throbber
The Journal of Pain, Vol 15, No 12 (December), 2014: pp 1366-1376
`Available online at www.jpain.org and www.sciencedirect.com
`
`Lenalidomide for Complex Regional Pain Syndrome Type 1: Lack of
`Efficacy in a Phase II Randomized Study
`z
`Donald C. Manning,*,y
`{
`Guillermo Alexander,
`Alyse Cooper,
`Joseph C. Arezzo,
`jj
`Anne Louise Oaklander,**,yy
`zz
`R. Norman Harden,
`Srinivasa N. Raja,
`Richard Rauck,
`z
`and Robert Schwartzman
`*Department of Anesthesiology, University of Virginia Health System, Charlottesville, Virginia.
`y
`Adynxx Inc, San Francisco, California.
`z
`Department of Neurology, Drexel University, Philadelphia, Pennsylvania.
`x
`Department of Neuroscience and Neurology, Albert Einstein College of Medicine, Bronx, New York.
`{
`Celgene Corporation, Summit, New Jersey.
`jj
`Center for Pain Studies, Rehabilitation Institute of Chicago, Chicago, Illinois.
`yy
`**Departments of Neurology and
`Pathology (Neuropathology), Massachusetts General Hospital and Harvard
`Medical School, Boston, Massachusetts.
`zz
`Division of Pain Medicine, Johns Hopkins University, Baltimore, Maryland.
`xx
`Center for Clinical Research, Salem, North Carolina.
`
`xx
`
`x
`
`Abstract: Complex regional pain syndrome (CRPS) is a potentially debilitating chronic pain syndrome
`with a poorly understood but likely neuroimmune/multifactorial pathophysiology associated with
`axonal injury. Based on the potential contribution of proinflammatory cytokines to CRPS pathogenesis
`and prior research with thalidomide, we investigated lenalidomide, a thalidomide derivative, for CRPS
`treatment. We conducted a phase II, randomized, double-blind, placebo-controlled study to evaluate
`the efficacy of oral lenalidomide 10 mg once daily in consenting patients with unilateral or bilateral
`CRPS type 1. The study comprised 12 weeks of treatment followed by a long-term extension. The pri-
`mary efficacy outcome was reduced pain in the index limb, defined as $30% improvement from base-
`line using an 11-point numeric rating scale. One hundred eighty-four subjects enrolled. The primary
`endpoint was not met because equal proportions of treated (16.1%) and control (16.1%) subjects
`achieved the outcome; however, lenalidomide was well tolerated, with no evidence of neuropathy
`or major adverse effects. This study is the largest controlled, blinded clinical trial in subjects with chronic
`CRPS using the Budapest research criteria. It demonstrates the feasibility of conducting high-quality
`clinical trials in CRPS type 1 and provides considerations for designing future trials.
`Perspective: This article reports an adequately powered, controlled clinical trial in subjects with
`CRPS. Treatment and placebo were equally effective, but the study demonstrated that lenalidomide
`treatment is feasible in this population. The study provides examples to consider in designing future
`CRPS trials.
`ª 2014 by the American Pain Society
`
`Open access under CC BY-NC-ND license.
`
`Received February 19, 2014; Revised September 10, 2014; Accepted
`September 22, 2014.
`This study was sponsored by Celgene Corporation. Celgene developed
`the protocol in conjunction with the clinical investigators. Celgene was
`solely responsible for investigative site selection, data collection and stor-
`age methods and technology, clinical monitoring of protocol and Good
`Clinical Practice guidelines compliance, statistical analysis plan and
`execution, reporting of safety data to regulatory agencies, and the pro-
`vision of study drug and matching control. The authors received editorial
`support for manuscript preparation from Amy Zannikos, PharmD, of Pel-
`oton Advantage, LLC, funded by Celgene Corporation. The authors, how-
`ever, directed and are fully responsible for all content and editorial
`decisions for this manuscript. D.C.M. was an employee of Celgene Corpo-
`ration during the execution of the reported study, an owner of Celgene
`stock, and a former member of the Board of Directors for Reflex Sympa-
`thetic Dystrophy Syndrome Association-USA. G.A. received standard
`compensation for participating as an investigator in this study. J.C.A.
`received standard compensation for assisting with the design and anal-
`ysis of the electrophysiology data from this study. A.C. is an employee
`
`1366
`
`of Celgene Corporation. R.N.H. received standard compensation for
`participating as an investigator in this study and also had an unrestricted
`grant from Celgene at the time of this study. He also has received research
`support from the Reflex Sympathetic Dystrophy Syndrome Association.
`A.L.O. reports no conflicts of interest. S.N.R. received standard compen-
`sation for participating as an investigator in this study and reports a
`research grant from Medtronic Inc for studies on the mechanisms of spi-
`nal cord stimulation–induced analgesia. R.R. received standard compen-
`sation for participating as an investigator in this study. R.S. received
`standard compensation for participating as an investigator in this study.
`Trial registration: clinicaltrials.gov #NCT00109772.
`Address reprint requests to Donald C. Manning, MD, PhD, 16 Johnston
`Drive, Bloomsbury, NJ 08804. E-mail: donaldcmanning@gmail.com
`1526-5900/
`ª 2014 by the American Pain Society
`Open access under CC BY-NC-ND license.
`http://dx.doi.org/10.1016/j.jpain.2014.09.013
`
`IPR2018-00685
`Celgene Ex. 2016, Page 1
`
`

`

`Manning et al
`
`The Journal of Pain
`
`1367
`
`Key words: Complex regional pain syndrome, cytokines, immunomodulation, lenalidomide, thalido-
`mide, clinical trial.
`
`Complex regional pain syndrome (CRPS) is an often
`
`debilitating neuropathic/neuroimmune syndrome
`usually triggered by injury to a limb.7,13,34,42 Its
`clinical features can include pain symptoms such as
`allodynia and hyperalgesia; difficulty moving, including
`reduced range of motion; changes
`in limb color,
`temperature, appearance, and sweating; edema; and
`bone changes.15,17,25,38,40 CRPS affects more females
`than males (relative risk 2:1–4:1) and has an average
`age of onset of 37 to 60 years.3,8,33 Its pathophysiology
`is likely multifactorial,13 and far more patients have
`type 1, which often involves soft-tissue and small-fiber
`axonal injury,24 than type 2, which requires injury to a
`major nerve.15,22,24 Treatment guidelines based on
`limited available data and expert opinion recommend
`physical and occupational therapy, analgesic and other
`medications,
`interventional
`procedures,
`and
`therapy and support.15,27 Proposed
`psychological
`mechanisms include peripheral afferent and efferent as
`well as central neural involvement; clinical presentation
`suggests a neurogenic inflammation and/or activation
`of the immune system leading to an exaggerated
`inflammatory process. Regardless, there is increasing
`consensus that inflammation plays an important role in
`CRPS type 1.
`Increased levels of proinflammatory cytokines (tumor
`necrosis factor-a [TNF-a], interleukin [IL]-1b, IL-2, and
`IL-6) and anti-inflammatory cytokines (IL-4 and IL-10)
`have been identified in plasma and cerebrospinal fluid
`and within the affected limbs of CRPS patients compared
`with nonaffected controls.1,20,26,39 Increased levels of
`TNF-a and IL-6 have been identified in blister fluid from
`the skin of the affected limb when compared with the
`unaffected limb.16 The elevated levels of proinflamma-
`tory and anti-inflammatory cytokines found in CRPS sug-
`gest that immunomodulating agents might be useful
`because of their anti-inflammatory effects. Few well-
`controlled and adequately powered clinical treatment
`studies of anti-inflammatory agents have been per-
`formed, and existing study designs vary.9,14 Small pilot
`studies have also suggested possible therapeutic
`avenues, including a report of observable but variable
`resolution of CRPS in 2 patients treated with the TNF-a
`antagonist infliximab.21 Thalidomide is a relatively selec-
`tive inhibitor of TNF-a production by human monocytes,
`both in vitro and in vivo, and it exerts costimulatory
`effects on T-cell responses, including inhibiting proin-
`flammatory
`cytokines
`and
`stimulating
`anti-
`inflammatory cytokines.6 Following demonstration of
`near resolution of CRPS symptoms after thalidomide
`treatment in a woman with CRPS for 3 years,28 thalido-
`mide was studied in 42 patients with long-standing, pre-
`viously
`treated CRPS. Objective
`and subjective
`improvement was observed in 17% of patients, with
`14% experiencing at least modest pain relief.36 However,
`use of thalidomide is limited by its well-known neurotox-
`icity and teratogenicity. Teratogenicity is a particular
`concern in CRPS, because most patients are female.
`
`Lenalidomide is a thalidomide derivative created in a
`program to enhance the anti-inflammatory and immu-
`nomodulatory properties of thalidomide while reducing
`its toxicity. It is approved in the United States and Europe
`for treating multiple myeloma and anemia in some
`patients with myelodysplastic syndromes12,30 and in
`several other countries (eg, China, Japan) for treating
`multiple myeloma.4,37 Lenalidomide has approximately
`1,000 times more anti-inflammatory activity in vitro
`than thalidomide.6 Its pharmacologic activities include
`inhibiting secretion of the proinflammatory cytokines
`TNF-a, IL-1b, and IL-6, and increasing secretion of the
`anti-inflammatory cytokine IL-10 from stimulated mono-
`cytes.6,19 Lenalidomide’s safety and effect on reported
`pain in CRPS type 1 were studied in a preliminary phase
`IIa, multicenter, single-arm, open-label, 40-patient pilot
`study initiated in 2003.35 The good response rates ($2-
`point
`improvement
`from baseline on an 11-point
`numeric rating scale [NRS]) were 28.9% at week 12 and
`52.0% at week 52, and patient-rated pain and sleep
`scores also improved (P < .05).35 In addition, lenalido-
`mide was well tolerated, with few treatment-related
`serious adverse events (AEs) and no evidence of neuro-
`logic toxicity. These findings provided the rationale for
`the current phase IIb, multicenter, double-blind, pla-
`cebo-controlled study.
`
`Methods
`
`Subjects
`The inclusion criteria were age $18 years and CRPS
`type 1 (as per Budapest research criteria17) for $1 year
`with unilateral or bilateral involvement of a distal hand
`or foot, with or without proximal spread, plus a CRPS-
`related pain intensity score of $4 in the index limb. In
`cases where both limbs were affected, subjects selected
`their limb to be assessed (designated as the index
`limb). For subjects with single-limb involvement, that
`limb was the index limb. Because lenalidomide is struc-
`turally similar to thalidomide, a human teratogen, men
`and women with reproductive potential had to follow
`pregnancy prevention requirements,
`including coun-
`seling about preventing pregnancy. Subjects had to
`agree to use reliable forms of contraception, and women
`had to agree to periodic pregnancy testing. Potentially
`fertile women had to submit 2 negative pregnancy tests
`before randomization.
`Subjects were excluded for history of stroke or deep
`vein thrombosis in the past 5 years (based on experi-
`ence with lenalidomide in the oncology program);
`peripheral neuropathy; severe, progressive, or uncon-
`trolled renal, hepatic, hematologic, endocrine, pulmo-
`nary, cardiac, neurologic, or cerebral disease; any
`serious medical diagnoses or abnormal laboratory test
`results; or psychiatric
`illness
`that
`could prevent
`informed consent. Additional exclusions were prior
`treatment with lenalidomide or allergy to thalidomide;
`
`IPR2018-00685
`Celgene Ex. 2016, Page 2
`
`

`

`1368
`
`The Journal of Pain
`
`Lenalidomide in Complex Regional Pain Syndrome
`
`pregnancy or lactation; or use of concomitant medica-
`tions that increase the risk of deep vein thrombosis.
`Low-dose aspirin (81 mg) was required for subjects
`receiving sex hormones in any form for contraception
`or treatment of menopausal symptoms.
`
`Study Design
`This multicenter, phase IIb study was conducted from
`February 2005 to April 2008 at 24 study sites in the United
`States.
`It included a 2-week screening phase and a
`12-week double-blind treatment phase, followed by an
`optional extension phase that allowed treatment to
`continue as long as benefit was maintained (Fig 1). All
`subjects meeting CRPS-related morning and evening
`pain intensity numerical rating scale (PI-NRS) score re-
`quirements averaged over the 7-day period before
`randomization ($8 recorded scores, average score $4
`in affected limb) were randomized 1:1 to receive lenali-
`domide 10 mg orally once daily or placebo for up to
`12 weeks in the treatment phase. Concomitant therapy
`for CRPS pain (opioid and nonopioid analgesics, nonste-
`roidal anti-inflammatory drugs, anticonvulsants, antide-
`pressants, and other nondrug therapies) was permitted if
`the dose/regimen had been stable for $4 weeks before
`randomization and remained stable throughout the
`study. No new CRPS medications or nondrug therapies
`were allowed except for limited use of a rescue medica-
`tion (short-acting opioid for no more than 7 days) to
`treat pain flare, trauma, or invasive procedure. During
`the extension phase, subjects were permitted to initiate,
`reduce, increase, or withdraw from concomitant CRPS
`pain medications/nondrug therapies except for initiating
`experimental therapies.
`Randomization codes were generated by a Celgene
`statistician not involved in the study using a randomiza-
`tion program in the Statistical Analysis Software system.
`Randomization was performed centrally across all cen-
`ters using blocked randomization with a block size of
`4. Eligible patients were randomized in a 1:1 ratio to 1
`of the 2 treatment groups. Sites were provided with
`drug for 4 subjects at a time, and additional shipments
`were sent as required. Each qualifying subject was as-
`signed the next sequential number in the series of
`numbers assigned to the site. Code-break envelopes
`were provided in the boxes containing the drug, and
`site personnel were instructed to contact the Celgene
`Medical Monitor before breaking the code. The Celgene
`Clinical Supplies group worked with Xeremis, which
`packaged the drugs. All subjects completing the entire
`12-week double-blind phase were eligible for the exten-
`sion phase. Subjects first randomized to placebo were al-
`lowed to cross over to lenalidomide 10 mg/d and
`continue treatment in the extension phase.
`The study protocol, amendments, and informed con-
`sent form were approved by the institutional review
`board at each site, and the study was conducted in accor-
`dance with the International Conference on Harmonisa-
`tion Guidelines for Good Clinical Practice and the
`Declaration of Helsinki. All subjects provided written
`informed consent before initiating study procedures.
`
`Figure 1. Study design.
`
`Dose Selection
`The lenalidomide dose for continuous dosing in
`myelodysplastic syndrome starts at 10 mg/d and can be
`adjusted downward for renal function. For intermittent
`dosing, such as for multiple myeloma, the dose can start
`at 25 mg/d for a defined portion of the chemothera-
`peutic cycle. AEs in general were dose related. Prelimi-
`nary data from the open-label study in CRPS showed
`lenalidomide to be well tolerated with an expected AE
`profile. Subjects who responded to lenalidomide
`10 mg/d reported return of their CRPS if their dose was
`reduced to 5 mg/d because of AEs, and most chose to
`resume 10 mg/d to complete the study. On the basis of the
`requirement for continuous dosing as well as experience
`in other patient populations, lenalidomide 10 mg/d was
`selected as the study dose.
`
`Efficacy and Safety Assessments and
`Endpoints
`Subjects used an electronic diary to collect daily
`morning and evening efficacy assessment data,
`including the PI-NRS. The PI-NRS was presented twice
`per day at set consistent times (morning and evening)
`as a discrete 11-box Likert scale from 0 (no pain) to
`10 (worst pain imaginable) with the instructions
`‘‘Please rate your pain due to CRPS right now.’’ The pri-
`mary efficacy endpoint was the PI-NRS responder rate,
`with response defined as completion of the treatment
`phase with a $30% reduction from baseline (ie,
`improvement) in the 7-day averaged morning and eve-
`ning PI-NRS score for the week before the week 12
`assessment as compared with the 7-day averaged
`morning and evening PI-NRS scores during the baseline
`period. The secondary efficacy endpoints included
`change from baseline in the Short-Form McGill Pain
`Questionnaire total score and sensory and affective
`subscale scores, pain intensity ratings (combined morn-
`ing and evening assessments) using PI-NRS in the index
`limb, sleep rating (NRS), activity level rating (NRS), sub-
`ject’s assessment of CRPS signs and symptoms in the in-
`dex limb, allodynia NRS score, allodynia rating
`between the index limb and the normal limb at the
`end of week 12 (unilateral CRPS subjects only), and
`concomitant use of pain medications.
`Safety assessments included type, frequency, and
`severity of AEs; physical examination; vital signs; and
`laboratory test results. A battery of noninvasive nerve
`conduction studies were included as a safety measure
`to explore the possibility of
`lenalidomide-induced
`
`IPR2018-00685
`Celgene Ex. 2016, Page 3
`
`

`

`Manning et al
`
`peripheral neuropathy, and to evaluate whether the
`anti-inflammatory
`effects of
`lenalidomide
`could
`improve nerve conduction. The principal electrophysio-
`logic outcome was maximal nerve conduction velocity.
`Responses were recorded from sural sensory, median
`sensory, peroneal motor, and median motor nerves on
`2 occasions before the onset of dosing and 2 occasions
`at the end of the initial 12-week dosing period.
`
`Statistical Analysis
`When the responder definition of this study was
`applied to the data from the pilot study,35 the response
`rate was calculated to be 34%. Based on this, plus the
`19% response rate in patients with postherpetic neural-
`gia who received placebo in a published study,32
`response rates for the current study were hypothesized
`to be 15% for placebo and 35% for lenalidomide. Using
`these assumptions, a sample size of 83 subjects per group
`would have 80% power to detect a significant difference
`in PI-NRS score between groups using a 2-sided,
`continuity-corrected c2 test at the .05 significance level.
`The primary analysis used the modified intent-to-treat
`population (subjects who received at least 1 dose of study
`medication and had at least 1 postdose measurement).
`Differences in responder rates between groups were
`assessed using the Cochran-Mantel-Haenszel test. Sub-
`jects who discontinued the treatment phase prematurely,
`regardless of reason, were considered nonresponders. All
`comparisons used 2-tailed tests at the .05 level of signifi-
`cance. Summary statistics were provided for continuous
`outcome measures. Secondary endpoints were evaluated
`with an analysis of covariance model, which included
`terms of treatment, center, baseline score, concomitant
`CRPS medication change status, and treatment-by-
`baseline score. Analysis of covariance results presented
`here are for the observed cases subpopulation of the
`modified intent-to-treat population with no imputation
`for missing data, thus giving varying sample sizes,
`because not all data were available for all subjects.
`
`Results
`Among the 184 subjects randomized, 180 received $1
`dose of study medication (97.8%; the safety population)
`and had $1 postdose diary measurement (the modified
`intent-to-treat population). The 12-week, double-blind
`treatment phase was completed by 68 subjects who
`received lenalidomide (78.2%) and 79 who received
`placebo (84.9%; Fig 2). The primary reason for discontin-
`uation during this phase was AEs. These affected 14 sub-
`jects who received lenalidomide (16.1%) and 5 subjects
`who received placebo (5.4%). The mean duration of
`treatment in the double-blind treatment phase was
`similar between groups: 10.6 weeks with lenalidomide
`and 11.4 weeks with placebo. During the 12-week
`double-blind treatment phase, small numbers of subjects
`receiving lenalidomide (11; 12.6%) and receiving
`placebo (7; 7.5%) altered their concomitant CRPS medi-
`cation (including addition, discontinuation, or dosage
`change).
`
`The Journal of Pain
`
`1369
`
`One hundred forty-two subjects entered the extension
`phase (lenalidomide, n = 64; placebo, n = 78). Ninety-four
`subjects discontinued—among them, 45 receiving only
`lenalidomide (70.3%) and 49 receiving lenalidomide
`(62.8%) after crossing over from placebo. The primary
`reason for discontinuation among subjects receiving
`lenalidomide was lack of therapeutic effect (29.7%).
`Among placebo subjects who crossed over to lenalido-
`mide, it was AEs (24.4%). The mean duration of treat-
`ment in the extension phase was 43.8 weeks for
`subjects receiving lenalidomide and 46.2 weeks for pla-
`cebo subjects who crossed over to lenalidomide. The
`cumulative durations of treatment were 41.1 weeks
`and 49.9 weeks, respectively. The baseline demographics
`and characteristics of subjects are summarized in Table 1.
`The study was discontinued prematurely during the
`extension phase because unblinding of the data from
`the double-blind treatment phase revealed that the pri-
`mary efficacy endpoint had not been met. At week 12,
`16.1% of subjects receiving lenalidomide and 16.1%
`receiving placebo were considered responders based
`on changes in PI-NRS scores. Changes (morning assess-
`ments, evening assessments, and combined morning
`and evening assessments) from baseline to week 12 (7-
`day averaged scores) did not differ between groups
`(Table 2). In addition, there were no significant differ-
`ences between groups in change from baseline in the
`daily sleep assessment, Short-Form McGill Pain Question-
`naire, activity level rating, and allodynia NRS score at
`week 12 (Table 3; no adjustment for multiplicity was
`made, so P values are for reference only).
`To better understand the types of subjects in the
`responder and nonresponder categories, use of concom-
`itant drugs (Table 4), nondrug therapies (Table 5), and
`rescue medications (Table 6) was analyzed in relation to
`responder status for the lenalidomide and placebo treat-
`ment groups. There were no significant differences in
`these outcomes between the lenalidomide and placebo
`groups, with responders using only slightly fewer
`concomitant treatments, but this did not determine
`responder status. The vast majority of subjects either
`took no rescue medication or used the described
`opioid-rescue protocol. More subjects in the placebo
`group (n = 15) than lenalidomide-treated subjects
`(n = 3) used nonallowed rescue therapies.
`Given lenalidomide’s mechanism of action, we evalu-
`ated whether subjects with higher baseline scores for
`potential markers of inflammation or microvasculopathy
`(changes in skin temperature, color, or swelling) on the
`subject’s assessment of CRPS symptoms (Table 7) were
`more likely to respond. The placebo group had slightly
`lower assessment ratings for these symptom categories
`compared to the lenalidomide-treated group, but there
`were no appreciable differences between responders
`and nonresponders.
`
`Safety Outcomes
`During the double-blind treatment phase, at least 1 AE
`was reported by 81 subjects receiving lenalidomide
`(93.1%) and 76 subjects receiving placebo (81.7%).
`
`IPR2018-00685
`Celgene Ex. 2016, Page 4
`
`

`

`1370
`
`The Journal of Pain
`
`Lenalidomide in Complex Regional Pain Syndrome
`
`Figure 2. Subject disposition. *Defined as all subjects who took at least 1 dose of study drug.
`
`Cumulative data for the double-blind plus open-label
`treatment phases indicated that at least 1 AE was
`reported by 83 subjects receiving lenalidomide (95.4%)
`and 85 placebo subjects who crossed over to lenalido-
`mide (91.4%). The most common AEs are summarized
`in Table 8. In the double-blind phase, at least 1 serious
`AE was reported by 4 subjects receiving lenalidomide
`(4.6%)
`(ie, decreased potassium, pain,
`joint sprain,
`angioneurotic edema) and 6 receiving placebo (6.5%)
`(ie, increased blood pressure, neuropathic pain, vomit-
`ing, tremor, clostridium colitis, and respiratory failure);
`cumulative data indicated that $1 serious AE was
`reported in 12 subjects receiving lenalidomide (13.8%).
`This included, in addition to the above, joint sprain
`(n = 2), foot fracture, deep vein thrombosis, positive hu-
`man chorionic gonadotropin, false-positive pregnancy
`test, sick sinus syndrome, pregnancy (n = 2), neuropathic
`pain, spontaneous abortion, and arthritis. Sixteen pla-
`cebo subjects who crossed over
`to lenalidomide
`(17.2%) also reported accidental overdose, hip fracture,
`
`deep vein thrombosis (n = 2), bradycardia, thyroiditis, ad-
`renal insufficiency, cholecystitis, gallbladder obstruction,
`allergic alveolitis, hemolytic anemia, thrombotic throm-
`bocytopenic purpura, upper abdominal pain, vomiting,
`or endometriosis. Treatment-related AEs led to discon-
`tinuation in 18 subjects receiving lenalidomide (20.7%)
`and 5 receiving placebo (5.4%); cumulative data showed
`that treatment-related AEs led to discontinuation in 29
`subjects receiving lenalidomide (33.3%) and 29 placebo
`subjects who crossed over to lenalidomide (31.2%).
`Most AEs leading to discontinuation were grade 1
`(mild) or 2 (moderate). AEs led to dose reduction in 11
`subjects receiving lenalidomide (12.6%) and 2 receiving
`placebo (2.2%) during the double-blind phase. The
`most common AE leading to reduction of lenalidomide
`was rash (3.4% of subjects), and the 2 AEs in the placebo
`group were clostridium colitis and conjunctivitis. Few
`shifts in laboratory values from baseline were noted in
`the double-blind treatment phase and the extension
`phase; of those that were different, few were reported
`
`IPR2018-00685
`Celgene Ex. 2016, Page 5
`
`

`

`The Journal of Pain
`
`1371
`
`.27
`.28
`.26
`
`PVALUEy
`
`yNoadjustmentformultiplicitywasmade,becausethePvalueisforreferenceonly;thePvalueisfromtheanalysisofcovariancemodeladjustingforcenterandbaselinescore.
`*AdjustedmeanchangefrombaselineinPainIntensityRatings(observedcases)atweek12.Noimputationwasemployedforthesedata.Negativechangeindicatesimprovement.
`Abbreviations:M,mean;SD,standarddeviation.
`
`ADJUSTEDCHANGE*
`
`.51
`.36
`.43
`
`(n=87).4(1.5)
`(n=88).3(1.5)
`(n=88).4(1.5)
`
`CHANGE
`
`(n=87)6.7(2.3)
`(n=88)6.5(2.3)
`(n=88)6.6(2.3)
`
`(n=87)7.1(1.6)
`(n=88)6.9(1.7)
`(n=88)7.0(1.6)
`
`WEEK12
`
`BASELINE
`
`ADJUSTEDCHANGE*
`
`.78
`.63
`.71
`
`(n=80).7(1.7)
`(n=80).6(1.7)
`(n=80).7(1.7)
`
`CHANGE
`
`(n=80)6.6(2.1)
`(n=80)6.3(2.1)
`(n=80)6.5(2.1)
`
`(n=80)7.3(1.4)
`(n=80)6.9(1.5)
`(n=80)7.1(1.4)
`
`PM,M(SD)
`AM,M(SD)
`AM1PM,M(SD)
`
`WEEK12
`
`BASELINE
`
`PI-NRS
`
`PLACEBO
`
`LENALIDOMIDE
`
`Table2.CRPSPI-NRSSummaryofChangeFromBaselineatWeek12inObservedCases
`
`Manning et al
`Table 1. Subject Demographic and Baseline
`Characteristics
`
`CHARACTERISTIC
`
`Age, y, M (SD)
`Age distribution, n (%)
`#65 y
`>65 y
`Sex, n (%)
`Male
`Female
`Race/ethnicity, n (%)
`White
`Black
`Hispanic
`Asian/Pacific Islander
`Baseline CRPS PI-NRS
`score, M (SD)*
`Daily sleep assessment
`average score, M (SD)
`
`LENALIDOMIDE
`(N = 87)
`
`PLACEBO
`(N = 93)
`
`TOTAL
`(N = 180)
`
`43.9 (11.4)
`
`45.1 (11.1)
`
`44.5 (11.2)
`
`83 (95)
`4 (5)
`
`22 (25)
`65 (75)
`
`81 (93)
`3 (3)
`1 (1)
`2 (2)
`7.2 (1.4)
`(n = 85)
`5.4 (1.1)
`(n = 85)
`
`89 (96)
`4 (4)
`
`14 (15)
`79 (85)
`
`86 (93)
`4 (4)
`3 (3)
`0 (0)
`7.0 (1.7)
`(n = 92)
`5.5 (1.3)
`(n = 92)
`
`172 (96)
`8 (4)
`
`36 (20)
`144 (80)
`
`167 (93)
`7 (4)
`4 (2)
`2 (1)
`–
`
`–
`
`Abbreviations: M, mean; SD, standard deviation.
`*Combined morning and evening assessments, based on a 0–10 scale, with
`higher ratings indicating more severe pain.
`
`as AEs. Likewise, no notable shifts in thyroid function or
`vital sign parameters (diastolic and systolic blood pres-
`sure, pulse, temperature) were reported.
`The pattern and variance of the electrophysiologic
`measures in the current study were consistent with previ-
`ous reports for multicenter clinical trials of pain.2 There
`were no significant differences in mean changes from
`baseline to study termination for the 2 sensory or the 2
`motor nerve velocities. Lenalidomide, at the doses and
`for the duration tested, was not associated with slowing
`of velocity, reduction in amplitude, or exacerbation of
`peripheral neuropathy.
`
`Discussion
`This phase IIb study evaluated the efficacy and safety of
`oral lenalidomide 10 mg once daily in subjects with CRPS
`type 1 based on pilot data indicating that lenalidomide
`had acceptable safety and some evidence of efficacy in
`CRPS.35 Although the current outcomes show no differ-
`ences in efficacy between lenalidomide and placebo for
`the primary and secondary endpoints, subjects tolerated
`daily lenalidomide for up to 2 years without significant
`safety issues, demonstrating that lenalidomide can be
`well tolerated in a population other than cancer patients.
`Importantly, all AEs reversed after lenalidomide was dis-
`continued, and neurophysiologic surveillance revealed
`no evidence of treatment-induced peripheral neuropa-
`thy, in distinction to the neuropathy associated with
`thalidomide use.28
`Both the pilot study and the phase IIb study helped
`identify design elements to consider for future CRPS tri-
`als. For efficiency purposes, we reevaluated the assess-
`ment tools and general tolerability measures used in
`the open-label pilot study, acknowledging that the po-
`tential for false positives was high, but the small sample
`ensured that fewer subjects would be exposed to the
`
`IPR2018-00685
`Celgene Ex. 2016, Page 6
`
`

`

`1372
`
`The Journal of Pain
`
`Table 3. Summary of Selected Secondary
`Efficacy Assessments
`
`Lenalidomide in Complex Regional Pain Syndrome
`Table 5. Concomitant Nondrug Therapy
`(Double-Blind Phase)
`
`MEAN ADJUSTED CHANGE
`FROM BASELINE AT WEEK 12
`
`Daily sleep assessment
`
`SF-MPQ
`Total
`Sensory
`Affective
`Activity level rating (NRS)
`
`Allodynia NRS score
`
`LENALIDOMIDE
`
`PLACEBO
`
`P VALUE*
`
`(n = 85)
`.11
`(n = 85)
`6.36
`4.55
`1.77
`(n = 85)
`.17
`(n = 76)
`3.65
`
`(n = 92)
`.24
`(n = 92)
`3.94
`2.85
`1.15
`(n = 85)
`.19
`(n = 87)
`4.37
`
`.35
`
`.09
`.12
`.16
`.90
`
`.16
`
`LENALIDOMIDE, N
`
`PLACEBO, N
`
`TOTAL
`(N = 87)
`
`RESPONDERS
`(N = 14)
`
`TOTAL
`(N = 93)
`
`RESPONDERS
`(N = 15)
`
`72
`3
`5
`1
`2
`10
`0
`1
`
`0
`0
`1
`0
`0
`4
`0
`1
`
`82
`2
`1
`3
`0
`7
`2
`0
`
`0
`0
`
`2
`0
`1
`0
`0
`
`NONDRUG THERAPY
`
`None
`TENS
`Acupuncture
`Spinal cord stimulation
`Psychological treatment
`Physical therapy
`Chiropractic therapy
`Stellate ganglia blocks
`
`Abbreviation: SF-MPQ, Short-Form McGill Pain Questionnaire.
`*No adjustment for multiplicity was made, because the P value is for reference
`only; the P value is from the analysis of covariance model adjusting for center
`and baseline score.
`
`Abbreviation: TENS, transcutaneous electrical nerve stimulation.
`NOTE. The total will not equal the sample size as some subjects received >1
`concomitant drug therapy.
`
`experimental agent in this initial study. The open-label
`pilot study accurately predicted safety, but not efficacy,
`in the current, adequately powered, controlled study.
`The lenalidomide 10-mg/d dose was well tolerated in
`this study; however, some AEs were observed. Given
`
`Table 4. Concomitant Medication Use During
`the Study Treatment Phase
`
`TREATMENT
`GROUP AND
`OUTCOME
`STATUS
`
`CONCOMITANT MEDICATIONS
`
`TOTM AD MR OP LA AC NSAID BZ OTHER
`
`86
`2.9
`
`18
`.2
`
`17
`.2
`
`66
`.8
`
`10
`.7
`
`11
`.1
`
`2
`.1
`
`39
`.5
`
`9
`.6
`
`16
`.2
`
`3
`.2
`
`7
`.1
`
`0
`.0
`
`21
`.2
`
`2
`.1
`
`the lack of clear efficacy and the dose dependency of
`AEs with moderate to long-term administration of lena-
`lidomide, higher doses approaching 25 mg are not
`advised. The quantitative sensory testing and measure-
`ments of limb volume used in the pilot study, initially
`thought
`important
`to include, proved difficult
`to
`execute and were inconsistently applied in the pilot,
`and thus they were not employed in this study, which
`focused on pain endpoints. Streamlining the battery of
`assessments was judged to be critical for larger studies.
`In future studies of CRPS, the need for objective assess-
`ments in pain clinical research must be balanced by feasi-
`bility considerations.17
`The current study is the largest controlled study of
`people with CRPS and one of the first to employ the
`Budapest research criteria for defining CRPS. These pro-
`vide a useful tool for carefully selecting subjects, but
`this symptom-based definition does not address poten-
`tial heterogeneity in etiology or disease mechanisms,
`which may have affected study results. This study was
`
`Lenalidomide
`Total
`n
`M
`Responder
`14
`n
`2.6
`M
`Nonresponder
`n
`72
`M
`3.0
`Placebo
`Total
`n
`M
`Responder
`15
`n
`2.3
`M
`Nonresponder
`n
`75
`M
`2.7
`
`90
`2.7
`
`3
`.2
`
`15
`.2
`
`18
`.2
`
`2
`.1
`
`16
`.2
`
`2
`.1
`
`15
`.2
`
`24
`.2
`
`5
`.3
`
`19
`.1
`
`56
`.8
`
`9
`.1
`
`30
`.4
`
`69
`.8
`
`9
`.6
`
`60
`.8
`
`10
`.1
`
`2
`.1
`
`8
`.1
`
`43
`.5
`
`7
`.5
`
`36
`.5
`
`13
`.2
`
`17
`.2
`
`4
`.3
`
`13
`.2
`
`7
`.1
`
`7
`.1
`
`0
`.0
`
`7
`.1
`
`19
`.3
`
`15
`.2
`
`2
`.1
`
`13
`.2
`
`Abbreviations: TotM, total medications used, including multiple agents within a
`class; AD, antidepressants, including tricyclic agents; MR, muscle relaxants and
`baclofen; OP, opioids; LA, local anesthetic preparations; AC, anticonvulsants,
`including gabapentin and pregabalin; NSAID, nonsteroidal anti-inflammatory
`agents; BZ, benzodiazepines, including clonazepam; Other, alpha adrenergic
`agents, mexiletine, and acetaminophen; M, mean.
`NOTE. The n value reflects the incidence of a parti

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