throbber
Articles
`
`TNF neutralization in MS
`Results of a randomized, placebo-controlled
`multicenter study
`
`The Lenercept Multiple Sclerosis Study Group and The University of British Columbia
`MS/MRI Analysis Group*
`
`Article abstract—Objective: A double-blind, placebo-controlled phase II study was conducted in 168 patients, most with
`relapsing-remitting MS, to evaluate whether lenercept would reduce new lesions on MRI. Background: Tumor necrosis
`factor (TNF) has been implicated in MS pathogenesis, has been identified in active MS lesions, is toxic to oligodendrocytes
`in vitro, and worsens the severity of experimental allergic encephalomyelitis (EAE) in animals. Lenercept, a recombinant
`TNF receptor p55 immunoglobulin fusion protein (sTNFR-IgG p55), protects against EAE. Methods: Patients received 10,
`50, or 100 mg of lenercept or placebo IV every 4 weeks for up to 48 weeks. MRI scans and clinical evaluations were
`performed at screening, at baseline, and then every 4 weeks (immediately before dosing) through study week 24. Results:
`There were no significant differences between groups on any MRI study measure, but the number of lenercept-treated
`patients experiencing exacerbations was significantly increased compared with patients receiving placebo (p 5 0.007) and
`their exacerbations occurred earlier (p 5 0.006). Neurologic deficits tended to be more severe in the lenercept treatment
`groups, although this did not affect Expanded Disability Status Scale scores. Anti-lenercept antibodies were present in a
`substantial number of treated patients; serum lenercept trough concentrations were detectable in only a third. Adverse
`events that increased in frequency in treated patients included headache, nausea, abdominal pain, and hot flushes.
`Conclusions: Lenercept failed to be beneficial, but insight into the role of TNF in MS exacerbations was gained.
`NEUROLOGY 1999;53:457–465
`
`MS is believed to be an inflammatory autoimmune
`disorder of the CNS with unknown myelin compo-
`nents as target. A number of findings have suggested
`that tumor necrosis factor (TNF) contributes to prop-
`agating the inflammatory response and to tissue in-
`jury in MS. In autopsy specimens, TNF has been
`demonstrated within active MS foci.1 TNF has been
`shown to have a direct toxic effect against oligoden-
`drocytes and a proliferation-inducing effect on astro-
`cytes in in vitro studies.2,3 In patients with MS,
`elevated TNF levels in the serum and CSF have been
`correlated in some studies with disease progres-
`sion.4,5 Blood mononuclear cells from MS patients,
`studied just before an exacerbation, secrete greater
`amounts of TNF in response to mitogen stimulation
`than at other times.6 Blood mononuclear cells from
`MS patients with active disease express higher lev-
`els of TNF mRNA than do cells from MS patients
`with stable disease or healthy controls.7,8
`
`Studies of experimental autoimmune encephalo-
`myelitis (EAE) have profoundly shaped views of MS
`pathogenesis. EAE is an autoimmune disease with
`pathologic features reminiscent of those seen in MS.
`TNF treatment worsens EAE,9 and TNF neutraliza-
`tion by anti-TNF antibody treatment consistently
`protects animals from EAE.10-12 Similarly, TNF cap-
`ture by lenercept, a TNFa receptor–immunoglobulin
`G (IgG)1 fusion protein, protects in EAE.13 The above
`indicates that TNF functions in EAE as a proinflam-
`matory mediator and suggests that TNF depletion
`might be protective in MS. The hypothesis that neu-
`tralization of TNF may reduce or halt MS progres-
`sion was evaluated in a phase II randomized,
`multicenter, placebo-controlled study of three doses
`of lenercept (sTNFR-IgG p55). Lenercept is a dimeric
`recombinant protein molecule built from two copies
`of the 55 kDa TNF receptor extracellular domain
`fused to a fragment of the human immunoglobulin
`
`See also pages 444 and 466
`
`*See the Appendix on page 464 for a listing of members of The Lenercept Multiple Sclerosis Study Group and The University of British Columbia MS/MRI
`Analysis Group.
`Funded by F. Hoffmann-LaRoche Ltd., Basel, Switzerland.
`Received September 11, 1998. Accepted in final form April 8, 1999.
`Address correspondence and reprint requests to Dr. Barry G.W. Arnason, University of Chicago, Department of Neurology, 5841 S. Maryland Ave., MC 2030,
`Chicago, IL 60637; e-mail: barnason@drugs.bsd.uchicago.edu
`
`Copyright © 1999 by the American Academy of Neurology 457
`
`Biogen Exhibit 2087
`Mylan v. Biogen
`IPR 2018-01403
`
`Page 1 of 9
`
`

`

`IgG1 heavy chain.14,15 In accordance with recently
`published recommendations, efficacy was assessed
`by means of MRI.16
`
`Methods. Patients. A total of 168 patients with clini-
`cally definite or laboratory supported definite MS were
`enrolled in a double-blind, placebo-controlled study. The
`study was approved by the institutional review boards of
`the participating centers, and all subjects gave informed
`consent. At enrollment, patients were between the ages of
`18 and 55 years and had an Expanded Disability Status
`Scale (EDSS) score #5.5. For patients with an EDSS score
`#3, the history of MS was limited to a maximum duration
`of 10 years. All patients had at least two exacerbations
`within the previous 2 years, but were clinically stable for 4
`weeks before the screening MRI and during the 4 weeks
`between screening and study entry. With the exception of
`glucocorticoids, any prior administration of agents with a
`putative effect on MS (including interferons, cyclophospha-
`mide, or azathioprine) led to exclusion. Treatment with
`glucocorticoids was not permitted within a 4-week period
`before the screening visit or between screening and base-
`line. Other exclusion criteria included the diagnosis of pri-
`mary progressive MS and inability to undergo MRI
`scanning. A randomization list with treatment blocks (four
`patients per block) was computer generated by Hoff-
`mann–La Roche (Basel, Switzerland) for each investiga-
`tion site. During the conduct of the study, the rando-
`mization list was available only to the Safety Review
`Board (SRB) members (see below). A limited number of
`Roche staff were unblinded at the time of the first analysis
`of efficacy as defined in the protocol. On study termination,
`each investigational site was provided with the site-
`specific randomization code.
`Eligible patients were randomized to 10, 50, or 100 mg
`of lenercept or to placebo, administered IV every 4 weeks.
`Study duration was 48 weeks, consisting of a 24-week,
`double-blind treatment period and a 24-week follow-up pe-
`riod. Of the 168 patients randomized to treatment, one
`patient (randomized to placebo) was identified as ineligible
`prior to the baseline visit; this patient did not receive
`treatment, have a baseline MRI scan, or return for follow-
`up. For the 167 patients who received treatment, compli-
`ance to treatment and study procedures was excellent.
`During the first 24 weeks, 99% (991/1002) of all planned
`doses were administered and 98% (1303/1336) of all MRI
`scan sets were performed.
`During the follow up period (study weeks 25–48), pa-
`tients could continue double-blind treatment on a volun-
`tary basis and 130 elected to do so. Those patients who
`opted not to continue treatment remained in the study and
`were followed on an intent-to-treat basis. For the full study
`duration, 10 doses (median) were administered to each
`treatment group.
`For safety purposes, three cohorts of up to 16 patients
`were enrolled in an ascending-dose design at approxi-
`mately 6-week intervals. The first cohort was randomized
`to placebo or 10 mg of lenercept whereas subsequent co-
`horts were randomized to placebo or 50 mg and finally to
`placebo or 100 mg of lenercept. An independent SRB eval-
`uated the unblinded study data before each dose escalation
`during the ascending dose phase of the study. Following
`these evaluations, the remaining patients were recruited.
`
`458 NEUROLOGY 53 August (1 of 1) 1999
`
`The SRB reviewed data at 3-month intervals throughout
`the study. This review included the MRI safety data but
`did not include a review of the MRI efficacy data.
`Magnetic resonance imaging. MRI scans were per-
`formed according to a predefined MRI protocol at screen-
`ing, baseline, and every 4 weeks (before each dose)
`throughout the first 24 weeks of the study for a total of
`eight scanning time points. At each time point, three scans
`with a slice thickness of 5 mm were obtained: 1) proton
`density/T2-weighted scan, 2) T1-weighted unenhanced
`scan, and 3) T1-weighted gadolinium (Gd)-enhanced scan 5
`minutes after the administration of Gd-DTPA 0.1 mmol/
`kg. All scans were analyzed according to a prospectively
`defined MRI analysis plan by the UBC MS/MRI Analysis
`Group in Vancouver, Canada. After comparison of each
`MRI follow-up scan with the prior scan, the number of
`newly active lesions was ascertained by summing the new,
`recurrent, or enlarging T2 lesions, and the new or recur-
`rent Gd-enhancing lesions. Newly active lesions identified
`on both the enhanced T1 scan and the T2 scan were
`counted as single lesions. The primary efficacy measure
`was the cumulative number of newly active lesions identi-
`fied on the six treatment scans. Definitions of new, re-
`current, and enlarging lesions have been reported
`previously.17,18 Persistently enhancing or enlarging lesions
`were separately identified as persistently active lesions, a
`secondary measure of efficacy. In this way, new lesions
`could easily be separated from other types of activity.
`Other secondary efficacy measures included the percent-
`age of active scans, defined as the proportion of scans with
`one or more newly active lesions, and the burden of dis-
`ease, which was assessed as reported previously, at base-
`line, and at 24 weeks.17,18 Burden of disease was
`determined by outlining each MS lesion identified on the
`T2-weighted MRI scan. These areas were summed slice by
`slice for a total lesion area recorded as mm3. In addition,
`the total number of Gd-enhancing lesions (a measure of
`safety) was counted for each patient at each scanning time
`point on an ongoing basis to allow MRI data to be reviewed
`by the SRB.
`Clinical assessments. At the baseline visit and every 4
`weeks thereafter for the first 24 weeks, a history was
`taken, physical and neurologic examinations performed,
`and adverse events noted. Study drug was administered at
`the end of each visit. Patients were encouraged to come for
`additional visits should exacerbations occur between visits.
`During the second 24-week period, two formal visits were
`planned at weeks 36 and 48. Whenever possible, patients
`who withdrew from treatment were asked to continue all
`study procedures including all MRI scans.
`Clinical endpoints. Exacerbations were defined as the
`appearance of a new sign or symptom or the worsening of
`an old sign or symptom attributable to MS, lasting at least
`24 hours in the absence of fever, and preceded by a period
`of stability of 28 days. An exacerbation was deemed to
`have ended when signs and symptoms had begun to im-
`prove. For those patients with permanent deficits, the first
`day of a 28-day period of stability was taken as the ending
`of an attack. The Neurological Rating Scale (NRS)19 was
`completed each time the neurologic examination was per-
`formed. As in other clinical trials in MS, a decline in NRS
`score of 15 points or more was considered to reflect a se-
`vere change in the patient’s neurologic condition, whereas
`declines of 8–14 or 1–7 points were considered to reflect
`
`Page 2 of 51
`
`Page 2 of 51
`
`Page 2 of 9
`
`

`

`moderate or mild changes, respectively.17,20 A difference of
`0 points was categorized as no change; an increase in the
`score as an improvement. The EDSS as recommended by
`Kurtzke21 was scored at screening and at weeks 24 and 48
`by the study neurologist. In 120/167 (72%) of patients,
`the EDSS was performed at all time points by the same
`neurologist.
`In accordance with the protocol, a first analysis was
`undertaken after all patients had completed 24 weeks of
`double-blind treatment and after all MRI scans had been
`evaluated. An increase in the exacerbation rate was noted
`in lenercept-treated patients. This finding resulted in the
`sponsor’s decision to terminate the study and to release
`the treatment code. All study drug administration was
`stopped promptly and, after a final visit, data collection
`was discontinued. For this reason, study data through
`week 48 are incomplete. The follow-up period through
`week 48, however, was similar in all treatment groups.
`Pharmacokinetic/dynamic parameters. Serum samples
`were obtained at baseline and before dosing every 4 weeks
`for 24 weeks and at study weeks 36 and 48. The concentra-
`tion of lenercept and titers of antibodies to lenercept were
`determined. All samples were analyzed centrally. Lener-
`cept concentrations were measured using an enzyme-
`linked immunologic and biologic binding assay (ELIBA)
`developed by Roche (Hoffmann-LaRoche Ltd., Basel,
`Switzerland); antibodies to lenercept were identified by
`means of a double antigen antibody test. Samples with
`detectable anti-lenercept antibodies were further evalu-
`ated to determine the neutralizing potential of the antibod-
`ies (Medi-Lab, Medicinsk Laboratorium A/S, Copenhagen,
`Denmark).
`Autoantibodies. Serum samples were obtained at base-
`line and at study weeks 24 and 48 and assayed for IgM–
`rheumatoid factor (RF), antinuclear antibodies (ANA)
`(Hep 2), and antibodies to dsDNA (DAKO; Carpinteria,
`CA) in a central laboratory (A. Wiik, Statens Seruminsti-
`tut Copenhagen, Denmark).
`Statistical analyses. The cumulative number of newly
`active lesions was tested with a closed test procedure
`based on an analysis of variance (ANOVA) of the Ln(x 1 1)
`transformed sum of the lesions. The protocol required that
`the analysis of the primary efficacy criterion be performed
`after imputation of the median number of lesions at a
`specific time point so as to compensate for missing values
`at that time point. Of the 1,008 expected values, 34 were
`missing, resulting in data imputation as noted above. The
`results of this analysis showed no differences among the
`treatment groups ( p 5 0.417) or between the pairs of
`treatment groups. Data imputation was not performed for
`the analyses presented herein.
`A closed tests procedure, based on an ANOVA with the
`factor “treatment” of Ln(1 1 x), where x denotes the cumu-
`lative number of newly active lesions, was used to compare
`the cumulative number of newly active lesions between the
`treatment groups. The closed tests procedure was first
`used to compare the means among all four treatment
`groups (global test)22; then, to compare the means among
`all combinations of three of the four treatment groups; and
`finally, to compare the means of each lenercept treatment
`group with that of the placebo group. For all comparisons,
`F tests were performed at the same significance level (a 5
`0.05); however, adjusted p values are provided. The mean
`
`of a treatment group is regarded as significantly different
`from that of the placebo group when all comparisons that
`include the two relevant treatment groups result in a p
`value #0.05; i.e., the adjusted p value is the maximum of
`the p values of these comparisons. The procedure stops
`early if the global test is nonsignificant. This procedure
`guarantees a multiple a 5 0.05. The closed tests procedure
`described above was also used to assess the cumulative
`number of persistently active lesions. Center effects were
`assessed using descriptive methods.
`The Kruskal-Wallis test was used to compare the mean
`change in EDSS scores, change in the burden of disease,
`and percent of active scans. To assess the influence of
`baseline imbalances in MRI activity among the treatment
`groups, covariance analyses using the corresponding trans-
`formed baseline MRI values as covariate were performed.
`Survival analysis methods (Kaplan-Meier estimates;
`i.e., product-limit estimates and logrank tests)23 were ap-
`plied to analyze the time to first exacerbation and the
`duration of exacerbations because of right censoring at the
`end of the observation period. Logrank tests, with a Bon-
`ferroni adjustment of the significance level (a 50.017),
`were used for the multiple comparisons among the three
`lenercept treatment and the placebo treatment Kaplan-
`Meier curves. As an exacerbation duration can only be
`observed in the presence of an exacerbation, we investi-
`gated the conditional distribution of their durations. After
`inspection of the exacerbation data, we assumed a count-
`ing process model according to Anderson and Gill24 with
`independent increments because the process is slow. For
`the same reason, exacerbation durations were assumed to
`be independently and identically distributed between pa-
`tients and within patients for those patients who had more
`than one exacerbation.
`A chi-square was used to evaluate the number of pa-
`tients with no, one, two, three, or four exacerbations in
`each treatment group after 24 weeks of treatment and at
`the end of the study (through week 48). Because of small
`frequencies in some cells, the table was collapsed to count-
`ing patients with and without exacerbations to allow a
`valid chi-square test. For the multiple comparisons the
`unadjusted p values should be compared with the Bonfer-
`roni adjusted a of 0.017 (0.05 4 3). Chi-square tests were
`also used to evaluate the NRS and the rate of RF or ANA
`among the treatment groups. Cox regression analysis was
`performed to assess potential predictive factors for the oc-
`currence of exacerbations. The data and analyses were
`performed on all data available, i.e., through week 48,
`unless otherwise stated in the text or tables. Two-tailed
`analyses were used throughout.
`
`Results. Figure 1 depicts the trial profile. The treatment
`groups were comparable at entry on all baseline disease
`characteristics and demographics (table 1). The protocol
`permitted enrollment of both relapsing-remitting and sec-
`ondary progressive patients, and from 4 to 10 patients
`with secondary progression were enrolled in each group
`(see table 1). Prestudy MRI characteristics were likewise
`comparable among the treatment groups (table 2) although
`there was a tendency (nonsignificant) for the higher lener-
`cept dose groups to have more MRI activity (median).
`MRI results. The results of the cumulative number of
`newly active MRI lesions, the percentage of persistently
`active lesions, the percentage of active scans, and the
`
`August (1 of 1) 1999 NEUROLOGY 53 459
`
`Page 3 of 51
`
`Page 3 of 51
`
`Page 3 of 9
`
`

`

`Figure 1. Profile of the lenercept MS
`clinical trial.
`
`change in burden of disease over 24 weeks of treatment are
`shown in figure 2 and table 3. There were no significant
`differences between the treatment groups for any measure.
`The results of the analyses of the primary efficacy criterion
`according to the protocol specifications were similar to
`those presented here. Because of the tendency for higher
`activity at baseline in the high-dose groups (see table 2),
`covariance analyses using the corresponding transformed
`baseline MRI values as covariate were performed, but
`
`these, too, failed to show a significant difference between
`the groups.
`Exacerbations. The number of
`Clinical endpoints.
`patients who developed exacerbations by week 24 and
`through study week 48 were both increased in the lener-
`cept groups as compared with the placebo treatment group
`as shown in table 4. A center effect was not present. Over
`the entire study period, a total of 36 exacerbations was
`reported in patients taking placebo as compared with 38,
`
`Table 1 Demographic and baseline characteristics of patients entered into the lenercept MS trial
`
`Characteristics
`
`% Female
`Age, y, mean (range)
`% White
`No. with SPMS
`Mean (range) no. exacerbations
`in prior 2 years
`EDSS, mean (range)
`NRS, mean (range)
`
`Placebo,
`n 5 44
`
`66
`36.5 (21–50)
`98
`10
`2.7 (2–5)
`
`2.45 (0–5.5)
`83.2 (51–100)
`
`10, n 5 44
`
`80
`34.6 (23–51)
`100
`5
`2.8 (2–8)
`
`2.52 (0–5.0)
`83.7 (44–100)
`
`Lenercept, mg
`
`50, n 5 40
`
`78
`35.1 (19–47)
`100
`10
`2.8 (2–8)
`
`2.83 (1.0–5.5)
`81.8 (54–100)
`
`100, n 5 40
`
`73
`34.9 (21–51)
`93
`4
`3.0 (2–6)
`
`2.55 (0–5.5)
`83.0 (57–99)
`
`SPMS 5 secondary progressive MS; EDSS 5 Expanded Disability Status Scale; NRS 5 Neurological Rating Scale.
`
`Table 2 MRI measurements at baseline of patients entered into the lenercept MS trial
`
`Lenercept, mg
`
`Variable
`
`Placebo, n 5 44
`
`10, n 5 44
`
`50, n 5 40
`
`100, n 5 40
`
`Newly active lesions, mean
`Median (range)
`
`Persistently active lesions, mean
`Median (range)
`
`1.8
`0 (0–16)
`
`1.2
`0 (0–15)
`
`1.5
`0 (0–55)
`
`0.5
`0 (0–6)
`
`2.1
`1 (0–11)
`
`1.6
`0 (0–22)
`
`1.9
`1 (0–14)
`
`0.6
`0 (0–7)
`
`Burden of disease, mean
`Median (range)
`
`2,459.9
`1,626.0 (26.2–9,888.7)
`
`2,236.2
`1,147.4 (0–14,377.4)
`
`3,757.2
`2,142.4 (67.3–16,319.0)
`
`2,707.2
`1,365.3 (58.7–10,884.8)
`
`460 NEUROLOGY 53 August (1 of 1) 1999
`
`Page 4 of 51
`
`Page 4 of 51
`
`Page 4 of 9
`
`

`

`Figure 2. (A) Number of cumulative
`newly active lesions as determined by
`MRI (see Methods) over the first 24
`weeks of the lenercept MS trial:
`placebo –—, lenercept 10 mg - - - -, le-
`nercept 50 mg — - — -, lenercept 100
`mg – – – –. The vertical bars give the
`standard error at the four weekly inter-
`vals at which MRI scans were per-
`formed. (B) The mean number of
`gadolinium (Gd)–positive lesions every
`4 weeks over the first 24 weeks of the
`trial. Vertical bars give the standard
`error. (C) Mean anti-lenercept antibody
`titers at four weekly intervals.
`
`57, and 49 exacerbations in patients taking 10, 50, and 100
`mg of
`lenercept, respectively. Exacerbation duration
`showed a tendency to increase with lenercept treatment,
`but this did not reach statistical significance (see table 4).
`This assessment was limited to exacerbations with an on-
`set date within the first 24 weeks of the study as exacerba-
`tion resolution dates were available in all but four
`exacerbations (one per treatment group).
`Exacerbation rate. The overall exacerbation rate in
`patients treated with placebo was approximately one exac-
`erbation/patient/year (the expected placebo rate). The ex-
`
`acerbation rate was increased over the placebo rate by 2%,
`68%, and 50% in patients treated with lenercept at doses
`of 10, 50, and 100 mg, respectively (see table 4). To control
`for a possible effect of unequal follow-up of patients be-
`tween treatment groups, exacerbation rates were deter-
`mined for each treatment group by 4-week intervals. The
`mean 4-week exacerbation rates were then converted to
`annual rates as presented in table 4.
`There was a dose-dependent decrease in the time to
`first exacerbation as shown in figure 3 (logrank test:
`global, p 5 0.0006; 10 mg versus placebo, p 5 0.498; 50 mg
`
`Table 3 MRI measurements over the first 24 weeks of the lenercept MS trial
`
`Lenercept, mg
`
`Variable
`
`Placebo, n 5 43
`
`10, n 5 44
`
`50, n 5 40
`
`100, n 5 40
`
`Newly active lesions, mean
`Median (range)
`Persistently active lesions, mean
`Median (range)
`Percent of active scans,† mean
`Median (range)
`Percent change in burden of disease,† mean
`Median (range)
`
`* Analysis of variance of Ln(1 1 x).
`† Kruskal-Wallis test.
`
`8.9
`4.0 (0–92)
`6.2
`1.0 (0–100)
`45.6
`50 (0–100)
`6.0
`22.2 (228.8–280.9)
`
`8.2
`3.0 (0–55)
`3.3
`1.0 (0–49)
`40.6
`33 (0–100)
`9.9
`0.0 (230.5–251.0)
`
`15.5
`5.5 (0–124)
`9.6
`1.0 (0–128)
`51.6
`50 (0–100)
`4.3
`1.4 (235.2–62.8)
`
`12.0
`4.5 (0–102)
`3.3
`1.0 (0–24)
`49.2
`55 (0–100)
`4.9
`20.2 (236.0–81.4)
`
`p
`Value
`
`0.43*
`
`0.36*
`
`0.58†
`
`0.74†
`
`August (1 of 1) 1999 NEUROLOGY 53 461
`
`Page 5 of 51
`
`Page 5 of 51
`
`Page 5 of 9
`
`

`

`Table 4 Number, duration, and annual rate of exacerbations during the lenercept MS trial
`
`Exacerbations
`
`Patients with at least one exacerbation through week 24, n
`Patients with at least one exacerbation through week 48, n
`Exacerbations with onset # week 24
`Duration (days) of these exacerbations, mean
`Median (range)
`Annualized exacerbation rate
`
`Placebo,
`n 5 43
`
`15
`22
`22
`28.3
`28 (1–91)
`0.98
`
`Lenercept, mg
`
`10, n 5 44
`
`50, n 5 40
`
`100, n 5 40
`
`21
`26
`28
`38.6
`31 (6–189)
`1.00
`
`28
`32
`37
`41.6
`31 (6–201)
`1.64
`
`27
`32
`33
`42.0
`25 (4–261)
`1.47
`
`p
`Value
`
`0.003*
`0.007†
`
`0.62‡
`
`* Chi-square tests: global.
`† Kruskal-Wallis test.
`‡ Kaplan-Meier (KM) (means and medians are estimated from the KM curves).
`
`versus placebo, p 5 0.0006; and 100 mg versus placebo,
`p 5 0.006). The comparisons with unadjusted p values for 50
`and 100 mg of lenercept as presented above remain signifi-
`cant when Bonferroni adjusted (a of 0.017, i.e., 0.05 4 3).
`The severity of exacerbations was assessed indirectly by
`computing the difference between the best pretreatment
`NRS score (highest value at baseline or screening) and the
`worst score recorded at any time during the study, i.e.,
`through week 48. Although a trend for increasing severity
`with lenercept treatment can be perceived (table 5), this
`difference did not reach statistical significance.
`Exacerbations did not appear to occur more frequently
`immediately before or after study drug administration, but
`this may reflect no more than difficulties in determining
`exacerbation onset dates with precision.
`Predictors of exacerbations. A Cox regression was per-
`formed. None of the following was identified as predictive
`of exacerbations: age, sex, MS category, number of exacer-
`bations within 2 years before the study, baseline EDSS
`score, or baseline number of newly active lesions.
`
`Figure 3. Proportion of patients remaining exacerbation
`free over the course of the lenercept MS trial: placebo –—,
`lenercept 10 mg - - - - , lenercept 50 mg - – - –, lenercept
`100 mg – – – –. Times of termination for individual pa-
`tients are shown as vertical bars.
`
`462 NEUROLOGY 53 August (1 of 1) 1999
`
`EDSS. There was no difference in EDSS score changes
`between the treatment groups after 24 weeks of treatment
`or at the last assessment (table 5). Sixty-seven percent of
`patients in the placebo group reported new or worsening
`MS symptoms during the study as compared with 77%,
`90%, and 93% of patients treated with lenercept in the 10-,
`50-, and 100-mg dose groups, respectively. Symptoms that
`increased in frequency or severity with lenercept treat-
`ment included sensory complaints, limb weakness, visual
`impairment, fatigue, vertigo, and spasm (table 6).
`Safety. Six patients withdrew from treatment during
`the study as a result of an adverse event. Depression wors-
`ened in one patient in the placebo group; development of a
`rash after the first injection led to the withdrawal of two
`patients, one in the 10-mg dose group and the other in the
`100-mg dose group. Three additional patients withdrew
`from the 100-mg dose group, one due to the occurrence of a
`transient episode of flushing, dyspnea, and gastralgia after
`the fifth dose and two because of exacerbation-related
`symptoms after the fifth and seventh doses, respectively.
`Ninety-five percent of patients treated with placebo had at
`least one adverse event reported as compared with 87%,
`90%, and 95% of patients treated with 10, 50, and 100 mg
`of lenercept, respectively. Adverse events that increased in
`frequency with active treatment included headache, hot
`flushes, nausea, and abdominal pain (see table 6). A tran-
`sient episode of dyspnea associated with the administra-
`tion of lenercept at doses of 50 and 100 mg occurred in six
`patients.
`Laboratory. RF (IgM) and ANA were present in 1%
`(2/162) and 20% (33/161) of patients at baseline, respec-
`tively. During the study, five patients developed a positive
`IgM-RF (all in the lenercept treatment groups) and 15
`patients developed ANA, 14 of whom were receiving lener-
`cept. One patient on lenercept developed both antibodies.
`Thus, new occurrences of RF or ANA were more frequent
`in lenercept-treated than in placebo-treated patients (18/
`124 versus 1/43; chi-square p 5 0.03). Clinical mani-
`festations of rheumatoid arthritis or systemic lupus
`erythematous were not observed, and dsDNA was consis-
`tently negative in all patients in whom ANA positivity was
`detected.
`Total serum IgM had increased in a dose-dependent
`fashion in lenercept-treated patients after 24 weeks of
`treatment by an average of 0.6, 0.9, and 1.0 g/L in the 10-,
`
`Page 6 of 51
`
`Page 6 of 51
`
`Page 6 of 9
`
`

`

`Table 5 Change in the NRS and the EDSS during the lenercept MS trial
`
`Lenercept, mg
`
`Rating scale
`
`Placebo, n 5 43
`
`10, n 5 44
`
`50, n 5 40
`
`100, n 5 40
`
`p
`Value
`
`0.37*
`
`0.24†
`
`NRS, weeks 1–48
`No change or improved
`Mild deterioration
`Moderate deterioration
`Severe deterioration
`EDSS, weeks 1–24, n
`Mean change (range)
`EDSS, weeks 1–48
`Mean change (range)
`
`* Chi-square.
`† Kruskal-Wallis test.
`
`14
`21
`3
`5
`43
`20.131 (22.0 to 2.0)
`
`13
`18
`10
`3
`44
`20.295 (23.0 to 5.5)
`
`9
`14
`8
`9
`40
`20.05 (22.0 to 2.0)
`
`9
`18
`7
`6
`40
`20.075 (112.0 to 4.0)
`
`0.014 (21.0 to 2.0)
`
`20.024 (23.0 to 5.5)
`
`20.026 (22.0 to 2.0)
`
`0.171 (22.0 to 4.0)
`
`0.71†
`
`NRS 5 Neurological Rating Scale; EDSS 5 Expanded Disability Status Scale.
`
`50-, and 100-mg dose groups. For those patients in the 100
`mg dose group who remained on treatment beyond 24
`weeks, the mean increase reached 1.2 g/L. Total serum IgG
`concentrations had increased by an average of 0.5 g/L at 24
`weeks on treatment in patients receiving 100 mg of lener-
`cept; for those patients who remained on treatment in the
`100-mg dose group, this increase ultimately reached 1.2
`g/L. Serum IgG concentrations were not increased in the
`other groups.
`Pharmacokinetics. Trough serum concentrations of le-
`nercept were detectable in only a third of the patients in
`all dose groups and tended to relate to the anti-lenercept
`antibody profile. Thus, patients without antibodies had
`persistently detectable lenercept trough serum concentra-
`
`Table 6 Patient complaints over the course of the lenercept MS
`trial
`
`Lenercept, mg
`
`Placebo,
`n 5 43
`
`10,
`n 5 44
`
`50,
`n 5 40
`
`100,
`n 5 40
`
`67
`44
`19
`19
`14
`2
`2
`
`23
`7
`5
`5
`0
`
`77
`45
`20
`14
`14
`7
`5
`
`36
`7
`11
`18
`0
`
`90
`58
`33
`28
`25
`13
`7
`
`35
`8
`23
`23
`2
`
`93
`63
`43
`40
`28
`15
`13
`
`45
`18
`15
`8
`13
`
`Patient complaints
`
`MS-related
`Any MS symptom
`Sensory complaints
`Limb weakness
`Visual impairment
`Fatigue
`Vertigo
`Spasm
`Other
`Headache
`Hot flushes
`Nausea
`Abdominal pain
`Dyspnea
`
`Values are %.
`
`tions, whereas patients with high antibody concentrations
`had consistently undetectable trough levels. Antibodies to
`lenercept at a concentration greater than 20 ng/mL, as
`determined by the double-antigen screening assay, were
`detected in the majority of patients on treatment (5%
`[2/43] of patients on placebo, as compared with 98% [43/
`44], 88% [35/40], and 100% [40/40] of patients in the 10-,
`50-, and 100-mg dose groups, respectively.) Antibody con-
`centrations varied considerably from patient to patient and
`over time; generally, a dose-dependent peak occurred after
`the first dose, followed by a relatively stable level that
`persisted for as long as the patient continued to receive
`lenercept (see figure 2).
`
`Discussion. Lenercept treatment in the phase II
`study reported here increased MS attack frequency.
`There was also a suggestion that lenercept increased
`attack duration and worsened attack severity as
`judged by the extent of changes noted on the NRS.
`The lenercept effect was more pronounced at the two
`higher doses employed (50 and 100 mg) than at the
`lowest dose (10 mg). An increase in attack frequency
`was noted in lenercept-treated patients within the
`first month on drug and persisted throughout the
`period during which drug was given. The magnitude
`of the treatment effect on exacerbations could be bi-
`ased because the trial was stopped early. We believe
`this is unlikely as the results of the second 24-week
`study period were consistent with those found during
`the first 24 weeks.
`Despite the seemingly deleterious clinical effects
`of lenercept, no meaningful difference between treat-
`ment groups was noted in terms of worsening of the
`EDSS score. Whereas MS attack frequency and sub-
`sequent development of disability have correlated in
`large patient series studied over many years, no such
`correlation was noted in the current study, perhaps
`because of the relative insensitivity of the EDSS and
`the prompt cessation of the study following the in-
`terim monitoring analysis.
`August (1 of 1) 1999 NEUROLOGY 53 463
`
`Page 7 of 51
`
`Page 7 of 51
`
`Page 7 of 9
`
`

`

`It is important to remember that had the current
`study shown a positive treatment effect, the results
`would have had to be validated in a second trial.
`Prudence dictates that similar caution be applied to
`the detrimental clinical effect noted in the current
`trial, particularly in view of the seemingly discor-
`dant MRI data. Setting caution aside, it is reason-
`able to conclude that lenercept is probably con-
`traindicated in MS based on the internal consistency
`of the clinical results presented here and the unto-
`ward results obtained in a preliminary study of two
`patients administered a humanized mouse monoclo-
`nal anti-TNF antibody.25 The dissociation seen be-
`tween a significantly increased clinical activity and,
`at best, a slight trend toward an increase in M

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