Está en la página 1de 10

ARTHRITIS & RHEUMATISM

Vol. 52, No. 2, February 2005, pp 563–572


DOI 10.1002/art.20860
© 2005, American College of Rheumatology

A Randomized, Double-Blind Clinical Trial of Two Doses of


Meloxicam Compared With Naproxen in Children With
Juvenile Idiopathic Arthritis
Short- and Long-Term Efficacy and Safety Results

Nicolino Ruperto,1 Irina Nikishina,2 Evgueni D. Pachanov,3 Ynessa Shachbazian,4


Anne Marie Prieur,5 Richard Mouy,5 Rik Joos,6 Francesco Zulian,7 Rudolf Schwarz,8
Valeria Artamonova,9 Wolfgang Emminger,10 Marcia Bandeira,1 Antonella Buoncompagni,1
Ivan Foeldvari,11 Fernanda Falcini,12 Eileen Baildam,13 Isabelle Kone-Paut,14 Maria Alessio,15
Valeria Gerloni,16 Alessandro Lenhardt,17 Alberto Martini,18 for the Pediatric Rheumatology
International Trials Organization (PRINTO), Gertraud Hanft,19 Ralf Sigmund,19 and Stefan Simianer19

Objective. In an international, multicenter, cacy and safety of 2 different doses of meloxicam oral
double-blind, randomized clinical trial we evaluated the suspension compared with the efficacy and safety of
short-term (3 months) and long-term (12 months) effi- naproxen oral suspension in children with
oligoarticular-course (oligo-course) or polyarticular-
Supported by a grant to the Pediatric Rheumatology Interna- course (poly-course) juvenile idiopathic arthritis (JIA).
tional Trials Organization from Boehringer Ingelheim Pharma GmbH
& Co. KG, Biberach, Germany. Methods. Children ages 2–16 years who had ac-
1
Nicolino Ruperto, MD, MPH, Marcia Bandeira, MD, An- tive oligo-course or poly-course JIA and who required
tonella Buoncompagni, MD: IRCCS G. Gaslini, Pediatria II, Reumato-
logia, Genoa, Italy; 2Irina Nikishina, MD: Institute of Rheumatology of
therapy with a nonsteroidal antiinflammatory drug
the Russian Academy of Medical Science, Moscow, Russia; 3Evgueni D. were eligible for this trial. Patients were randomly
Pachanov, MD: Scientific Research Institute of Pediatrics, Moscow, allocated to receive therapy with meloxicam oral sus-
Russia; 4Ynessa Shachbazian, MD: Moscow Medical Academy, Moscow,
Russia; 5Anne Marie Prieur, MD, Richard Mouy, MD: Hôpital Necker pension, 0.125 mg/kg body weight in a single daily dose;
des Enfants Malades, Paris, France; 6Rik Joos, MD: Universitair Zieken- meloxicam oral suspension, 0.25 mg/kg body weight in a
huis Gent, Ghent, Belgium; 7Francesco Zulian, MD: Unità di Rheuma-
tologia Pediatrica, Padua, Italy; 8Rudolf Schwarz, MD: Landeskinderk-
single daily dose; or naproxen, 10 mg/kg body weight in
linik Linz, Linz, Austria; 9Valeria Artamonova, MD: Morozovskaya 2 daily doses. The trial drugs were administered in a
Children City Clinical Hospital, Moscow, Russia; 10Wolfgang Emminger, double-blind, double-dummy design for up to 12
MD: University of Wien, Vienna, Austria; 11Ivan Foeldvari, MD: Kinder-
rheumatologische Praxis am AK Eilbek, Hamburg, Germany; 12Fernanda months. Response rates were determined according to
Falcini, MD: Clinica Pediatrica III, Florence, Italy; 13Eileen Baildam, the American College of Rheumatology pediatric 30%
MD: Booth Hall Children’s Hospital, Manchester, UK; 14Isabelle Kone- improvement criteria (ACR pediatric 30). Safety para-
Paut, MD: Hôpital Nord, Pavillon Mère-Enfant, Marseilles, France;
15
Maria Alessio, MD: Università Federico II, Naples, Italy; 16Valeria meters were assessed by evaluating the frequency of
Gerloni, MD: Instituto Gaetano Pini, Milan, Italy; 17Alessandro Len- adverse events in the 3 groups.
hardt, MD: IRCCS Burlo Garofalo, Trieste, Italy; 18Alberto Martini, MD,
IRCCS G. Gaslini, Pediatria II, Reumatologia, and University of Genoa, Results. Of 232 patients enrolled, 225 received
Genoa, Italy; 19Gertraud Hanft, MD, Ralf Sigmund, Dipl.-Math. Dec., treatment, 6 were not eligible for randomization, and 1
Stefan Simianer, MD: Boehringer Ingelheim Pharma GmbH & Co. KG, randomized patient was not treated. One hundred
Biberach, Germany.
Dr. Foeldvari has received consulting fees of less than $10,000 eighty-two patients (81%) completed the 12-month
per year from Bristol-Meyers Squibb. treatment period. Response rates according to the ACR
Address correspondence and reprint requests to Nicolino Ru-
perto, MD, MPH, Pediatric Rheumatology International Trials Organi-
pediatric 30 criteria improved from month 3 to month
zation, IRCCS G. Gaslini Pediatria II–Reumatologia, University of 12, as follows: from 63% to 77% in the meloxicam 0.125
Genoa, Largo Gaslini, 5, 16147 Genoa, Italy. E-mail: nicolaruperto@ mg/kg group, from 58% to 76% in the meloxicam 0.25
ospedale-gaslini.ge.it.
Submitted for publication July 5, 2004; accepted in revised mg/kg group, and from 64% to 74% in the naproxen
form November 4, 2004. group. No statistically significant differences in re-
563
564 RUPERTO ET AL

sponse rates were observed between the groups. There PATIENTS AND METHODS
were no differences in the frequency of adverse events or The trial was conducted in accordance with the 1996
abnormal laboratory values between the 3 groups. Declaration of Helsinki. Each investigational site obtained
Conclusion. The short- and long-term safety and approval from its independent institutional review board
efficacy of meloxicam oral suspension appear to be and/or relevant ethics committee before the start of the trial,
comparable with the safety and efficacy of naproxen oral and all parents/legal guardians of the patients gave written
informed consent. Children who were able to understand the
suspension in the treatment of oligo-course and poly- given information also had to provide their active assent to
course JIA. The once-daily administration of meloxicam participate in the study.
oral suspension might represent an improvement in the Patients. Children ages 2–16 years with a diagnosis of
treatment of JIA. JIA, as defined according to the Durban criteria (9), and for
whom therapy with an NSAID was required were candidates
for the trial. In order to be eligible, patients had to have at least
Juvenile idiopathic arthritis (JIA) encompasses a 2 joints with active arthritis plus abnormal results in at least 2
heterogeneous group of disorders involving chronic ar- of any of the 5 remaining JIA core set criteria (10,11).
Exclusion criteria included the presence of current systemic
thritis, disease onset before 16 years of age, and un- manifestations (e.g., fever, rash, hepatosplenomegaly); abnor-
known etiology. With a reported prevalence of 86.1–94 mal, clinically relevant laboratory values unrelated to JIA;
per 100,000 children (1), JIA is the most common pregnancy, breast-feeding, and/or inadequate contraception in
chronic rheumatic disease in childhood and is one of the females of childbearing potential; history of bleeding disor-
ders; active peptic ulcer within the past 6 months; known or
leading causes of pediatric acquired disability (2). suspected hypersensitivity reaction to NSAIDs, analgesics, or
Nonsteroidal antiinflammatory drugs (NSAIDs), antipyretics; a planned surgical procedure during the course of
either as monotherapy or in combination with other the trial; participation in another clinical trial; known drug
agents, are considered to be the agents of first choice for abuse; and other concomitant rheumatic and clinically signifi-
cant nonrheumatic conditions. Other exclusion criteria were
controlling joint inflammation in patients with all forms successful previous treatment with an NSAID; concomitant
of juvenile arthritis (3,4). In order to maintain control of therapy with other NSAIDs or with prednisone (or equivalent)
symptoms, therapy with NSAIDs may need to be con- at a dosage exceeding 0.2 mg/kg/day or 10 mg/day, whichever
tinued for several years. was lower; treatment with ⬎1 disease-modifying antirheumatic
drug within the past 3 months before enrollment, including
Meloxicam is an NSAID that selectively inhibits hydroxychloroquine, cyclosporine, methotrexate, cytotoxic
the cyclooxygenase 2 (COX-2) enzyme and has the agents, gold compounds, D-penicillamine, etanercept, or other
advantage of once-daily dosing. An oral suspension biologic agents; and intraarticular corticosteroid injections
formulation is available for use in children. Data regard- during the last month before enrollment and intended for the
first 4 weeks of therapy. Concomitant therapy had to be
ing use of meloxicam for treating the signs and symp- maintained at a stable dosage throughout the trial.
toms of adult rheumatoid arthritis demonstrate its com- Study design. The trial was performed according to a
parability with other NSAIDs in terms of efficacy and double-blind, double-dummy design in patients with oligo-
safety (5,6). Results of postmarketing surveillance stud- course or poly-course JIA. Eligible patients were allocated to 1
of the 3 treatment groups in a 1:1:1 randomization scheme.
ies also demonstrated that meloxicam is comparable Patients were seen after 2 weeks, 4 weeks, 6 weeks, 8 weeks,
with other, more selective COX-2 inhibitors with regard and 3 months, and thereafter every 3 months until month 12,
to the reduced frequency of upper gastrointestinal (GI) by their treating physician.
complications such as perforation or bleeding (7). Al- After a washout period of 1–7 days, depending on the
serum half-life of the previous NSAID, the children were
though meloxicam is currently registered in many coun- randomly allocated to 1 of 3 therapies, as follows: meloxicam,
tries for the treatment of adult rheumatoid arthritis, few 0.125 mg/kg body weight in a single daily dose; meloxicam, 0.25
data exist for its use in children (8). mg/kg body weight in a single daily dose; or naproxen, 5 mg/kg
For these reasons, we conducted an interna- body weight twice daily. To keep the trial blinded, children in
tional, multicenter, double-blind, double-dummy, ran- the meloxicam group also received naproxen placebo suspen-
sion and vice versa, in a double-dummy design. Meloxicam oral
domized clinical trial to evaluate the short-term (3 suspension or placebo was administered once daily in the
months) and long-term (12 months) efficacy and safety morning with food, while naproxen oral suspension or placebo
of 2 different doses of meloxicam oral suspension com- was administered twice daily (morning and evening). The
pared with the safety and efficacy of naproxen oral selected dosage of naproxen is the dosage registered for JIA in
most of the countries participating in the trial. The doses of
suspension in children ages 2–16 years with meloxicam chosen were equivalent to the registered adult
oligoarticular-course (oligo-course) or polyarticular- doses of 7.5 mg and 15 mg, based on pharmacokinetic and
course (poly-course) JIA. clinical data obtained in children (8). The trial drug was
MELOXICAM VERSUS NAPROXEN FOR JIA 565

administered continuously until the end of the trial. Patients Statistical analysis. Analyses were completed accord-
who missed taking their trial medication on ⬎7 consecutive ing to the recommendation of the Consolidated Standards of
days were considered noncompliant. Reporting Trials statement (25). Each patient who received at
End points. As primary outcome measures, we used least 1 dose of the trial drug was included in the analysis,
the validated American College of Rheumatology pediatric 30 according to the intent-to-treat (ITT) principle.
definition of improvement (ACR pediatric 30) (10–12). Ac- The primary analysis of efficacy was performed for the
cording to the ACR pediatric 30 criteria, patients were con- ITT population after 3 months of therapy (and every 3 months
sidered to be responders to treatment if they demonstrated at thereafter) according to the last observation carried forward
least 30% improvement from baseline in at least 3 of any 6 JIA approach for missing data. Patients who withdrew were
core set variables, with no more than 1 of the remaining counted with their response status as reported in the last
variables worsening by ⬎30% (11). The ACR pediatric 30 observation. In a second, more conservative approach, all
definition allows researchers or clinicians to dichotomize pa- patients who withdrew were considered to be nonresponders.
tients into responders or nonresponders. Patients were also
Descriptive statistics were used for reporting demo-
evaluated by ACR response levels of at least 50% and at least
graphics, clinical characteristics, efficacy variables, and adverse
70% (ACR pediatric 50 and ACR pediatric 70, respectively) in
at least 3 of any 6 JIA core set variables, with no more than 1 events. For the other secondary efficacy parameters, treatment
of the remaining variables worsening by ⬎30%. comparisons were performed using analysis of covariance at
As secondary end points, we used the individual vali- months 3, 6, 9, and 12, controlling for baseline data. Confi-
dated JIA core set variables (10,11), which include the number dence intervals were expressed with limits of 95% (95% CIs).
of joints with active arthritis, defined as a joint with swelling or The area under the curve (AUC) approach was used to
a joint with pain and limitation on movement (range 0–75) provide an average response over the entire 1-year treatment
(13); the number of joints with limited range of motion (range period. To obtain the AUC, the percent response was summed
0–67) (14); the physician’s global evaluation of disease activity for certain periods of time according to the trapezoidal rule,
on a double-anchored 100-mm visual analog scale (VAS) and the average was calculated by division through the respec-
(anchoring words 0 ⫽ inactive, 100 ⫽ very severe); the parent’s tive time intervals used. The chi-square test or the unadjusted
global assessment of the child’s overall well-being on a double- Fisher’s exact test was applied for the paired comparisons, as
anchored 100-mm VAS (anchoring words 0 ⫽ very well, 100 ⫽ appropriate. Logistic regression was used, if necessary, to
very poor) as reported on the Childhood Health Assessment analyze potential confounder variables, such as the center from
Questionnaire (C-HAQ) (15,16); the disability index of the which the patient was referred, disease severity, age, or sex.
C-HAQ; and the Westergren erythrocyte sedimentation rate Safety data were analyzed descriptively by tabulating
(ESR). The C-HAQ (15,16) is a validated disease-specific the reported adverse events according to the Medical Diction-
instrument for JIA adapted from the adult Health Assessment ary for Regulatory Activities (26). Laboratory values were
Questionnaire. It measures functional ability in 8 activities of standardized and normalized before analysis, based on consid-
daily living: dressing and grooming, arising, eating, walking, eration of age- and-sex specific differences in the trial popu-
hygiene, reach, grip, and activities. These 8 domains are then lation.
averaged into a summary score ranging from 0 to 3, with higher For the primary end point, a sample size of 219
scores meaning greater disability. The C-HAQ has been fully evaluable patients (73 per group) was determined to be
cross-culturally adapted, validated, and translated in all of the sufficient to show superiority of meloxicam (both doses)
languages of the countries participating in this trial (16–23). compared with naproxen according to the ACR pediatric 30
As additional assessments, we reported the parent’s criteria, based on an expected response rate of 60% for
evaluation of the child’s pain on a double-anchored 100-mm naproxen and a clinically meaningful delta of 20%, power of
VAS (anchoring words 0 ⫽ no pain, 100 ⫽ very severe pain) as
90%, using a 1-sided Fisher’s exact test (␣ ⫽ 5%).
reported on the C-HAQ, the parent’s global assessment of the
Random numbers were generated centrally. Individual
child’s arthritis on a double-anchored 100-mm VAS (anchor-
sealed envelopes containing the randomization for each pa-
ing words 0 ⫽ very well, 100 ⫽ very poor), the child’s
tient were sent to the referring centers together with the trial
assessment of discomfort on a facial affective scale (range 1–9
points) (24), and the final global assessment of efficacy and medication.
tolerability by the investigator (at months 3 and 12) on a verbal
rating scale (with the ratings good, satisfactory, not satisfac- RESULTS
tory, or bad).
Safety was evaluated at every visit by tabulating the A total of 232 children were enrolled from Sep-
number of reported adverse events (graded as mild, moderate, tember 2000 to December 2001. These children were
or severe) that occurred during the course of the trial, as well
as by reviewing the results of routine laboratory tests (e.g., recruited from 34 pediatric rheumatology tertiary care
differential blood cell count, and levels of liver enzymes units in 7 countries: Austria (n ⫽ 21; 3 units), Belgium
and serum creatinine) performed in local laboratories. Assess- (n ⫽ 11; 2 units), France (n ⫽ 29; 7 units), Germany
ments of efficacy and safety were performed at baseline (n ⫽ 20; 6 units), Italy (n ⫽ 45; 9 units), Russia (n ⫽ 97;
and after 2 weeks, 4 weeks, 6 weeks, 8 weeks, 3 months, 6
months, 9 months, and 12 months of therapy (except for the 4 units), and the UK (n ⫽ 9; 3 units). In a confounder
ESR, which was determined only at week 4 and at months 3, 6, analysis, the origin of the patient was found to have no
9, and 12). impact on the primary end point.
566 RUPERTO ET AL

ized to receive meloxicam, 0.25 mg/kg/day; and 78


patients were randomized to receive naproxen, 5 mg/kg
twice daily. Some differences in the patients’ baseline
characteristics or medical histories were observed be-
tween the treatment groups (Table 1), especially for age
and age group. A confounder analysis revealed that
none of the observed differences had a meaningful
impact on the primary end point, and that the effects
were not working unidirectionally for any treatment
arm. Because there is no evidence that differences in age
would impact the efficacy or safety of treatments in JIA,
the observed difference in age was considered irrelevant
with respect to the analysis of the trial.
One hundred eighty-two patients (81%) com-
pleted the 12-month treatment period. Nine patients in
the 0.125 mg/kg meloxicam group, 4 patients in the 0.25
mg/kg meloxicam group, and 13 patients in the naproxen
Figure 1. Flow diagram of the patients with juvenile idiopathic ar-
group discontinued treatment due to adverse events or
thritis.
lack of efficacy (Figure 1). Among the patients who
discontinued treatment for other reasons, 2 patients in
As shown in Figure 1, 6 of the 232 patients did each of the meloxicam groups discontinued because of
not meet the eligibility criteria for randomization, and 1 disease remission.
randomized patient withdrew consent on the same day On average, patients in the 0.125 mg/kg meloxi-
and therefore did not receive the trial drug. Of the 225 cam group were treated for 327 days with a mean daily
treated patients, 73 were randomized to receive treat- dose of 0.122 mg/kg, patients in the 0.25 mg/kg meloxi-
ment with meloxicam, 0.125 mg/kg/day; 74 were random- cam group were treated for 326 days with a mean daily

Table 1. Characteristics and disease history of the study patients at baseline*


Meloxicam, Meloxicam, Naproxen,
0.125 mg/kg 0.25 mg/kg 10 mg/kg
Characteristic (n ⫽ 73) (n ⫽ 74) (n ⫽ 78) P
Disease duration, mean ⫾ SD months 41.6 ⫾ 40.6 30.0 ⫾ 33.4 27.7 ⫾ 24.9 0.154
Age, mean ⫾ SD years 8.9 ⫾ 3.8 9.0 ⫾ 3.9 7.5 ⫾ 3.7 0.021
Age group
ⱕ6 years 23 (31.5) 20 (27.0) 37 (47.4) 0.022
7–16 years 50 (68.5) 54 (73.0) 41 (52.6)
Sex
Male 24 (32.9) 25 (33.8) 18 (23.1) 0.276
Female 49 (67.1) 49 (66.2) 60 (76.9)
JIA onset type
Oligoarticular 60 (82.2) 59 (79.7) 56 (71.8) 0.156
Polyarticular 13 (17.8) 12 (16.2) 21 (26.9)
Systemic 0 3 (4.1) 1 (1.3)
JIA subtype at enrollment†
Oligoarticular 49 (67.1) 42 (56.8) 46 (59.0) 0.398
Polyarticular 24 (32.9) 32 (43.2) 32 (41.0)
Uveitis at baseline 9 (12.3) 7 (9.5) 6 (7.7) 0.628
Use of any DMARD at baseline 18 (24.7) 21 (28.4) 29 (37.2) 0.225
Use of methotrexate at baseline 15 (20.5) 19 (25.7) 23 (29.5) 0.449
Previous intake of other NSAID 71 (97.3) 67 (90.5) 74 (94.9) 0.214
within past 5 years

* Except where indicated otherwise, values are the number (%). P values refer to the differences between
the 3 treatment groups. JIA ⫽ juvenile idiopathic arthritis; DMARD ⫽ disease-modifying antirheumatic
drug; NSAID ⫽ nonsteroidal antiinflammatory drug.
† According to investigator classification.
MELOXICAM VERSUS NAPROXEN FOR JIA 567

meloxicam 0.125 mg/kg increased from 38% (28 of 73


patients) at month 3 (95% CI 27–50%) to 53% (39 of 73
patients) at month 12 (95% CI 42–65%); in the group
assigned to receive meloxicam 0.25 mg/kg, response
rates increased from 26% (19 of 74 patients) at month 3
(95% CI 16–36%) to 43% (32 of 74 patients) at month
12 (95% CI 32–55%); and in the group receiving
Figure 2. Frequency of treatment response as determined by the naproxen, response rates increased from 29% (23 of 78
American College of Rheumatology pediatric 30 definition of im- patients) at month 3 (95% CI 19–40%) to 50% (39 of 78
provement in the 3 treatment groups. No statistically significant patients) at month 12 (95% CI 39–61%). Again, the
differences in the response rate among the 3 groups were observed. difference between the pooled meloxicam groups and
Bars show the mean and 95% confidence intervals. MEL L ⫽
meloxicam 0.125 mg/kg/day; MEL H ⫽ meloxicam 0.25 mg/kg/day;
the naproxen group was not significant (P ⫽ 0.7).
NAP ⫽ naproxen 5 mg/kg twice daily. When applying the AUC approach, according to
the ACR pediatric 30 definition, the average response
rates per month over the whole treatment period were
dose of 0.261 mg/kg, and those in the naproxen group 57%, 55%, and 54%, respectively, in the groups receiv-
were treated for 315 days with a mean daily dose of 11.52 ing meloxicam 0.125 mg/kg, meloxicam 0.25 mg/kg, and
mg/kg. naproxen. In a more conservative approach, all patients
Efficacy results. The response rate, according to who withdrew from the study were counted as nonre-
the ACR pediatric 30 criteria, increased over time (from sponders. In this analysis, the response rates according
month 3 to month 12) in all 3 treatment groups (Figure to ACR pediatric 30 criteria at month 12 decreased to
2). In the group receiving meloxicam 0.125 mg/kg, ACR 68% (50 of 73 patients) in the group receiving meloxi-
pediatric 30 response rates increased from 63% (46 of 73 cam 0.125 mg/kg (95% CI 58–79%), to 69% (51 of 74
patients) at month 3 (95% CI 52–74%) to 77% (56 of 73 patients) in the group receiving meloxicam 0.25 mg/kg
patients) at month 12 (95% CI 67–86%); in the group (95% CI 58–79%), and to 63% (49 of 78 patients) in the
receiving meloxicam 0.25 mg/kg, response rates in- group receiving naproxen (95% CI 52–74%). Similar
creased from 58% (43 of 74 patients) at month 3 (95% trends were observed for the ACR pediatric 50 and
CI 47–69%) to 76% (56 of 74 patients) at month 12 ACR pediatric 70 criteria (data not shown).
(95% CI 66–85%); and in the group receiving naproxen, As shown in Table 2 and Figure 3, patients in all
response rates increased from 64% (50 of 78 patients) at 3 treatment groups demonstrated improvement from
month 3 (95% CI 53–75%) to 74% (58 of 78 patients) at baseline in all 6 JIA core set variables and in all
month 12 (95% CI 65–84%). The primary end point additional measures; a statistically significant improve-
analysis between the pooled meloxicam groups and the ment for all JIA core set variables except the ESR, and
naproxen group showed no statistical difference (P ⫽ secondary efficacy end point parameters was observed
0.6). during the first 3 months of the trial. This improvement
According to the ACR pediatric 50 criteria, re- was maintained for the rest of the trial (P values not
sponse rates in the group receiving meloxicam 0.125 shown). The investigators rated global efficacy as good
mg/kg increased from 52% (38 of 73 patients) at month or satisfactory in 88% of patients treated with meloxi-
3 (95% CI 41–64%) to 68% (50 of 73 patients) at month cam 0.125 mg/kg, in 85% of those treated with meloxi-
12 (95% CI 58–79%); response rates in the group cam 0.25 mg/kg, and in 87% of patients treated with
receiving meloxicam 0.25 mg/kg increased from 43% (32 naproxen.
of 74 patients) at month 3 (95% CI 32–55%) to 65% (48 As exploratory analyses, we evaluated the re-
of 74 patients) at month 12 (95% CI 54–76%); and in sponse rate, according to the ACR pediatric 30 criteria,
the group receiving naproxen, response rates increased in the subgroups of patients with oligo-course JIA and
from 50% (39 of 78 patients) at month 3 (95% CI those with poly-course JIA. At month 3, all treatments
39–61%) to 68% (53 of 78 patients) at month 12 (95% had shown a trend toward better efficacy response rates
CI 58–78%). The analysis between the pooled meloxi- in patients with poly-course JIA compared with those
cam groups and the naproxen group showed no statisti- with oligo-course disease, with only the meloxicam 0.25
cal difference (P ⫽ 0.7). mg/kg group showing a statistically significant difference
Finally, according to the ACR pediatric 70 crite- (73% for poly-course versus 48% for oligo-course; P ⫽
ria, response rates in the group randomized to receive 0.03). Beyond month 3, the difference in efficacy be-
568

Table 2. Measures of disease activity and improvement from baseline*


Meloxicam, 0.125 mg/kg (n ⫽ 73) Meloxicam, 0.25 mg/kg (n ⫽ 74) Naproxen, 10 mg/kg (n ⫽ 78)

Measure Month 0 Month 3 Month 12 Month 0 Month 3 Month 12 Month 0 Month 3 Month 12
JIA core set measures
No. of active joints (0–75 joints) 6.2 ⫾ 8.4 3⫾5 2.5 ⫾ 5.2 7.3 ⫾ 8.3 3.9 ⫾ 6 3.5 ⫾ 6.3 6.7 ⫾ 7.9 3.8 ⫾ 5.2 2.9 ⫾ 4.6
No. of limited joints (0–67 joints) 6.1 ⫾ 8.5 3.4 ⫾ 5.3 2.8 ⫾ 5.8 6.6 ⫾ 7.9 4.7 ⫾ 7.1 4.7 ⫾ 8.9 6.5 ⫾ 8 4.1 ⫾ 5.7 3.3 ⫾ 5.6
Physician’s global assessment of 37.1 ⫾ 19.6 19.4 ⫾ 20.7 15.4 ⫾ 20.5 38.5 ⫾ 21.6 20.6 ⫾ 20.3 16.8 ⫾ 19 37.6 ⫾ 17.9 21.1 ⫾ 19.2 14.4 ⫾ 16.7
disease activity by VAS (0–100
mm)
Erythrocyte sedimentation rate, 16.2 ⫾ 14.7 16.5 ⫾ 15.3 14 ⫾ 14.7 21.4 ⫾ 22.8 17.2 ⫾ 17.5 14.8 ⫾ 13.5 20.5 ⫾ 18.6 19.4 ⫾ 17.1 16.7 ⫾ 16.8
mm/hour
Parent’s global assessment of 36.9 ⫾ 20.1 21.1 ⫾ 19.3 18.2 ⫾ 19.2 38.7 ⫾ 23.3 23.5 ⫾ 21.9 17.7 ⫾ 20.3 38 ⫾ 19.5 22.5 ⫾ 20.4 16.9 ⫾ 18.2
overall well-being by VAS
(0–100 mm)
Disability index of the C-HAQ 0.6 ⫾ 0.6 0.4 ⫾ 0.5 0.3 ⫾ 0.4 0.8 ⫾ 0.6 0.5 ⫾ 0.6 0.4 ⫾ 0.6 0.8 ⫾ 0.6 0.5 ⫾ 0.6 0.3 ⫾ 0.5
(0–3 points)
Additional measures
Parent’s global assessment of 35 ⫾ 22.4 17.6 ⫾ 20.2 13.4 ⫾ 17.6 39 ⫾ 24.6 21.9 ⫾ 23.6 17.2 ⫾ 22.5 38.1 ⫾ 23.4 20.8 ⫾ 22.4 15.9 ⫾ 21.3
pain by VAS (0–100 mm)
Parent’s global assessment of 40.8 ⫾ 21.9 21.7 ⫾ 19.6 16.9 ⫾ 19 42.7 ⫾ 22.8 26 ⫾ 22 19.3 ⫾ 20.4 44 ⫾ 22 24 ⫾ 21.9 17.9 ⫾ 20.5
arthritis by VAS (0–100 mm)
Child’s assessment of discomfort 0.5 ⫾ 0.2 0.3 ⫾ 0.2 0.3 ⫾ 0.2 0.5 ⫾ 0.2 0.4 ⫾ 0.2 0.3 ⫾ 0.2 0.4 ⫾ 0.2 0.3 ⫾ 0.2 0.2 ⫾ 0.2
by facial affective scale (1–9
points)

* Values are the mean ⫾ SD. Month 0 is baseline, month 3 is the end of the short-term efficacy phase, and month 12 is the end of the long-term extension phase. JIA ⫽ juvenile
idiopathic arthritis; VAS ⫽ visual analog scale; C-HAQ ⫽ Childhood Health Assessment Questionnaire.
RUPERTO ET AL
MELOXICAM VERSUS NAPROXEN FOR JIA 569

group experienced reversible hyperbilirubinemia that


resulted in withdrawal from the trial.
Table 3 shows all adverse events occurring in at
least 10% of the patients, irrespective of severity or
relatedness to the study drugs, according to the Medical
Dictionary for Regulatory Activities. The majority of ad-
verse events were considered to be mild. The incidence
of musculoskeletal disorders was highest in the group
receiving meloxicam 0.25 mg/kg, and these disorders
were attributable mainly to an arthritis flare.
Bleeding-associated disorders and skin disorders
were more often observed in the naproxen group.
Among the skin disorders, 1 case of moderate pseu-
doporphyria was observed in the naproxen group. From
baseline until the end of the trial, the number of patients
with active uveitis decreased, from 9 to 4 in the meloxi-
cam 0.125 mg/kg group, from 7 to 4 in the meloxicam
0.25 mg/kg group, and from 6 to 5 in the naproxen
group. Global tolerability was assessed by the investiga-
tor as “good” or “satisfactory” in 93% of patients in the
Figure 3. Analysis of covariance for the 6 individual variables in-
0.125 mg/kg meloxicam group and in 95% of the patients
cluded in the juvenile idiopathic arthritis core set of outcome mea-
sures, in the 3 treatment groups. P values refer to the differences treated with meloxicam 0.25 mg/kg or naproxen.
between the 3 treatment groups. Bars show the mean and 95% No remarkable changes in the laboratory para-
confidence intervals. Mel H ⫽ meloxicam 0.25 mg/kg/day; Mel L ⫽ meters were observed (data not shown), except for
meloxicam 0.125 mg/kg/day; Nap ⫽ naproxen 5 mg/kg twice daily; changes in bilirubin values. During the trial, 14 patients
ESR ⫽ erythrocyte sedimentation rate; C-HAQ ⫽ Childhood Health
in the meloxicam 0.125 mg/kg group (20% of all patients
Assessment Questionnaire.
with available data), 5 patients in the meloxicam 0.25
mg/kg group (7% of all patients with available data), and
10 patients in the naproxen group (13% of all patients
with available data) showed an increase in the bilirubin
tween patients with oligo-course JIA and those with concentration above the upper limit of normal.
poly-course JIA was vanishing or not visible. Four patients receiving the 0.125 mg/kg dose of
Safety results. Fifty-four patients (74%) in the meloxicam, 7 receiving the 0.25 mg/kg dose of meloxi-
meloxicam 0.125 mg/kg group, 59 patients (80%) in the cam, and 10 patients receiving naproxen experienced a
meloxicam 0.25 mg/kg group, and 66 patients (85%) in serious adverse event (data not shown). All serious
the naproxen group experienced at least 1 adverse event adverse events were categorized as “severe” due to the
during the course of the 12-month trial period. The fact that hospitalization was required to treat either the
investigators considered adverse events to be drug- underlying disease or intercurrent diseases such as ap-
related in 7 patients (10%) in the meloxicam 0.125 pendicitis, and none of these adverse events were con-
mg/kg group, in 11 patients (15%) in the meloxicam 0.25 sidered to be drug-related.
mg/kg group, and in 10 patients (13%) in the naproxen
group.
DISCUSSION
Seven patients in the meloxicam 0.125 mg/kg
group, 3 patients in the meloxicam 0.25 mg/kg group, The present trial was performed to assess the
and 10 patients in the naproxen group discontinued efficacy and safety of meloxicam oral suspension in the
therapy prematurely due to an adverse event (Figure 1). treatment of JIA. This study represents the first example
Among these patients, treatment was most often discon- of a large-scale randomized trial to evaluate the efficacy
tinued because of an arthritis flare (2% of all patients and safety of an NSAID selective for COX-2 inhibition
treated), followed by GI system disorders (2% of all in children with JIA.
patients treated) and skin disorders (1% of all patients Apart from aspirin, only a few NSAIDs have
treated). One patient in the meloxicam 0.125 mg/kg been studied at all, and even fewer have been approved
570 RUPERTO ET AL

Table 3. Adverse events occurring in at least 10% of the study patients*


Meloxicam, Meloxicam, Naproxen,
0.125 mg/kg 0.25 mg/kg 10 mg/kg
Adverse event (n ⫽ 73) (n ⫽ 74) (n ⫽ 78) P
Eye disorders 5 (7) 6 (8) 8 (10) 0.7
Gastrointestinal disorders 28 (38) 27 (37) 25 (32) 0.7
Pain, diarrhea, nausea, vomiting† 21 (29) 19 (26) 19 (24) 0.8
Pharyngolaryngeal pain 9 (12) 5 (7) 4 (5) 0.2
General disorders 13 (18) 14 (19) 19 (24) 0.6
Pyrexia 11 (15) 13 (18) 14 (18) 0.9
Infections and infestations 30 (41) 38 (51) 39 (50) 0.4
Nasopharyngitis 4 (6) 9 (12) 7 (9) 0.3
Physical examination‡ 9 (12) 6 (8) 4 (5) 0.3
Musculoskeletal and connective tissue disorders 11 (15) 22 (30) 10 (13) 0.02
Nervous system disorders 10 (14) 11 (15) 7 (9) 0.5
Headache NOS 9 (12) 10 (14) 5 (6) 0.3
Respiratory, thoracic, and mediastinal disorders 22 (30) 19 (26) 26 (33) 0.6
Cough 7 (10) 9 (12) 14 (18) 0.3
Rhinitis NOS 13 (18) 11 (15) 16 (21) 0.66
Skin and subcutaneous tissue disorders 4 (6) 5 (7) 13 (17) 0.049§
Eczema, erythema, pruritus, rash† 0 (0) 3 (4) 8 (10) 0.008§
Bleeding disorders (rectal hemorrhage, 3 (4) 2 (3) 9 (12) 0.07§
epistaxis, hematuria, hematoma, Henoch-
Schönlein purpura)†

* Values are the number (%). Adverse events were defined according to the Medical Dictionary for Regulatory Activities (26). Except
where indicated otherwise, P values were determined by chi-square test. NOS ⫽ not otherwise specified.
† Selected preferred terms were grouped together, and each patient was counted only once.
‡ Abnormal laboratory results or findings from physical examination (e.g., increased body temperature).
§ By Fisher’s exact test.

for use in children (e.g., naproxen, tolmetin, and ibupro- symptoms of JIA over time. The treatment effect in this
fen in the US), and most of them are used off-label, trial was expressed almost completely after 3 months of
meaning that no indication for pediatric use is reported therapy. Still, further improvement appeared to occur
on the drug label. Nonvalidated drug treatment may not until 1 year, which confirms the data obtained from an
provide optimal benefit to the individual child and open trial with meloxicam (8). In the absence of a
usually does not generate new information for identify- placebo group, it remains speculative whether this fur-
ing the best use of the medication. ther improvement is the result of additional effects of
A placebo group, although desirable, was not the trial drug beyond the underlying trend of the disease
included due to ethical considerations. Ethics commit- to be self-limiting, a bias from natural fluctuations of the
tees and expert guidelines prefer the exclusive use of disease, or the variability in the assessment of the
active comparators instead of placebo in children, and disease by investigator and parent.
alternative trial designs are suggested (27). Some regu-
The therapeutic effect was clear in all 3 treatment
latory authorities, such as the US Food and Drug
groups, but the responses observed in this trial were
Administration and the European Medicines Agency,
higher than expected. Response to treatment was de-
refer to the systematic advantages of comparing investi-
fined according to the ACR pediatric 30 criteria. The
gational drugs with placebo during the evaluation of new
drugs (28,29). It is interesting to note that a placebo ACR pediatric 30 criteria were proposed with the intent
response rate of 29% (95% CI 24–34%) was obtained that they would be robust enough to cover all different
from a meta-analysis of previous placebo-controlled subtypes of JIA (10), but the performance of this criteria
trials in children with JIA (30), applying the ACR set in children with oligo-course JIA, who constituted
pediatric 30 criteria. The CIs of the response rates in this the majority of the patients enrolled in this trial, has
trial are clearly separate from those assessed in the never been formally tested. It is conceivable that the use
meta-analysis, showing indirectly the superiority of the of the ACR pediatric 30 definition in patients with
active treatments compared with placebo. All 3 treat- oligo-course disease, in whom the total number of joints
ment groups showed substantial improvement of the involved is low, may have led to an overestimation of the
MELOXICAM VERSUS NAPROXEN FOR JIA 571

percentage of change, leading to an unreasonably high comparable with the safety and efficacy of naproxen in
response rate. the treatment of oligo-course and poly-course JIA. The
Anecdotal information and common clinical benefit of the once-daily administration of meloxicam
practice suggest that higher doses of naproxen might oral suspension might represent an improvement in the
be needed in certain cases of JIA (31). However, from treatment of children with JIA.
published literature it is impossible to demonstrate that
a higher dosage of naproxen would be more effective
ACKNOWLEDGMENTS
than the registered dosage of 10 mg/kg/day divided into
2 daily doses. Indeed, 3 published trials of naproxen used We thank the following members of the Pediatric
the registered dosage of 10 mg/kg/day (32–34), with the Rheumatology International Trials Organization (PRINTO)
remaining 2 studies (35,36) using dosages up to 15–20 who enrolled children for the trial: C. Huemer, MD, H.
Schacherl, MD (Austria); C. Wouters, MD (Belgium); C.
mg/kg/day. A dosage of 5 mg/kg twice daily in children Barbier, MD, L. Gourmy, MD, C. Job-Deslandre, MD, J.
corresponds to an adult dose of 1,000 mg (37), the Terzic, MD, I. Lemelle, MD (France); H. Girschick, MD, G.
highest recommended daily dose. In the absence of any Horneff, MD, H. I. Huppertz, MD, R. Küster, MD, M.
other comparative data on the use of different doses of Metzler, MD (Germany); R. Di Toro II, MD, M. G. Alpigiani,
naproxen in JIA, we performed an exploratory subgroup MD (Italy); J. Anderson, MD, and P. Woo, MD (UK).
analysis within the naproxen-treated patient group in
this trial, based on the actual naproxen doses adminis- REFERENCES
tered to every child (data not shown). The administra-
1. Peterson LS, Mason T, Nelson AM, O’Fallon WM, Gabriel SE.
tion of higher doses of naproxen did not translate into Juvenile rheumatoid arthritis in Rochester, Minnesota 1960–1993:
better response rates. is the epidemiology changing? Arthritis Rheum 1996;39:1385–90.
The safety profile of meloxicam was largely com- 2. Ruperto N, Levinson JE, Ravelli A, Shear ES, Tague BL, Murray
K, et al. Longterm health outcomes and quality of life in American
parable with that of naproxen, including the frequency and Italian inception cohorts of patients with juvenile rheumatoid
and nature of GI adverse events. Although GI adverse arthritis. I. Outcome status. J Rheumatol 1997;24:945–51.
events would be expected side effects of NSAID use in 3. Hull RG, and the British Paediatric Rheumatology Group. Guide-
adults, the majority of such events in this trial were not lines for management of childhood arthritis. Rheumatology (Ox-
ford) 2001;40:1309–12.
considered by the investigators to be drug-related. This 4. Ilowite NT. Current treatment of juvenile rheumatoid arthritis.
could be attributable to the fact that many concomitant Pediatrics 2002;109:109–15.
diseases, especially infections in younger children, are 5. Distel M, Mueller C, Bluhmki E, Fries J. Safety of meloxicam: a
global analysis of clinical trials. Br J Rheumatol 1996;35 Suppl
accompanied by GI symptoms. No ulcerations, perfora- 1:68–77.
tions, or major bleeds from the upper GI tract were 6. Schoenfeld P. Gastrointestinal safety profile of meloxicam: a
reported, which supports data from literature indicating meta-analysis and systematic review of randomized controlled
trials. Am J Med 1999;107:48–54S.
that children are less likely than adults to experience
7. Layton D, Hughes K, Harris S, Shakir SA. Comparison of the
significant GI problems (4,38). incidence rates of selected gastrointestinal events reported for
Although a higher frequency of increases in the patients prescribed celecoxib and meloxicam in general practice in
serum bilirubin level was observed in the group receiving England using prescription-event monitoring (PEM) data. Rheu-
matology (Oxford) 2003;42:1332–41.
meloxicam 0.125 mg/kg compared with the other 2 8. Foeldvari I, Burgos-Vargas R, Thon A, Tuerck D. High response
treatment groups, this observation, in the absence of rate in the phase I/II study of meloxicam in juvenile rheumatoid
dose dependency, is not considered to affect the safety arthritis. J Rheumatol 2002;29:1079–83.
9. Petty RE, Southwood TR, Baum J, Bhettay E, Glass DN, Manners
assessment of meloxicam. Also, data from studies of P, et al. Revision of the proposed classification criteria for juvenile
meloxicam in adults did not reveal an increase in the idiopathic arthritides, Durban, 1997. J Rheumatol 1998;25:1991–4.
bilirubin level above the upper limit of normal, as was 10. Giannini EH, Ruperto N, Ravelli A, Lovell DJ, Felson DT,
observed in the current study. The observed higher Martini A. Preliminary definition of improvement in juvenile
arthritis. Arthritis Rheum 1997;40:1202–9.
incidence of musculoskeletal adverse events in the group 11. Ruperto N, Ravelli A, Falcini F, Lepore L, de Sanctis R, Zulian F,
receiving meloxicam 0.25 mg/kg was mainly attributable et al. Performance of the preliminary definition of improvement in
to an arthritis flare; however, because such an increased juvenile chronic arthritis patients treated with methotrexate. Ann
Rheum Dis 1998;57:38–41.
incidence of these adverse events was not detected in the 12. Albornoz MA. ACR formally adopts improvement criteria for
group receiving meloxicam 0.125 mg/kg, this observation juvenile arthritis (ACR pediatric 30). ACR News 2002;21:3.
was not considered relevant. 13. Cassidy JT, Levinson JE, Bass JC, Baum J, Brewer EJ Jr, Fink
CW, et al. A study of classification criteria for a diagnosis of
In conclusion, the short- and long-term safety juvenile rheumatoid arthritis. Arthritis Rheum 1986;29:274–81.
and efficacy of meloxicam oral suspension appear to be 14. Ruperto N, Giannini EH. Redundancy of conventional articular
572 RUPERTO ET AL

response variables used in juvenile chronic arthritis clinical trials. 26. Brown EG. Methods and pitfalls in searching drug safety data-
Ann Rheum Dis 1996;55:73–5. bases utilising the Medical Dictionary for Regulatory Activities
15. Singh G, Athreya BH, Fries JF, Goldsmith DP. Measurement of (MedDRA). Drug Saf 2003;26:145–58.
health status in children with juvenile rheumatoid arthritis. Arthri- 27. Committee on Drugs, American Academy of Pediatrics. Guide-
tis Rheum 1994;37:1761–9. lines for the ethical conduct of studies to evaluate drugs in
16. Ruperto N, Ravelli A, Pistorio A, Malattia C, Cavuto S, Gado- pediatric population. Pediatrics 1995;95:286–94.
West L, et al. Cross-cultural adaptation and psychometric evalu- 28. Food and Drug Administration. Regulations requiring manufac-
ation of the Childhood Health Assessment Questionnaire turers to assess the safety and effectiveness of new drugs and
(CHAQ) and the Child Health Questionnaire (CHQ) in 32 biologic products in pediatrics patients. Federal Register 1998;63:
countries: review of the general methodology. Clin Exp Rheuma- 66631–72.
tol 2001;19(4 Suppl 23):S1–9. 29. European Medicines Evaluation Agency. Report on the experts
17. Huemer C, Ruperto N, Huemer M, Sailer-Hoeck M, Kaulfersch round table on the difficulties related to the use of new medicinal
W, Schwarz R, et al. The Austrian version of the Childhood Health products in children held on 18 December 1997. EMEA, 1998.
Assessment Questionnaire (CHAQ) and the Child Health Ques- (27164/98 Rev. 1.)
tionnaire (CHQ). Clin Exp Rheumatol 2001;19:(4 Suppl 23): 30. Ruperto N, Pistorio A, Martini A, Sigmund R, Hanft G, Simianer
S15–9. S. A meta-analysis to estimate the “real” placebo effect in juvenile
18. Joos R, Ruperto N, Wouters C, Boven K, Raat H, Landgraf JM, rheumatoid arthritis (JRA) trials [abstract]. Arthritis Rheum
et al. The Belgian-Flemish version of the Childhood Health 2003;48 Suppl 9:S90.
Assessment Questionnaire (CHAQ) and the Child Health Ques- 31. Cassidy JT, Petty RE. Juvenile rheumatoid arthritis. In: Cassidy
tionnaire (CHQ). Clin Exp Rheumatol 2001;19(4 Suppl 23): JT, Petty RE, editors. Textbook of pediatric rheumatology. 4th ed.
S20–4. Philadelphia: WB Saunders; 2001. p. 218–322.
19. Pouchot J, Ruperto N, Lemelle I, Sommelet D, Grouteau E, David 32. Kvien TK, Hoyeraal HM, Sandstad B. Naproxen and acetylsali-
L, et al. The French version of the Childhood Health Assessment cylic acid in the treatment of pauciarticular and polyarticular
Questionnaire (CHAQ) and the Child Health Questionnaire juvenile rheumatoid arthritis: assessment of tolerance and efficacy
(CHQ). Clin Exp Rheumatol 2001;19:S60–5. in a single-centre 24-week double-blind parallel study. Scand
20. Foeldvari I, Ruperto N, Dressler F, Hafner R, Kuster RM, J Rheumatol 1984;13:342–50.
Michels H, et al. The German version of the Childhood Health 33. Leak AM, Richter MR, Clemens LE, Hall MA, Ansell BM. A
Assessment Questionnaire (CHAQ) and the Child Health Ques- crossover study of naproxen, diclofenac and tolmetin in seroneg-
tionnaire (CHQ). Clin Exp Rheumatol 2001;19(4 Suppl 23):S71–5. ative juvenile chronic arthritis. Clin Exp Rheumatol 1988;6:
21. Ruperto N, Ravelli A, Pistorio A, Malattia C, Viola S, Cavuto S, 157–60.
et al. The Italian version of the Childhood Health Assessment 34. Makela AL. Naproxen in the treatment of juvenile rheumatoid
Questionnaire (CHAQ) and the Child Health Questionnaire arthritis: metabolism, safety and efficacy. Scand J Rheumatol
(CHQ). Clin Exp Rheumatol 2001;19(4 Suppl 23):S91–5. 1977;6:193–205.
22. Nikishina O, Ruperto N, Kuzmina N, Shelepina T, Illarionova O, 35. Williams PL, Ansell BM, Bell A, Cain AR, Chamberlain MA,
Salougina S, et al. The Russian version of the Childhood Health Clarke AK, et al. Multicentre study of piroxicam versus naproxen
Assessment Questionnaire (CHAQ) and the Child Health Ques- in juvenile chronic arthritis, with special reference to problem
tionnaire (CHQ). Clin Exp Rheumatol 2001;19(4 Suppl 23): areas in clinical trials of nonsteroidal anti-inflammatory drugs in
S131–5. childhood. Br J Rheumatol 1986;25:67–71.
23. Nugent J, Ruperto N, Grainger J, Machado C, Sawhney S, 36. Laxer RM, Silverman ED, St-Cyr C, Tran MT, Lingam G. A
Baildam E, et al. The British version of the Childhood Health six-month open safety assessment of a naproxen suspension for-
Assessment Questionnaire (CHAQ) and the Child Health Ques- mulation in the therapy of juvenile rheumatoid arthritis. Clin Ther
tionnaire (CHQ). Clin Exp Rheumatol 2001;19:S163–7. 1988;10:381–7.
24. McGrath PA, Seifert CE, Speechley KN, Booth JC, Stitt L, Gibson 37. Physicians’ Desk Reference. Montvale (NJ): Medical Economics;
MC. A new analogue scale for assessing children’s pain: an initial 2001.
validation study. Pain 1996;64:435–43. 38. Keenan GF, Giannini EH, Athreya BH. Clinically significant
25. Moher D, Schulz KF, Altman DG. The CONSORT statement: gastropathy associated with nonsteroidal antiinflammatory drug
revised recommendations for improving the quality of reports of use in children with juvenile rheumatoid arthritis. J Rheumatol
parallel-group randomised trials. Lancet 2001;357:1191–4. 1995;22:1149–51.

También podría gustarte