throbber
Plenary paper
`
`Iin1nun01nodulatory drug CC-5013 overc01nes drug resistance and is well
`tolerated in patients with relapsed multiple myel01na
`
`Paul G. Richardson , Robert L. Schlossman , Edie Weller, Teru Hideshima , Constantine Mitsiades , Faith Davies , Richard LeBlanc,
`Laurence P. Calley, Deborah Doss, Kathleen Kelly, Mary McKenney, Julie Mechlowicz, Andrea Freeman, Reggie Deocampo,
`Rebecca Rich , Joan J . Ryoo, Dharminder Chauhan, Kathe Balinski, Jerome Zeldis, and Kenn eth C. Anderson
`
`Thalidomide (Thal) can overcome drug
`resistance in multiple rnyelorna (MM) but
`is associated with somnolence, constipa(cid:173)
`tion, and neuropathy. In previous in vitro
`studies, we have shown that the potent
`irnrnunornodulatory derivative of thalido(cid:173)
`mide (IMiD) CC-5013 induces apoptosis
`or growth arrest even in resistant MM cell
`lines and patient cells, decreases binding
`of MM cells to bone marrow strornal cells
`(BMSCs), inhibits the production in the
`BM milieu of cytokines (interleukin-6 [IL-
`6), vascular endothelial growth factor
`[VEGFJ, tumor necrosis factor-a [TNF-a))
`mediating growth and survival of MM
`cells, blocks angiogenesis, and stimu(cid:173)
`lates host anti-MM natural killer (NK) cell
`immunity. Moreover, CC-5013 also inhib(cid:173)
`its tumor growth, decreases angiogen-
`esis, and prolongs host survival in a
`
`human plasmacytoma mouse model. In
`the present study, we carried out a phase 1
`CC-5013 dose~scalation (5 mg/d, 10 mg/d,
`25 mg/d, and 50 mg/d) study in 27 patients
`(median age 57 years; range, 40-71 years)
`with relapsed and refractory relapsed MM.
`They received a median of 3 prior regi(cid:173)
`mens (range, 2-6 regimens), including
`autologous stern 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 first 28
`days; however, grade 3 myelosuppres(cid:173)
`sion developed after day 28 in all 13
`patients treated with 50 mg/d CC-5013. In
`12 patients, dose reduction to 25 mg/d
`was well tolerated and therefore consid(cid:173)
`ered the maximal tolerated dose (MTD).
`Importantly, no significant somnolence,
`
`Introduction
`
`constipation, or neuropathy has been
`seen in any cohort. Best responses of at
`least 25% reduction in paraprotein oc(cid:173)
`curred in 17 (71 %) of 24 patients (90%
`confidence interval [Cl], 52%-85%), includ(cid:173)
`ing 11 (46%) patients who had received
`prior Thal. Stable disease (less than 25%
`reduction in paraprotein) was observed in
`an additional 2 (8%) patients. Therefore,
`17 (71 %) of 24 patients (90% Cl, 52%-85%)
`demonstrated benefit from treatment. Our
`study therefore provides the basis for the
`evaluation of CC-5013, either alone or in
`combination, to treat patients with MM at
`earlier stages of disease. (Blood. 2002;
`100:3063-3067)
`
`Multiple myeloma (MM) is the second most common hemato(cid:173)
`logic malignancy, affecting 14 400 new and 50 000 total patients
`in the United States in 200 I, 1 and remains incurable despite
`conventional and high-dose chemotherapy. To overcome resis(cid:173)
`tance lo current therapies and improve patient outcome, novel
`biologically based treatment approaches are needed that target
`mechanisms whereby MM cells grow and survive in the bone
`marrow (BM). Thalidomide (Thal), used empirically to treat
`MM based upon its antiangiogenic activity and the increased
`angiogenesis observed in MM BM, ach ieves responses even in
`refractory, relapsed discase. 1 Importantly, our in vitro and in
`vivo preclinical studies both define a role for MM-host interac(cid:173)
`tions in regulating MM cell growth, survival, drug resistance,
`and migration in the BM 3• 12 and demonstrate that Thal targets
`the MM cell in its BM milieu to overcome classical drug
`resistance both in vitro and in vivo in animal models. 1J- Ir,
`However, Thal has significant and dose-limiting side effects,
`including somnolence, constipation, and neuropathy, which has
`
`prompted the search for more potent and less toxic Thal
`derivatives.
`Tmmunomodulatory drugs (!MiDs) are potent Thal derivatives
`that markedly stimulate T-cell proliferation, as well as interleukin-2
`(IL-2) and interferon--y (IFN--y) production, but do not inhibit
`phosphodiesterase-4 (PDE-4). 17 CC-5013 (REVIMID) is 50 to
`2000 times more potent than Thal in stimulating T-cell proliferation
`triggered via the T-ccll receptor (TCR) and 50 to I 00 times more
`potent lhan Thal in augmenting IL-2 and IFN--y production. In
`addition, CC-5013 triggers dose-dependent decreased secretion of
`tumor necrosis factor-a (TNF-a), IL-I 13, and IL-6 and triggers
`increased secretion of IL- I 0. The CC-5013 concentration for 50%
`inhibition (TC50) of lipopolysaccharide (LPS)-induccd TNF-a
`secretion by peripheral blood mononuclear cells (PBMCs) is about
`I 00 nM (25.9 ng/mL), whereas Thal has an IC50 of about 194 µM
`(50.2 µg/mL). 1x Based upon these more potent effects of !MiDs
`than Thal on normal cells, we compared their relative anti-MM
`activitit:s. Our in vitro studies show an IC50 of 0.4 µM (I 03.6 ng/mL)
`
`From the Jerome Lipper Multiple Myeloma Center, Dana-Farber Cancer
`Institute, Harvard Medical School, Boston , MA; and Celgene, Warren, NJ.
`
`Submitted April 2, 2002; accepted May 1, 2002. Prepublished online as Blood
`First Edition Paper, July 12, 2002; DOI 10.1182/blood-2002-03-0996.
`
`Supported by National Institutes of Health grants R0-1 50947 and P0-78378,
`the Multiple Myeloma Research Foundation (T.H., C.M., R.L.), the Myeloma
`Research Fund (K C.A.), and the Doris Duke Distinguished Clinical Research
`
`Scientist Award (K.C.A.).
`
`Reprints: Kenneth C. Anderson, Dana-Farber Cancer Institute, Mayer 557, 44
`Binney St, Boston, MA 02115; e-mail: kenneth_anderson@dfci.harvard.edu.
`
`The publication costs of this article were defrayed in part by page charge
`payment. Therefore, and solely to indicate this fact, this article is hereby
`marked "advertisement" in accordance with 18 U.S.C. section 1734.
`
`BLOOD, 1 NOVEMBER 2002 • VOLUME 100, NUMBER 9
`
`3063
`
`DR. REDDY’S LABS., INC. EX. 1044 PAGE 1
`
`

`

`3064
`
`RICHARDSON el al
`
`BLOOD, 1 NOVEMBER 2002 • VOLUME 100, NUMBER 9
`
`Figure 1. Mechanisms of action of CC-5013 targeting
`MM cells and the BM microenvironment. (A) MM cell
`G1 growlh arrest and apoplosis. (B) Decreased MM cell
`lo BMSC binding. (C) Decreased cylokine activity. (D)
`Decreased cylokine production in BM. (E) Decreased
`angiogenesis. (F) Induced host anti-MM immune re(cid:173)
`sponse.
`
`I C. CC-5013 I
`MM cells ~ tt.' D. CC-5013 ]
`~Ge;) I B. CC-5013 1~~~:tt
`I A. CC-5013 j...
`.,
`Bone Marrow
`J
`ICAM-1 ~ StrornaJ Cells
`-~ Bone Marrow
`{
`e ~
`VEGFt
`• @~'\IL-2t
`«:>Vessels
`bF~ -@~
`~ @~
`
`PBMC
`
`CDS+ cells
`NK cells
`
`IFNyt
`
`t
`IF. CC-5013 [
`
`for CC-5013 against MM cell lines and patient cells that are
`resistant to conventional therapy; in contrast, even at concentra(cid:173)
`tions up to 100 µM (25.8 µg/mL), Thal decreases MM cell
`proliferation by only 15% or 20%. 13 Our studies further demon(cid:173)
`strate that CC-5013 decreases binding of MM cells to bone marrow
`stromal cells (BMSCs), inhibits the production in the BM milieu of
`cytokines (IL-6, vascular endothelial growth factor [VEGF], TNF-a)
`mediating growth and survival of MM cells, blocks angiogenesis,
`and stimulates host anti-MM natural killer (NK) cell immunity 13·15
`(Figure I). In addition, we detennined that CC-5013 inhibits tumor
`growth, decreases angiogenesis, and prolongs host survival in a
`human plasmacytoma mouse model. 16 These preclinical studies
`suggest that CC-5013 may overcome drug resistance, even to Thal,
`in MM cells.
`The remarkable in vitro and in vivo activity ofCC-5013 against
`resistant MM cells in preclinical studies provided the framework
`for this phase I dose-escalation trial of CC-5013 in patients with
`relapsed and refractory MM. Importantly, CC-5013 achieved either
`response or stabilization of disease in 17 (71 %) of 24 evaluable
`patients (90% confidence interval [CI], 52%-85%) and demon(cid:173)
`strated a favorable toxicity profile. Our study therefore provides the
`basis for the evaluation of CC-5013, either alone or in combination,
`to treat patients with MM at earlier stages of disease.
`
`Patients, materials, and methods
`
`Study objectives
`
`The primaiy objectives of this study were to identify the maximum
`tolerated dose (MTD) and to evaluate the safety of CC-5013 when given at
`doses of 5 mg/d to 50 mg/din patients with rcfractmy and/or relapsed MM.
`A seconda1y objective was to evaluate the response to CC-5013.
`
`Study design
`
`A standard dose escalation of CC-50 J3 (5, I 0, 25, and 50 mg/d) was
`pe1to1med to identify the MTD under the auspices ofan institutional review
`board (!RB) protocol and after obtaining infonned consent. Three patients
`were enrolled at each dose level, with up to 6 patients assigned to each dose
`level, depending on dose-limiting toxicity (DLT). DLT is defined to be at
`least grade 2 cardiac a1i-hythmia, at least grade 3 nonhematologic toxicity,
`or grade 4 hematologic toxicity using National Cancer Institute (NCI)
`common toxicity criteria during the first 4 weeks of treatment. Following
`
`the standard design, if no DLTs were observed among the first cohort of 3
`patients at each dose level, the next set of3 patients was entered on the next
`highest dose. lf2 or more DLTs were observed, the previous dose level was
`identified as the MTD. If I DLT was observed among the initial 3 patients,
`an additional 3 patients would be entered. If none of !he additional 3
`patients experienced a DLT, the dose was escalated; othe1wise, the lower
`dose was identified as the MTD. Ten additional patients were enrolled at the
`MTD to better define the toxicity rate (Figure 2).
`Patients were assessed before and weekly for the first month after
`treatment. Patients were hospitalized in the clinical research unit for at least
`72 hours after the first dose of CC-5013 and overnight on day 28. Patients
`who did not have either disease progression or DLT at day 28 continued on
`treatment until disease relapse or progression, with monthly follow-up.
`Safety and efficacy assessments were pe1formed at each visit, with blood
`and urine samples collected for phannacokinetics on days I to 4 and day 28.
`
`Patient selection
`
`Patients with relapsed and/or refractory MM were eligible and must have
`foiled to respond to at least 2 prior regimens of treatment. Patients were
`considered refracto1y if they had progressive disease by Southwest
`Oncology Group (SWOG) criteria (an increase by more than I 00% of the
`lowest level of protein production). Relapse following a remission was
`defined by SWOG criteria as any of !he following: (I) more than 25%
`increase in M protein from baseline levels, (2) reappearance of M
`paraprotcin, or (3) a definite increase in the size and number of lytic bone
`lesions recognized on radiographs (compression fractures per se did not
`constitute a relapse). Prior therapy with Thal was allowed, provided that it
`was tolerated. Exclusion criteria included a prolonged QT interval (> 430
`ms); predisposition to cardiac anythmias; concomitant medications known
`to prolong the QT segment; at least grade 3 peripheral ncuropathy;
`inadequate renal function (serum creatinine > 1.5 mg/dL); evidence of
`
`N=3
`
`5 mg/day
`
`N=3
`
`10 mg/day -
`
`N=J
`25 mg/day
`
`-
`
`N=3
`50 mg/day
`
`DLT:2: grade 2 cardiac arrythmia, 2: grade 3 nonhematologic
`toxicity, 2: grade 4 hematologic toxicity. If 1 DLT, add 3 p ati<mtsl<-(cid:173)
`at that dose level: If 2:2 DLT, stop : MTD=dose lcliol below DLT
`At MTD add 10 patients
`
`Figure 2. Schema of CC-5013 phase 1 trial in myeloma.
`
`DR. REDDY’S LABS., INC. EX. 1044 PAGE 2
`
`

`

`BLOOD, 1 NOVEMBER 2002 • VOLUME 100, NUMBER 9
`
`CC-5013 IN MYELOMA
`
`3065
`
`mucosa! or internal bleeding; thrombocytopenia (platelets < 50 X I 09/L
`[50 0001µ.L]); neutropcnia (absolute ncutrophil count< I X 109/L [10001 Results
`µ.L]); pregnant or lactating women; and women of childbearing potential
`who were not using adequate contraception.
`
`Patients treated
`
`Treatment
`
`Each patient received CC-5013 as a single daily oral dose. Depending on
`the order of study entiy and the tolerability of prior dose levels, patients
`received CC-5013 at 5, I 0, 25, or 50 mgld for the first 4 weeks. Accrual to
`the next higher dose level did not occur until the safety and tolerability of
`CC-5013 at prior dose level(s), when given for at least 4 weeks, had been
`established. Patients experiencing DLT during the first 28 days had therapy
`discontinued. After the first 4 weeks of treatment, dose reduction was
`permitted to manage any toxicity.
`
`Safety parameters
`
`Prior to enrollment, at each weekly and monthly visit, and either at the
`completion of the study or at premature discontinuation, evaluation of each
`patient included medical history and physical examination (to include
`assessment of peripheral neuropathy and measurement of vital signs), query
`for adverse events and concomitant medication use, and clinical laboratory
`testing (blood chemistry, hematology, urinalysis, and thyroid function
`testing [every 3 months]). Electrocardiograms (ECGs) were recorded at the
`first 4 weekly visits and monthly thereafter for the first 12 months of
`treatment. During the 72-hour inpatient period following the first dose
`administration, vital signs, ECGs, and adverse events were monitored and
`recorded at I, 2, 4, and 8 hours. Adverse events and vital sign measurements
`were recorded evety 8 hours thereafter, and ECGs were perfonned on days
`2, 3, and 4. Severity of adverse experiences, including any clinical
`laboratory and vital sign abnormalities, were evaluated using common
`toxicity criteria. No drug-related renal compromise was observed.
`
`Response
`
`Response to treatment was assessed by using M-protein quantification (by
`protein electrophoresis) in serum and a 24-hour urine collection at
`screening, start of therapy, day 14 and day 28, as well as monthly thereafter
`( or upon early termination). Bone marrow aspirations and biopsies were
`petformed at baseline, 3, 6, and 12 months, and/or at completion of therapy.
`A skeletal survey was also pe1formed in patients who had abnonnal
`pretreatment studies or as clinically indicated.
`
`Pharmacokinetics
`
`Blood and urine samples were collected for analysis of phannacokinetic
`parameters on days I and 28. Specifically, blood samples were collected
`before dose as well as at 0.25, 0.5, 0.75, I, 1.5, 2, 2.5, 3, 4, 6, 8, I 0, 12, 18,
`and 24 hours after dose. In addition, blood samples were collected weekly
`for CC-5013 level detenninations. Total urine was collected and assayed at
`0 to 4, 4 to 8, 8 to 12, and 12 to 24 hours after drug administration.
`Appropriate noncompartmental phannacokinetic parameters were esti(cid:173)
`mated using the actual blood sampling times for each patient on day I and
`day 28. Values for noncompartmental pharmacokinetic parameters were
`calculated based on CC-5013 plasma concentration-time data obtained
`during each dosing period.
`
`Twenty-seven patients with a median age of 57 years (range, 40-71
`years) were enrolled. Two patients were removed from study on the
`first day of treatment due to rapid disease progression resulting in
`renal dysfunction, a known complication of MM, which rendered
`them ineligible. Of the 25 patients who received therapy, 19 were
`men and 6 were women. Immunoglobulin G (IgG) MM was
`present in 15 (60%), IgA in 7 (28%), and light-chain-only disease
`in 3 (12%) patients. Fifteen (60%) patients had undergone prior
`autologous stem cell transplantation (SCT), and 16 (64%) had
`received prior Thal. Patients received a median of 3 prior regimens
`(range, 2-6 regimens); all patients had relapsed MM, and 18 (72%)
`were refractory to salvage therapy.
`
`Toxicity profile
`
`The first 3 patients were treated for 28 days with CC-5013 5 mg/d
`without DLT. In the second cohort of3 patients treated at 10 mg/d,
`I patient had DLT characterized by grade 3 leukopenia and
`neutropenia, resulting in her removal from study before day 28; 2
`other patients tolerated the drug without complication, and an
`additional 3 patients treated at IO mg/d also demonstrated no DLT.
`The patient who had the DLT previously experienced a similar
`reaction to Thal and to dexamethasone (Dex). CC-5013 was well
`tolerated within the first 28 days in all 3 patients treated at 25 mg/d;
`however, grade 3 thrombocytopenia and grade 4 neutropenia
`developed during the second month, resulting in 2 patients being
`removed from study. All 3 patients treated with CC-5013 at 50
`mg/d tolerated therapy without DLT in the first 28 days, and an
`additional IO patients were treated at 50 mg/d to better define
`toxicity and outcome. None of the patients (0 of 13) treated at 50
`mg/d had DLT in the first 28 days (90% CI, 0%-21 %).
`Overall, the median duration ofCC-5013 therapy was 6 months
`(range, 2 weeks to 16 months), and 10 patients continued on
`treatment (4 patients at 25 mg/d, 3 patients at 10 mg/d, and 3
`patients at 5 mg/d). Importantly, no significant somnolence,
`constipation, or neuropathy has been seen in any cohort. Grade 3
`and 4 myelosuppression developed in 12 of 13 patients treated with
`50 mg/d CC-5013 beyond 28 days, eventually prompting dose
`reduction and granulocyte colony-stimulating factor (G-CSF) sup(cid:173)
`port in all 12 patients, with a median duration of 3 months (range,
`2-4 months) of CC-5013 therapy at 50 mg/d. All 12 patients who
`were reduced to 25 mg/d of CC-5013 tolerated the lower dose, and
`in aggregate 13 patients remained on therapy at this dose for a
`median of 4 months (range, 2-8 months). Based on greater
`exposures to CC-5013 for more than 28 days, we therefore
`conclude that 25 mg/d is the MTD. The most common adverse
`events for treated patients (n = 25), including those observed after
`day 28, are presented in Figure 3. Grade 3 neutropenia occurred in 15
`(60%) of25 patients (90% CI, 42%-76%) and grade 4 neutropenia in 4
`(16%) of 25 patients (90% CI, 6%-33%). Grade 3 thrombocytopenia
`occurred in 5 (20%) of25 patients (90% CI, 8%-38%).
`
`Biostatistical analysis
`
`Response
`
`The MTD was identified using a standard phase I design (Figure 2). With
`this design, the probability of dose escalation was 0.97 if the trne DLT rate
`was 5%. The 90% confidence intervals are repmted on the percentage of
`patients who experience toxicity at the MTD and the percentage of patients
`who achieved reduction in their paraprotein.
`
`Maximal paraprotein reductions observed in 24 patients who
`received at least 28 days of treatment are summarized in Table I.
`Best responses of at least 25% reduction in paraprotein occurred in
`17 (71 %) of 24 evaluable patients (90% CI, 52%-85%), including
`at least 50% reduction in paraprotein in 7 (29%) of 24 patients
`
`DR. REDDY’S LABS., INC. EX. 1044 PAGE 3
`
`

`

`3066
`
`RICHARDSON et al
`
`BLOOD, 1 NOVEMBER 2002 • VOLUME 100, NUMBER 9
`
`Llghlheadedness
`
`t=:::J
`
`• Grade4
`llilGrade3
`0Grad&2
`DGrade 1
`
`Raoh ~
`
`Leg cramps
`
`Thrombocytopenla -
`
`0
`
`10
`
`20
`
`30
`
`40
`
`50
`
`60
`
`70
`
`BO
`
`90 100
`
`% of Patients
`Figure 3. Profile of adverse events. Severity of adverse events was assessed using
`common toxicity criteria for the entire duration of therapy (n = 25 patients).
`
`(90% CI, 15%-48%). Stable disease (25% or less reduction in
`paraprotein) was noted in an additional 2 (8%) patients. Therefore,
`17 (71%) of24 patients (90% CI, 52%-85%) demonstrated benefit
`from treatment, including 11 (46%) patients who had received prior
`Thal. Eleven (85%) of 13 patients treated at 50 mg/d had at least
`25% paraprotein reductions (90% CI, 59%-97%), and 5 (38%) of
`13 patients (90% CI, 17%-65%) had at least 50% declines. Most
`responses have been at 25 mg/d (2 of 3 patients) and 50 mg/d
`CC-5013 (12 of 13 patients), although clinical activity was also
`seen in patients treated at both 5 mg/d (3 of3 patients) and IO mg/d
`(1 of 5 patients) dose levels. Median time to best response was 2
`months (range, 1-11 months), and median duration of response was
`6 months (range, 2-18 months).
`
`Pharmacokinetic analysis
`
`CC-5013 was rapidly absorbed, with maximum plasma concentrations
`at a median T max (time of maximum concentration) of I hour or 1.5
`hours on day 1 and day 28 in patients treated at each dose level. No trend
`in T max was noted with either increasing dose level or multiple <losings.
`Following Cmox (maximum concentration), plasma concentrations of
`CC-5013 declined in a predominantly monophasic manner, with elimi(cid:173)
`nation phase starting at I to 8 hours after dose on day I and day 28. The
`mean terminal elimination half-lives were 3.1 to 4.2 hours on both day I
`and day 28. There was little orno accumulation ofCC-5013: The mean
`accumulation ratio ofCC-5013 in plasma was 0.7 to 1.0, with a Cmax and
`area under the curve (AUC) (0-) of0.8 to 1.2 on day 28 compared with
`day 1. Intersubject variability was generally low to moderate for AUC
`and Cmax, with values ranging from 10.6% to 51.8% and 3% to 33% on
`day 1 and day 28, respectively.
`
`Bone marrow examination
`
`Of the 13 patients treated with CC-5013 at 50 mg/d, 12 patients and
`a single patient developed grades 3 and 4 neutropenia, respectively;
`BM hemopoiesis was nonnal or improved in IO and reduced in just
`2 of these patients. In the 5 patients treated with 50 mg/d CC-5013
`who developed grade 2 or 3 thrombocytopenia, megakaryocyte
`numbers were nonnal in 3 and mildly reduced in 2 cases. In
`aggregate, the BM findings were therefore most remarkable for the
`absence ofhypoplasia despite low circulating peripheral counts.
`
`Discussion
`
`Our long-tenn studies, both in vitro and in animal models, have
`shown the importance of host BM-MM cell interactions in
`promoting MM cell growth, survival, drug resistance, and migra(cid:173)
`tion in the BM microenvironment.3·t 2 Importantly, our studies have
`also shown that CC-5013 can target the MM cell in its BM milieu
`to overcome drug resistance in vitro as well as decrease tumor
`growth and extend survival in vivo in a murine model. 13-t 6 The
`present clinical study now demonstrates both the safety and
`efficacy of CC-5013 in patients with refractory and relapsed MM.
`In aggregate, these studies therefore provide the framework for
`development of a new treatment paradigm using CC-5013 to target
`both the MM cell and its microenvironment, overcome drug
`resistance, and improve patient outcome.
`The choice of dose levels in this phase 1 clinical trial (5-50
`mg/d) was targeted to achieve CC-5013 concentrations of approxi(cid:173)
`mately 25.9 to 259 ng/mL, levels that modulate the production of
`cytokines and inhibit MM cell proliferation in vitro. Importantly,
`except for an idiosyncratic reaction to CC-5013 that had also
`occurred to Thal, no DLT occurred within the first 28 days in any
`dose cohort; remarkably, no sedation, constipation, or neuropathy
`was seen. Twenty-five percent or greater reductions in paraprotein
`were observed in 17 (71 %) of 24 evaluable patients. This anti-MM
`activity was remarkable given that 15 patients (60%) had under(cid:173)
`gone prior SCT and 16 patients (64%) had progressive disease
`despite Thal treatment. In addition, less than 25% paraprotein
`reductions were observed in an additional 2 patients. These data,
`coupled with the absence of somnolence, constipation, and neurop(cid:173)
`athy, support future phase 2 studies of CC-5013 for MM patients
`earlier in the course of their disease.
`Pharmacokinetic studies completed in 24 subjects reveal rapid
`absorption (Tmax 1-1.5 hours), monophasic elimination (half-life
`[T 112 ] 3. 1-4.2 hours), and low to moderate intersubject variability
`for AUC (11 %-52%) and Cmax (3%-33%). In addition, there was no
`significant accumulation by day 28. The myelosuppression after
`day 28 seen in the patients treated with 50 mg/d CC-5013 is
`considered to be DLT. Furthermore, 13 of 15 patients treated at 25
`mg/d remained on therapy at this dose for a median of 4 months
`(range, 2-8 months); therefore, 25 mg/d is considered MTD. The
`
`Table 1. Maximal changes In paraproteln after CC-5013 treatment
`
`Decrease
`
`Dose, mg/d
`
`No. of patients
`
`<25%
`
`2: 25% < 50%
`
`2: 50% < 75%
`
`2: 75% < 99%
`
`5
`10
`25
`50
`Subtotal
`
`3
`5
`3
`13
`24
`
`0
`0
`
`2(8%)
`
`2
`0
`2
`6
`10 (42%)
`
`0
`2
`4(17%)
`
`0
`0
`0
`3
`3 (13%)
`
`Increase
`
`2: 25%
`
`0
`4
`0
`
`5(21%)
`
`DR. REDDY’S LABS., INC. EX. 1044 PAGE 4
`
`

`

`BLOOD, 1 NOVEMBER 2002 • VOLUME 100, NUMBER 9
`
`CC-5013 IN MYELOMA
`
`3067
`
`absence of significant marrow hypoplasia observed in most patients
`with cytopenias suggests etiologies other than BM suppression,
`which will be evaluated in future studies.
`Based upon the current study showing efficacy and tolerability of
`CC-5013, several future clinical phase 2 trials are planned in patients
`with refractory relapsed disease. First, because some patients treated
`with CC-5013 at 25 mgid developed granulocytopenia and thrombocy(cid:173)
`topenia, a dosing regimen that cycles drug exposure, with time for
`granulocyte and platelet recovery, will attempt to achieve clinical benefit
`while avoiding myelosuppression. Second, given the activity of the drug
`seen at lower dose levels and the T 112 of3 to 4 hours, a comparative trial
`of2 cycling regimens will be undertaken: 30 mgid as a single daily dose
`versus 30 mgid in divided doses. Third, because laboratory and clinical
`data have suggested at least additive effects of dexamethasone in
`
`combination with Thal, 13•19,20 and our preclinical data suggest that Dex
`also enhances anti-MM activity ofCC-5013, 13,21 clinical trials will also
`determine whether the addition of dexamethasone to CC-5013 enhances
`its clinical efficacy in MM.
`Thal has achieved response rates of 30% in patients with refractory,
`relapsed MM and, when combined with dexamethasone, response rates
`of 64% in patients with newly diagnosed disease. 2•20 However, somno(cid:173)
`lence, constipation, and neuropathy preclude its use in some patients.
`Because our study now shows that oral CC-5013 has antitumor activity,
`favorable pharmacokinetics, and acceptable toxicity in patients with
`relapsed and refractory MM, we will carry out clinical protocols of
`CC-5013, alone and in combination with dexamethasone, for the
`treatment of MM earlier in the disease course, including as initial
`treatment, therapy for first relapse, and maintenance therapy.
`
`References
`
`1. Greenlee RT, Hill-Harmon MB, Murray T, Thun M.
`Cancer Statistics, 2001. CA Cancer J Clin. 2001;
`51:15-36.
`2. Singha! S, Mehta J, Desikan R, et al. Anti-tumor
`activity of thalidomide in refractory multiple my-
`eloma. N Engl J Med. 1999;341 :1565-1571.
`3. Chauhan D, Uchiyama H, Akbarali Y, et al. Mui-
`tiple myeloma cell adhesion-induced interleukin-6
`expression in bone marrow stromal cells involves
`activation of NF-• B. Blood. 1996;87:1104-1112.
`4. Teoh G, Urashima M, Greenfield EA, et al. The 86
`kD subunit of Ku autoantigen mediates homotypic
`and heterotypic adhesion of multiple myeloma
`cells. J Clin Invest. 1997;101 :1379-1388.
`5. Tai YT, Teoh G, Chauhan D, et al. Ku86 variant
`expression and function in multiple myeloma cells
`is associated with increased sensitivity to DNA
`damage. J lmmunol. 2000; 165:634 7-6355.
`6. Ogata A, Chauhan D, Teoh G, et al. lnterleukin-6
`trtggers cell growth via the ras-dependent mite-
`gen-activated protein kinase cascade. J lmmunol.
`1997;159:2212-2221.
`7. Chauhan D, Pandey P, Hideshima T, et al. SHP2
`mediates the protective effect of interleukin-6
`against dexamethasone-induced apoptosis in
`multiple myeloma cells. J Biol Chem. 2000;275:
`27845-278450.
`8. Hideshima T, Nakamura N, Chauhan D, Ander-
`son K. Biologic sequelae of interleukin-6 induced
`
`P13-K/AKT signaling in multiple myeloma. Once-
`gene. 2001;20:5991-6000.
`9. Hideshima T, Chauhan D, Schlossman R, Rich-
`ardson P, Anderson KC. The role of tumor necro-
`sis factor a in the pathophysiology of human mul-
`tiple myeloma: therapeutic applications.
`Oncogene. 2001 ;20:4519-4527.
`10. Podar K, Tai YT, Davies FE, et al. Vascular endo-
`thelial growth factor triggers signaling cascades
`mediating multiple myeloma cell growth and mi-
`gration. Blood. 2001 ;98:428-435.
`11. Podar K, Tai YT, Lin B, et al. Vascular endothelial
`growth factor-induced migration of multiple my-
`eloma cells is associated with 13 1 integrin- and
`phosphatidylinositol 3-kinase-dependent PKC a
`activation. J Biol Chem. 2002;277:7875-7881.
`
`12. Urashima M, Chen BP, Chen S, et al. The devel-
`opment of a model for the homing of multiple my-
`eloma cells to human bone marrow. Blood. 1997;
`90:754-765.
`
`13. Hideshima T, Chauhan D, Shima Y, et al. Thalido-
`mide and its analogs overcome drug resistance
`of human multiple myeloma cells to conventional
`therapy. Blood. 2000;96:2943-2950.
`
`14. Gupta D, Treon SP, Shima Y, et al. Adherence of
`multiple myeloma cells to bone marrow stromal
`cells upregulates vascular endothelial growth fac-
`tor secretion: therapeutic applications. Leukemia.
`2001 ;15:1950-1961.
`
`15. Davies FE, Raja N, Hideshima T, et al. Thalido-
`mide and immunomodulatory derivatives aug-
`ment natural killer cell cytotoxicity in multiple my-
`eloma. Blood. 2001 ;98:210-216.
`16. Lentzsch S, LeBlanc R, Podar K, et al. Thalido-
`mide and its immunomodulatory analogs inhibit
`human multiple myeloma cell growth and angio-
`genesis in vivo. Leukemia. 2001. In press.
`17. Corral LG, Haslett PAJ, Muller GW, et al. Differen-
`tial cytokine modulation and T cell activation by
`two distinct classes of thalidomide analogues that
`are potent inhibitors ofTNF-a. J lmmunol. 1999;
`163:380-386.
`18. Muller G, Chen R, Huang SY, et al. Amino-substi-
`luted thaldidomide analogs: potent inhibitors of
`TNF-a production. Bioorg Med Chem Lett. 1999;
`9:1625-1630.
`19. Weber OM, Rankin K, Gavina M, Delasalle K, Al-
`exanian R. Thalidomide with dexamethasone for
`resistant multiple myeloma [abstract]. Blood.
`2000;96:167a.
`20. Rajkumar VJ, Hayman SR, Gertz MA, et al. Com-
`bination therapy with thalidomide plus dexameth-
`asone for newly diagnosed multiple myeloma [ab-
`stract]. Blood. 2001 ;98:849a.
`21. Mitsiades N, Mitsiades CS, Poulaki V, et al. Apo-
`ptotic signaling induced by immunomodulatory
`thalidomide analogs in human multiple myeloma
`cells: therapeutic implications. Blood. 2002;99:
`4525-4530.
`
`DR. REDDY’S LABS., INC. EX. 1044 PAGE 5
`
`

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