III
`Dex
`III
`Dex
`IV
`
`Patient
`Rel/ref
`Rel/ref
`Rel/ref
`Newly diagnosed
`Rel/ref
`Rel/ref
`Newly diagnosed
`Newly diagnosed
`Previously treated
`Previously treated
`
`Lenalidomide plus dexamethasone in multiple myeloma
`
`Direct anti-tumor activities
`of lenalidomide
`Although the targets of whereby lelalidomide mediates anti-
`tumor activity of lenalidomide have not been fully delineated,
`several studies have examined the molecular mechanisms
`mediating sequelae of lenalidomide. Our previous studies
`demonstrated that lenalidomide induces G0/G1 growth arrest
`associated with p21Cip1 upregulation and/or apoptosis which
`is mediated via caspase-8 activation (Hideshima et al 2000;
`Mitsiades et al 2002). Lenalidomide inhibits LPS-mediated
`induction of Cox-2 and prostaglandin E2 (PGE2) production by
`a post-transcriptional mechanism in RAW 364.7 cells (Fujita
`et al 2001), suggesting that the anti-tumor activity induced
`by lenalidomide may also be due to inhibition of Cox-2 and
`PGE2. Lenalidomide inhibits nuclear factor (NF)-κB subunit
`activity in MM cell lines (Mitsiades et al 2002), which is
`consistent with reports that Thal inhibits DNA binding activ-
`ity of the p50/p65 NF-κB triggered by TNFα and IL-1β in
`Jurkat cell line (Keifer et al 2001) and in PBMCs (Rowland
`et al 2001). Since NF-κB plays an essential role in cell cycle
`regulation, cell survival, anti-apoptosis, and cytokine produc-
`tion in MM cells (Hideshima et al 2001b, 2002), inhibition of
`NF-κB activity by lenalidomide may also enhance or restore
`sensitivity to other chemotherapeutic agents. Specifi cally, we
`have demonstrated that MM cell adhesion-mediated upregula-
`tion of IL-6 is mediated via NF-κB activation (Chauhan et al
`1996; Hideshima et al 2002). Recently, Stewart et al (2004)
`reported pharmacogenomic studies suggesting that hyperac-
`tivation of the Wnt signaling antagonist DKK-1 is associated
`with response to the immunomodulators Thal and lenalido-
`mide. Furthermore, β-catenin expression is downregulated
`by lenalidomide in MM cell lines.
`Lenalidomide in combination with Dex is one of the most
`promissing MM novel treatment options. It induces at least
`additive direct cytotoxicity in MM cells (Hideshima et al
`2000), associated with activation of dual apoptotic signal-
`ing cascades: Dex induces caspase-9 (Chauhan et al 2001;
`Hideshima et al 2001a) and lenalidomide triggers caspase-8
`activation (Mitsiades et al 2002) (Figure 2). Most recently,
`enhanced anti-MM activity of rapamycin, a specifi c mTOR
`inhibitor, in combination with lenalidomide has been reported
`(Raje et al 2004). In this study, the combination of rapamycin
`plus lenalidomide overcomes drug resistance in MM cell
`lines resistant to conventional chemotherapy. Interestingly,
`differential signaling cascades, including the ERK and PI3-
`K/Akt pathways, are targeted by these drugs individually
`and in combination, suggesting the molecular mechanism
`by which they inhibits MM growth and survival.
`
`Therapeutics and Clinical Risk Management 2008:4(1)
`
`131
`
`IPR2018-01714
`Celgene Ex. 2001, Page 3
`
`

`

`Hideshima et al
`
`Figure 2 Potential mechanisms of synergistic cytotoxicity by lenalidomide plus
`Dex treatment in MM cells. Lenalidomide triggers caspase-8 dependent apoptosis,
`whereas Dex induces caspase-9 dependent apoptosis. The combination therefore
`triggers dual apoptotic signaling cascades.
`
`Anti-angiogenic activity
`Previous studies have shown that oral administration of
`lenalidomide attenuates growth factor-induced angiogenesis
`in vivo. This effect is correlated with the inhibitory effect
`of lenalidomide on growth factor-induced Akt phosphoryla-
`tion, thereby providing a potential mechanism for its anti-
`migratory and subsequent anti-angiogenic effects (Dredge
`et al 2005). In MM, an anti-angiogenic effect of Thal in
`vitro has been demonstrated (D’Amato et al 1004; Singhal
`et al 1999; Lentzsch et al 2002; Fujita et al 2004); however,
`to date no strong evidence of an anti-angiogenic effect of
`lenalidomide in vivo has been demonstrated. Moreover,
`Singhal et al (1999) reported no correlation of BM angiogen-
`esis with response to Thal in patients with relapsed refractory
`MM, suggesting that lenalidomide may mediate its anti-MM
`activity via mechanisms other than anti-angiogenesis.
`
`Immunomodulatory activities
`A unique feature of the anti-tumor effect of Thal and lenalido-
`mide is their ability to modulate and potentiate host immune
`responses against MM. Several studies have demonstrated
`the effects of lenalidomide on peripheral blood lymphocytes
`(Davies et al 2001; Haslett et al 2003; LeBlanc et al 2004;
`Hayashi et al 2005). Co-culture of naive splenocytes with
`
`anti-CD3 monoclonal antibody and IMiD1 (Actimid®)
`directly costimulates T cells and increases Th-1-type cyto-
`kines. Most excitingly, IMiDs augment CTL and NK cell
`activity against MM cell lines and autologous MM cells,
`associated with increased IL-2 levels in serum (Davies et al
`2001). Although Thal/IMiDs induce IL-2 secretion from
`T cells (Corral et al 1999; Shannon et al 2000), the mecha-
`nisms whereby these compounds induce IL-2 production
`from T cells has not totally been defi ned. Importantly, our
`recent studies demonstrated that lenalidomide signifi cantly
`costimulates proliferation of CD3+ T cells induced by CD3
`ligation, immature dendritic cells (DCs; SI, 2.1), or mature
`DCs (SI, 2.6). T-cell proliferation triggered by DCs is abro-
`gated by cytotoxic T lymphocyte antigen 4-immunoglobulin
`(CTLA-4-Ig). Lenalidomide also overcomes the inhibitory
`effects of CTLA-4-Ig on Epstein-Barr virus and infl uenza-
`specifi c CD4 and CD8 T-cell responses, as measured by
`cytokine capture and enzyme-linked immunosorbent spot
`(ELISPOT) assays. Importantly, lenalidomide triggers
`tyrosine phosphorylation of CD28 on T cells, followed by
`activation of NF-κB (LeBlanc et al 2004). Furthermore, we
`have demonstrated that IMiDs facilitate the nuclear translo-
`cation of nuclear factor of activated T cells (NF-AT)-2 and
`activator protein-1 via activation of PI3-K/Akt signaling,
`with resultant IL-2 secretion. IMiDs enhance both NK cell
`cytotoxicity and ADCC induced by triggering IL-2 produc-
`tion from T cells (Hayashi et al 2005). These studies therefore
`defi ne the molecular mechanisms whereby lenalidomide trig-
`gers NK cell-mediated cytotoxicity against MM cells, further
`supporting their therapeutic use in MM. More recently, we
`have shown that lenalidomide enhances ADCC induced by
`SGN-40, a humanized IgG1 anti-CD40 monoclonal antibody
`(Tai et al 2005).
`
`Clinical studies of lenalidomide
`Pharmacokinetics
`Pharmacokinetics (PK) of lenalidomide in MM patients
`has been reported by Wu and Scheffl er (2004) at American
`Society of Clinical Oncology in 2004. In this single-center,
`open-label, non-randomized, phase I dose escalation study
`in relapsed and refractory MM, the doses of lenalidomide
`used were 5, 10, 25 or 50 mg/day orally for 28 days. Blood
`samples were collected before and at 15 min, 30 min, 45 min,
`1 h, 1.5 h, 2 h, 2.5 h, 3 h, 4 h, 6 h, 8 h, 10 h, 12 h, 18 h, 24 h,
`48 h, and 72 h after administration on both days 1 and 28. No
`lenalidomide dose-limiting toxicity was observed at any dose
`level within the fi rst 28 days. Absorption of lenalidomide was
`rapid on both day 1 and 28, with tmax ranging from 0.7 to 2.0 h
`
`132
`
`Therapeutics and Clinical Risk Management 2008:4(1)
`
`IPR2018-01714
`Celgene Ex. 2001, Page 4
`
`

`

`at all dose levels. Plasma levels of lenalidomide declined in a
`monophasic manner, with elimination half-life ranging from
`2.8 to 6.1 h on both days 1 and 28 at all four doses. No plasma
`accumulation was observed upon multiple dosing. Importantly,
`daily oral doses of lenalidomide up to 50 mg produced no
`dose-limiting toxicity within the fi rst 28 days.
`The other PK study has been reported by Richardson et al
`(2006). In this study, plasma concentration of lenalidomide
`was determined in 39 patients during the fi rst and second
`cycles in both 15 mg and 30 mg dose groups, and when Dex
`was added due to progressive disease (PD) or stable disease
`(SD) on lenalidomide alone. The mean minimum (Cmin)
`plasma lenalidomide concentrations on days 1, 2, 3, 4, and
`21 during the fi rst and second 21-day cycles of lenalidomide
`alone and with the addition of Dex are shown for the 30 mg
`once-daily and 15 mg twice-daily cohorts. The average Cmin
`plasma levels were less in the twice-daily compared with
`daily dosing regimens. No obvious effect on lenalidomide
`plasma concentrations was seen with addition of Dex in either
`once- or twice-daily treatment.
`
`Clinical trials of lenalidomide
`Only a limited number of reports are available for clini-
`cal studies of lenalidomide (Bartlett et al 2004). A phase I
`clinical study of lenalidomide was completed at Dana-Farber
`Cancer Institute (Richardson et al 2002a). In this study,
`dose-escalation (5 mg/day, 10 mg/day, 25 mg/day, and
`50 mg/day) of lenalidomide was evaluated in 27 patients
`(median age 57 years; range, 40–71 years) with relapsed
`and refractory relapsed MM (Richardson et al 2002b). These
`patients received a median of 3 (range, 2–6) prior regimens,
`including autologous stem cell transplantation and Thal in
`15 and 16 patients, respectively. In 24 evaluable patients,
`no dose-limiting toxicity (DLT) was observed in patients
`treated at any dose level within the fi rst 28 days; however,
`grade 3 myelosuppression developed after day 28 in all 13
`patients treated with 50 mg/day lenalidomide. Dose reduction
`to 25 mg/day was well tolerated in 12 patients and therefore
`considered to be the maximal tolerated dose (MTD). Most
`importantly, no signifi cant somnolence, constipation, or
`neuropathy, the most common toxicities of Thal, have been
`seen in any cohort. Best responses of at least 25% reduction
`in paraprotein occurred in 17 of 24 (71%) patients (90%
`confi dence interval [CI], 52%–85%), including 11 (46%)
`patients who had received prior Thal; stable disease (less
`than 25% reduction in paraprotein) was observed in an addi-
`tional 2 (8%) patients. This study therefore demonstrates that
`lenalidomide can overcome conventional drug resistance,
`
`Lenalidomide plus dexamethasone in multiple myeloma
`
`even resistance to Thal. Given that lenalidomide is an oral
`agent, it is currently being evaluated in a randomized trial
`post autografting in an attempt to prolong progression free
`and overall survival.
`A multicenter, open-label, randomized phase II study
`to evaluate 2 dose regimens of lenalidomide for relapsed,
`refractory MM has been performed. In this study, 70 patients
`were randomized to receive either 30 mg once-daily or 15 mg
`twice-daily oral lenalidomide for 21 days of every 28-day
`cycle. An additional 32 patients received 30 mg once daily.
`Patients with progressive or stable disease after 2 cycles
`received additional Dex. Responses were evaluated according
`to European Group for Blood and Marrow Transplantation
`(EBMT) criteria. Overall response rate (CR+PR+MR) to
`lenalidomide alone was 25%; 24% for 30 mg once-daily and
`29% for 15 mg twice-daily cohort. Median overall survival in
`30-mg once-daily and 15 mg twice-daily groups was 28 and 27
`months, respectively. However, median progression-free sur-
`vival was 7.7 months on 30 mg once-daily versus 3.9 months
`on 15 mg twice-daily lenalidomide. Dex was added in 68
`patients and 29% responded. Importantly, time to fi rst occur-
`rence of clinically signifi cant grade 3/4 myelosuppression
`was shorter in the 15 mg twice-daily group (1.8 months) than
`30 mg once-daily (5.5 months, p = 0.05) group. Moreover,
`analysis of the fi rst 70 patients showed increased grade 3/4
`myelosuppression in patients receiving 15 mg twice-daily
`(41% vs 13%, p = 0.03). This study indicate that lenalidomide
`is active and well tolerated in relapsed, refractory myeloma,
`with the 30-mg once-daily regimen providing the basis for
`future studies as monotherapy and with Dex (Richardson
`et al 2006).
`
`Clinical studies of lenalidomide
`in combination with Dex
`As described above, preclinical studies have demonstrated
`the effi cacy of combination treatment of lenalidomide with
`Dex in MM and several clinical trials of this combination
`treatment have been completed.
`In two double blind, multicenter, international phase
`III clinical trials (MM-009, North American, 353 patients;
`MM-010, Europe, Australia, and Israel, 351 patients),
`patients with relapsed or refractory MM not resistant to Dex
`were treated with Dex 40 mg daily on days 1–4, 9–12, and
`17–20 every 28 days and were randomized to receive either
`lenalidomide 25 mg daily orally on days 1–21 every 28 days
`or placebo. At a median follow-up from randomization of
`17.1 months (MM-009) and 16.5 months (MM-010), both
`studies show signifi cant improvement with lenalidomide
`
`Therapeutics and Clinical Risk Management 2008:4(1)
`
`133
`
`IPR2018-01714
`Celgene Ex. 2001, Page 5
`
`

`

`Hideshima et al
`
`plus Dex compared to Dex in overall response (OR)
`(MM-009: 1% vs 20.5%, p ⬍ 0.001; MM-010: 59.1%
`vs 24%, p ⬍ 0.001, respectively), time to progression
`(TTP) (MM-009: 11.1 months vs 4.7 months, p ⬍ 0.001;
`MM-010: 11.3 months vs 4.7 months, p ⬍ 0.001, respec-
`tively), and overall survival (OS) (MM-009: 29.6 months
`vs 20.5 months, p ⬍ 0.001; MM-010: not estimable vs 20.6
`months, p ⬍ 0.001, respectively). In a subgroup analysis on
`patients with impaired creatinine clearance, no signifi cant
`difference in response rate, TTP, or OS was observed in
`patients with creatinine clearance above or below 50 mL/
`min who were treated with lenalidomide plus Dex; however,
`for 16 patients with creatinine clearance ⬍30 mL/min,
`median TTP and OS was shorter than for those with creati-
`nine clearance ⬎30 mL/min, but still signifi cantly longer
`than for patients treated with Dex (Weber et al 2006).
`Treatment with lenalidomide plus Dex in newly diagnosed
`MM patients has also reported by Rajkumar et al (2005). In
`this study, lenalidomide was given orally 25 mg daily on days
`1–21 of a 28-day cycle. Dex was given orally 40 mg daily
`on days 1–4, 9–12, and 17–20 of each cycle. Thirty-one of
`34 patients achieved an objective response, including 2 (6%)
`achieving complete response (CR) and 11 (32%) meeting
`criteria for both very good partial response and near complete
`response, resulting in an overall objective response rate of
`91%. This study indicated that lenalidomide plus Dex is a
`highly active regimen with manageable side effects in the
`treatment of newly diagnosed MM.
`A number of studies demonstrated that MM is character-
`ized by cytogenetic abnormalities causing dysregulation of
`the genes at the breakpoints, and by point mutations (Kuehl
`et al 2002; Fonseca et al 2004; Carrasco et al 2006). Spe-
`cifi cally, chromosome 13 deletions are present in over 50%
`of MM patients and considered to be associated with poor
`prognosis. In addition, t(4; 14) in MM also predicts poor
`response to conventional and high dose treatment and short-
`ened survival. A recent study has shown that lenalidomide
`overcomes the poor prognosis conferred by chromosome 13
`deletion and t(4; 14) in MM patients, evidenced by event free
`survival and response rate (Bahlis et al 2006).
`Recently, a phase I/II 3 combination treatment of
`lenalidomide, Dex and adriamycin (RAD therapy) for
`relapsed MM patient has been reported. In this study, 31
`patients were evaluated for response and toxicity: 26 patients
`achieved reduction of paraprotein levels of at least 50% for
`a response rate of 84%, including one confi rmed CR and
`14 PRs according to the EBMT criteria. Importantly, 8 of
`10 patients who displayed del (13) on cytogenetic analysis
`
`responded, including 6 confi rmed PRs. One patient each
`experienced acute renal failure due to emesis and hypo-
`volemia, pneumocystis pneumonitis, and catheter related
`infection. Somnolence, constipation, thromboembolism,
`or neuropathy was not observed. This study showed that
`RAD induces substantial responses with an acceptable
`toxicity profi le, and thus signifi cantly contributes to the
`therapeutic armamentarium even in heavily pretreated MM
`patients (Knop et al 2006). Most recently, a phase I study of
`lenalidomide and dexamethasone in combination with Akt
`inhibitor perifosine for patients with relapsed or refractory
`MM, and a phase I/II study of lenalidomide, dexamethasone
`and bortezomib combination therapy for newly diagnosed
`MM patients are ongoing.
`The common side effects of lenalidomide treatment in
`phase 2 clinical trials of relapsed refractory MM are sum-
`marized in Table 1. The most common toxicities associated
`with Thal (eg, constipation, neuropathy, tremors) were not
`observed. Toxicities associated with lenalidomide were
`primarily hematologic and reversible. The most common
`grade 3 or higher adverse events during lenalidomide therapy
`were neutropenia and thrombocytopenia. Grade 4 neutro-
`penia occurred in 2 of 34 (5.8%) patients treated at 15 mg
`twice daily vs 4 of 68 (5.9%) patients treated with 30 mg
`daily. Grade 4 thrombocytopenia occurred in 2 (5.8%) of
`34 patients on 15 mg twice daily vs 2 of 68 patients (2.9%)
`treated with 30 mg daily. Deep vein thrombosis (DVT)
`was reported in 1 patient on the 30 mg daily and 2 patients
`(5.8%) on the 15 mg twice-daily treated regimen. Sedation
`or neurologic toxicities were not observed in most of these
`studies (Richardson et al 2006). The differences in the side
`effect profi le between Thal and lenalidomide may refl ect
`distinct patterns of antiangiogenic, cytokine-related, micro-
`environmental, and immunomodulatory activity, rather than
`distinct separate mechanisms of action.
`In phase III trials of lenalidomide plus Dex for newly
`diagnosed MM patients, 47% of patients experienced grade
`III or higher nonhematologic toxicity. The most common
`adverse effects were fatigue (15%), muscle weakness (6%),
`anxiety (6%), pneumonitis (6%), and rash (6%) (Rajkumar
`et al 2006a). Recently, a randomized phase III trial of
`lenalidomide plus high-dose Dex versus lenalidomide plus
`low-dose Dex in newly diagnosed MM has also reported
`by Rajkumar et al (2006b). In this study, patients in both
`arms received lenalidomide 25 mg/day orally on days 1–21
`every 28 days. In addition, patients in the high-dose Dex
`arm received Dex 40 mg on days 1–4, 9–12, and 17–20
`orally every 28 days, while patients in the low-dose Dex
`
`134
`
`Therapeutics and Clinical Risk Management 2008:4(1)
`
`IPR2018-01714
`Celgene Ex. 2001, Page 6
`
`

`

`arm received Dex 40 mg on days 1, 8, 15, and 22 orally
`every 28 days. Although response rate (RR) has not yet
`been reported, toxicity rates are higher in the high-dose
`Dex arm than low-dose Dex arm. For example, Grade 3
`and above toxicities in cardiac ischemia (2.7% vs 0.5%),
`hypercalcemia (5.8% vs 1.8%), infection (18.8% vs 9%),
`thromboembolism (18.4% vs 5.4%), and non-hematologic
`toxicities (22% vs 12.6%) are higher in high-dose Dex arm
`than low-dose Dex arm. If RR is similar in both arms, dos-
`age of Dex can be reduced to 25 mg.
`
`Conclusion
`Lenalidomide plus Dex treatment is highly effective in
`both preclinical and clinical studies. It is one of the prom-
`ising treatment options against both relapsed/refractory
`and newly diagnosed MM patients. Adverse effects of this
`combination can be markedly reduced by lowering the
`Dex dosage.
`
`Future directions
`Lenalidomide plus Dex treatment can be further combined
`with other novel or conventional agents to improve patient
`outcome in MM. Indeed, potent Akt inhibitor Perifosine,
`proteasome inhibitor bortezomib (Velcade®; Millennium
`Pharmaceuticals Inc.), and anti-angiogenic agent bevaci-
`zumab (Avastin®; Genentech) are already under evaluation
`in combination clinical trials.
`
`Acknowledgments
`Supported by National Institutes of Health Grant PO-1
`78378 and RO-1 CA 50947; the Doris Duke Distinguished
`Clinical Research Scientist Award (KCA); the Multiple
`Myeloma Research Foundation (TH, NR, KCA); and the
`Myeloma Research Fund (KCA).
`
`References
`
`Akiyama M, Hideshima T, Hayashi T, et al. 2002. Cytokines modulate
`telomerase activity in a human multiple myeloma cell line. Cancer
`Res, 62:3876–82.
`Anderson G, Gries M, Kurihara N, et al. 2006. Thalidomide derivative
`CC-4047 inhibits osteoclast formation by down-regulation of PU.1.
`Blood, 107:3098–105.
`Attal M, Harousseau JL, Facon T, et al. 2003. Single versus double auto-
`logous stem-cell transplantation for multiple myeloma. N Engl J Med,
`349:2495–502.
`Bahlis NJ, Mansoor A, Lategan JC, et al. 2006. Lenalidomide overcomes
`poor prognosis conferred by deletion of chromosome 13 and t(4; 14)
`in multiple myeloma: MM016 Trial. Blood, 108:1016a.
`Bartlett JB, Dredge K, Dalgleish AG. 2004. The evolution of thalidomide and
`its IMiD derivatives as anticancer agents. Nat Rev Cancer, 4:314–22.
`Carrasco DR, Tonon G, Huang Y, et al. 2006. High-resolution genomic
`profi les defi ne distinct clinico-pathogenetic subgroups of multiple
`myeloma patients. Cancer Cell, 9:313–25.
`
`Lenalidomide plus dexamethasone in multiple myeloma
`
`Catley L, Tai YT, Shringarpure R, et al. 2004. Proteasomal degradation of
`topoisomerase I is preceded by c-Jun NH2-terminal kinase activation,
`Fas up-regulation, and poly(ADP-ribose) polymerase cleavage in
`SN38-mediated cytotoxicity against multiple myeloma. Cancer Res,
`64:8746–53.
`Chauhan D, Catley L, Li G, et al. 2005. A novel orally active proteasome
`inhibitor induces apoptosis in multiple myeloma cells with mechanisms
`distinct from Bortezomib. Cancer Cell, 8:407–19.
`Chauhan D, Hideshima T, Rosen S, et al. 2001. Apaf-1/cytochrome c
`independent and Smac dependent induction of apoptosis in multiple
`myeloma cells. J Biol Chem, 276:24453–6.
`Chauhan D, Li G, Hideshima T, et al. 2004. Blockade of ubiquitin-
`conjugating enzyme CDC34 enhances anti-myeloma activity of
`Bortezomib/Proteasome inhibitor PS-341. Oncogene, 23:3597–602.
`Chauhan D, Uchiyama H, Akbarali Y, et al. 1996. Multiple myeloma cell
`adhesion-induced interleukin-6 expression in bone marrow stromal
`cells involves activation of NF-kB. Blood, 87:1104–12.
`Corral LG, Haslett PAJ, Muller GW, et al. 1999. Differential cytokine modu-
`lation and T cell activation by two distinct classes of thalidomide ana-
`logues that are potent inhibitors of TNF-α. J Immunol, 163:380–6.
`D’Amato RJ, Loughman MS, Flynn E, et al. 1994. Thalidomide is an inhi-
`bitor of angiogenesis. Proc Natl Acad Sci USA, 91:4082–5.
`Damiano JS, Cress AE, Hazlehurst LA, et al. 1999. Cell adhesion mediated
`drug resistance (CAM-DR): Role of integrins and resistance to apoptosis
`in human myeloma cell lines. Blood, 93:1658–67.
`Dankbar B, Padro T, Leo R, et al. 2000. Vascular endothelial growth factor
`and interleukin-6 in paracrine tumor-stromal cell interactions in multiple
`myeloma. Blood, 95:2630–6.
`Davies FE, Raje N, Hideshima T, et al. 2001. Thalidomide and immu-
`nomodulatory derivatives augment natural killer cell cytotoxicity in
`multiple myeloma. Blood, 98:210–16.
`Desikan R, Barlogie B, Sawyer J, et al. 2000. Results of high-dose therapy
`for 1000 patients with multiple myeloma: durable complete remissions
`and superior survival in the absence of chromosome 13 abnormalities.
`Blood, 95:4008–10.
`Dredge K, Horsfall R, Robinson SP, et al. 2005. Orally administered
`lenalidomide (CC-5013) is anti-angiogenic in vivo and inhibits endo-
`thelial cell migration and Akt phosphorylation in vitro. Microvasc
`Res, 69:56–63.
`Fermand J-P, Ravaud P, Chevret S, et al. 1998. High-dose therapy and
`autologous peripheral blood stem cell transplantation in multiple
`myeloma: Up-front or rescue treatment? Results of a multicenter
`sequential randomized clinical trial. Blood, 92:3131–6.
`Fonseca R, Barlogie B, Bataille R, et al. 2004. Genetics and cytogenetics of
`multiple myeloma: a workshop report. Cancer Res, 64:1546–58.
`Fujita J, Mestre JR, Zeldis JB, et al. 2001. Thalidomide and its analogues
`inhibit lipopolysaccharide-mediated Iinduction of cyclooxygenase-2.
`Clin Cancer Res, 7:3349–55.
`Fujita K, Asami Y, Tanaka K, et al. 2004. Anti-angiogenic effects of
`thalidomi

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