You are on page 1of 8

Current Medical Research & Opinion Vol. 30, No.

2, 2014, 279–286

0300-7995 Article RT-0338.R1/855631


doi:10.1185/03007995.2013.855631 All rights reserved: reproduction in whole or part not permitted

Original article
A randomized saline-controlled trial of NASHA
hyaluronic acid for knee osteoarthritis

d
Curr Med Res Opin Downloaded from informahealthcare.com by Queen's University on 01/01/15

ite

n
Nigel K. Arden Abstract

na ow io
d,
py us is Lim
NIHR Musculoskeletal Biomedical Research Unit,

se oa
so d ut
University of Oxford, UK Objective:

l u nl
NASHA hyaluronic acid is administered as a single intra-articular injection to treat the symptoms of

b
Christian Åkermark

K
osteoarthritis (OA). In a previous trial, post-hoc analysis indicated that NASHA provides significantly

rp c i
fo ers tr
Sport Med, Stockholm, Sweden

aU
greater pain relief than saline in patients with OA confined to the study knee. We aimed to evaluate the

er n
Mats Andersson safety and efficacy of NASHA in patients with unilateral knee OA.

a
rm
Q-Med AB, Uppsala, Sweden

D
Research design and methods:
Martin G. Todman le is al
rin ted m fo

Smith & Nephew UK Ltd, Research Centre, York


This was a randomized, double-blind, saline-controlled trial. All patients had knee OA confirmed by
ng or i
For personal use only.

American College of Rheumatology criteria and a WOMAC pain score of 7–17 in the study knee, but no
si th rc

Science Park, York, UK


d p ibi om In

co ed
pain in the previous 3 months in the non-study knee. Treatment comprised a single intra-articular injection
Roy D. Altman
t a . Au e

of NASHA or saline control. The follow-up period was 6 weeks.


14

UCLA David Geffen School of Medicine, LA, USA


20

Clinical trial registration:


Address for correspondence: ClinicalTrials.gov NCT01806207.
la d le ©

Prof. Nigel Arden, NIHR Musculoskeletal Biomedical


C

Research Unit, Nuffield Orthopaedic Centre, University


Main outcome measures:
ht

ew p r

of Oxford, Windmill Road, OX3 7LD, UK.


vi e o

nigel.arden@ndorms.ox.ac.uk
The primary efficacy endpoint was the responder rate, defined as the percentage of patients with 40%
an h
rig

improvement from baseline in WOMAC pain score and an absolute improvement of 5 points.
ro
sp ize a

Keywords:
py

Results:
di hor r S
y, u s

Knee – NASHA hyaluronic acid gel – Osteoarthritis –


A total of 218 patients received study treatment (NASHA: 108, saline: 110). In the main intention-to-treat
Co

Saline control
(ITT) analysis, no statistically significant difference in responder rate was found between the two groups at
au fo

Accepted: 10 October 2013; published online: 4 November 2013 6 weeks (NASHA: 30.6%; saline: 26.4%). A post-hoc subgroup analysis of patients without clinical effusion
Citation: Curr Med Res Opin 2014; 30:279–86
in the study knee at baseline showed a significantly higher 6 week responder rate with NASHA than with
Un t
No

saline: 40.6% versus 19.7% (p ¼ 0.0084). A total of 68 adverse events were reported among 44 patients in
t

the NASHA group, compared with 69 adverse events among 44 patients in the saline group. The main
weakness of the study was the short, 6 week follow-up duration. In addition, image guidance was not used
to ensure injection as intended into the intra-articular space.

Conclusions:
Single-injection NASHA was well tolerated and, although there was no significant benefit versus saline
control in the primary analysis, post-hoc analysis showed a statistically significant improvement in pain relief
at 6 weeks among patients without clinical effusion at baseline.

Introduction
Intra-articular (IA) injection of hyaluronic acid (HA) is a well established
means of treating pain in patients with osteoarthritis (OA) of the knee.
According to the American College of Rheumatology guidelines, such treat-
ment is indicated for patients who have not responded to a program of non-

! 2014 Informa UK Ltd www.cmrojournal.com NASHA hyaluronic acid for knee osteoarthritis Arden et al. 279
Current Medical Research & Opinion Volume 30, Number 2 February 2014

pharmacologic therapy and simple analgesics1, while the 2000, and the International Conference on
2008 and 2010 OARSI guidelines conclude that injections Harmonization Guidelines for Good Clinical Practice.
of IA hyaluronate may be useful in patients with knee All patients provided signed, informed consent upon
or hip OA2,3. enrolment for the study. The results are presented in line
The risks associated with the most commonly used with Consolidated Standards of Reporting Trials
medications for OA pain management – hepatic toxicity (CONSORT) guidelines18. Independent Ethics
with acetaminophen4, gastrointestinal safety concerns Committee (IEC)/Institutional Review Board (IRB)
with non-steroidal anti-inflammatory drugs (NSAIDs)5,6 approval was obtained at each participating site in the
and cardiovascular safety concerns with cyclooxygenase-2 UK and Germany. In Sweden, the Independent Ethics
(COX-2) inhibitors7 – have received increasing attention Committee at Lunds University served as co-ordinating
in recent years3. This has prompted renewed interest in IEC for all sites. In Germany, the protocol was approved
alternative treatment options, including viscosupplemen- by ‘Bundesamt für Strahlenschutz’ prior to commencement
tation with IA HA. A number of HA products are avail- of the study. The study is registered at ClinicalTrials.gov
Curr Med Res Opin Downloaded from informahealthcare.com by Queen's University on 01/01/15

able for the treatment of knee OA pain, with dosing (NCT01806207).


regimens ranging from one to five weekly injections. Normally active men and women aged 450 years with
NASHA hyaluronic acid has a unique cross-linked the ability to walk 50 metres unaided and with knee pain
molecular structure8 that provides a prolonged IA resi- meeting the American College of Rheumatology criteria
dence time (half-life of 4 weeks)8–10. This allows adminis- for the diagnosis of OA1 were eligible for inclusion,
tration of NASHA as a single injection, providing benefits provided that OA was confirmed in the study knee radio-
such as improved patient convenience and potential cost graphically (Kellgren–Lawrence grades II or III) and by a
reductions through fewer clinic visits. The cross-linking Western Ontario and McMaster Universities
(stabilization) process used to produce NASHA introduces Osteoarthritis Index (WOMAC) pain score of 7–17 at
only minimal molecular changes, thereby preserving the the baseline visit. Key exclusion criteria were: pain
inherent biocompatibility of HA. This, together with the during the previous 3 months in the non-study knee, radio-
For personal use only.

sourcing of HA from bacterial fermentation as opposed to graphically verified OA of the non-study knee (Kellgren–
animal tissue, may reduce the risk of allergic reactions Lawrence grade4I), OA or clinically significant pain from
and other adverse events. Clinical studies of NASHA any part of the musculoskeletal system other than the study
administered as a single injection have demonstrated knee, previous IA steroid injection into the study knee
that it provides significant improvement in the symptoms within the last 3 months, previous IA HA injection into
of OA compared to baseline, both in the knee11–14 and the study knee within the last 9 months, use of systemic
the hip15,16. steroids (excluding inhaled steroids) within the last
A previous 6 month, randomized, saline-controlled 3 months, and arthroscopy or other surgical procedures
study of NASHA for the treatment of knee OA pain13 in the study knee within the last 12 months. The first
included patients with late-stage OA (Kellgren– patient was enrolled on August 15, 2003 and the last
Lawrence grade IV), OA or clinically significant pain patient completed the 6 week visit on April 22, 2004.
from joints other than the knee (poly-articular pain), Study participants were randomized 1:1 to receive a
and clinical effusion at baseline. Improved efficacy was single IA injection in the study knee of either NASHA
observed in patients with OA confined to the study (Durolane* 60 mg in 3 ml) or saline (3 ml phosphate-
knee. Other studies have also shown that pain from buffered saline, pH 7). Investigators with experience of
joints other than the one being treated can mask the treat- administering intra-articular treatment administered
ment effect of IA HA injection13,17. We designed a 6 NASHA or saline into the synovial space; lateral mid-
week, saline-controlled study to investigate the safety patellar, lateral upper-patellar or medial injection tech-
and efficacy of NASHA in patients with mild–moderate nique was advised. Image guidance was not used. Rescue
structural OA confined to the study knee. medication with acetaminophen up to 4 g per day was
allowed throughout the study except during the 48 hour
period preceding each study visit, and all such medication
was recorded. NSAIDs, including topical agents for the
Patients and methods knee, were not permitted during the study period or
Study design and participants during the week before administration of study treatment.
Follow-up visits were scheduled at 2, 4 and 6 weeks
This was a saline-controlled, double-blind, multicentre post-treatment. Randomization codes were computer gen-
study of NASHA, with a 6 week follow-up period. erated by the contract research organization running the
Patients were enrolled at 13 sites in Sweden, Germany study, with stratification by site. Patients’ initials were
and the UK. The study was conducted in accordance
*
with the Declaration of Helsinki 1975, as revised in Durolane is a registered trade name of Q-Med AB, Uppsala, Sweden.

280 NASHA hyaluronic acid for knee osteoarthritis Arden et al. www.cmrojournal.com ! 2014 Informa UK Ltd
Current Medical Research & Opinion Volume 30, Number 2 February 2014

written on the outside of treatment code envelopes after Statistical methods


randomization and, to determine which product to admin-
ister (NASHA or saline), the treating investigator opened The intention-to-treat (ITT) and safety populations con-
each patient’s envelope before treatment. Patients were sisted of all patients who were randomized and treated. The
enrolled by the evaluating investigator, and the treating primary population for all efficacy analyses was ITT, with
investigator administered the study treatment (any joint supportive analyses performed on the per-protocol (PP)
effusion was removed prior to NASHA or saline injec- population (patients in the ITT population who com-
tion). Patients were not informed which treatment they pleted all scheduled efficacy evaluations without major
received and they were blinded to the injection with the protocol violations).
use of surgical drapes. The evaluating investigator, who Improvements from baseline in WOMAC pain score,
was blinded to the treatment given (no access to the treat- WOMAC stiffness score and WOMAC physical function
ment code envelopes and no involvement in the treatment score and total number of rescue medication tablets were
procedure), conducted the post-treatment assessments. compared between treatments using a two-sample t-test.
WOMAC pain responder rates were compared between
Curr Med Res Opin Downloaded from informahealthcare.com by Queen's University on 01/01/15

The study was terminated when the sample size (see


Statistical methods below) was fulfilled and the last patient treatments using chi-squared tests. Treatment-group dif-
enrolled had completed the 6 week follow-up visit. ferences in the percentage of patients reporting adverse
Adverse events were reported at all study visits and events were assessed with Fisher’s exact test. Within-
classified according to World Health Organization patient analysis of WOMAC pain responder rate over
(WHO) preferred terms and body systems. All adverse time was performed using McNemar’s test.
events occurring on or after the day of injection were con- As a post-hoc analysis, treatment-group differences in
sidered as treatment emergent. WOMAC pain responder rates were assessed within
patient subgroups defined by the variables shown in
Table 1. The continuous variables were dichotomized as
follows: age (below/at least 60), body mass index (BMI)
For personal use only.

Outcomes
(below/at least 27), duration of diagnosed knee OA
The primary endpoint was the WOMAC pain responder (below/at least 2 years) and WOMAC pain score at base-
rate, defined as the percentage of patients achieving line (below/at least 10 units).
at least 40% reduction from baseline in WOMAC Sample size calculation was based on the assumption
pain score, together with an absolute improvement of that the proportion of patients responding to treatment
at least 5 points19. A Likert version of the WOMAC would be 45% with NASHA and 25% with saline. Using
pain score was used (range 0–20). Secondary outcome a two-sided 5% significance level and a power of 80%, a
measures included Likert versions of the WOMAC sample size of 172 patients (86 per group) was calculated.
stiffness score (range 0–8), WOMAC physical function To compensate for patients lost to follow-up, the total
score (range 0–68) and patient assessment of global sample size was increased to 218. All statistical analyses
status (range 1–5). were performed using SAS software.

Table 1. Baseline characteristics of the study population.

Variable NASHA (n ¼ 108) Saline (n ¼ 110) p-Value

Age in years, median (range) 64.5 (29–84) 60.9 (30–86) 0.0403


Female:male, n (%) 59:49 (55:45) 51:59 (46:54) 0.2223
BMI female, median (range) 26.4 (20.4–39.7) 26.9 (20.1–41.0) 0.7198
BMI male, median (range) 28.2 (20.3–37.8) 28.1 (22.2–39.5) 0.7984
Duration of OA in years, median (range) 2.2 (0–21.2) 3.1 (0–44.1) 0.0794
Kellgren–Lawrence score*
II, n (%) 33 (30.6) 40 (36.4) 0.3636
III, n (%) 75 (69.4) 70 (63.6)
WOMAC pain score, median (range)* 10.0 (7–17) 9.5 (7–15) 0.6284
Clinical effusion present, * n (%) 39 (36.1) 44 (40.0) 0.5544
NSAIDs or analgesics for OA in the last 3 months, n (%) 45 (41.7) 45 (40.9) 0.9096
Previous treatment with IA steroids in the study knee, n (%) 21 (19.4) 32 (29.1) 0.0969
Previous treatment with IA HA in the study knee, n (%) 8 (7.4) 16 (14.5) 0.0923
Previous knee surgery in the study knee, n (%) 46 (42.6) 45 (40.9) 0.8010

p-values: chi-squared test (categorical variables) and Wilcoxon rank-sum test (other variables).
*Relating to the study (signal) knee.
BMI, body mass index; HA, hyaluronic acid; IA, intra-articular; NSAID, non-steroidal anti-inflammatory drug; OA, osteoarthritis;
WOMAC, Western Ontario and McMaster Universities Osteoarthritis Index.

! 2014 Informa UK Ltd www.cmrojournal.com NASHA hyaluronic acid for knee osteoarthritis Arden et al. 281
Current Medical Research & Opinion Volume 30, Number 2 February 2014

All efficacy analyses were based on a last observation A significant improvement in responder rate was observed
carried forward (LOCF) approach. The p-values obtained in the NASHA group between 2 and 6 weeks (30.6% vs
for secondary and post-hoc analyses were not adjusted 19.4%, p ¼ 0.0143). There was no significant improve-
for multiple testing. A result was considered statistically ment in the saline group over this period (26.4% vs
significant when the statistical test yielded a two-tailed 25.5%, p ¼ 0.8273).
p-value of 0.05 or less. For all secondary efficacy endpoints (WOMAC pain
score, WOMAC stiffness score, WOMAC physical func-
tion score and patient assessment of global status),
improvements from baseline were numerically larger in
Results the NASHA group compared with saline, but the differ-
A total of 218 patients were randomized to receive study ences did not reach statistical significance. Data for the
treatment (NASHA: 108, saline: 110). There were five three WOMAC subscales are shown in Table 3. In the PP
discontinuations during the 6 week study period, all from population, a statistically significantly higher percentage
Curr Med Res Opin Downloaded from informahealthcare.com by Queen's University on 01/01/15

the NASHA group (Figure 1). Demographic and other of patients receiving NASHA showed improvement in
baseline characteristics of the patients are presented in global status of at least one point versus baseline at 4
Table 1. A statistically significant difference in age was weeks (NASHA: 44.6% [37/83]; saline: 26.4% [24/91];
seen between groups but, within the context of OA as a p ¼ 0.0119) and 6 weeks (NASHA: 37.3% [31/83];
chronic condition, this was not considered to be clinically saline: 20.9% [19/91]; p ¼ 0.0165). No other statistically
significant. The volume of effusion removed immediately significant between-group differences were observed in
before administration of NASHA or saline was higher in secondary outcomes. Mean consumption of rescue medi-
the saline group (median 12.5 ml [range 1–50 ml], versus cation during the follow-up phase was 34.2 tablets in the
5.0 ml [1–20 ml] in the NASHA group), but the percent- NASHA group and 26.1 tablets in the saline group
age of patients with effusion was similar in the two groups. (p ¼ 0.3057).
For personal use only.

There was no statistically significant difference between The post-hoc subgroup analysis showed statistically
the two study groups in the primary outcome measure at significantly higher WOMAC pain responder rates at
any time point (both ITT and PP analyses; Table 2). 4 weeks (p ¼ 0.0292) and 6 weeks (p ¼ 0.0084) with

Randomized (n=218)

Allocated to NASHA (n=108) Allocated to saline (n=110)


Allocation Received allocated intervention (n=108) Received allocated intervention (n=110)

Premature discontinuation (n=5*) Premature discontinuation (n=0)


Consent withdrawn (n=2)
Unrelated serious adverse event (n=1)
Lost to follow-up (lost interest) (n=1)
Lack of effect (n=1)

Follow-up Major protocol deviation (n=25 in 22 subjects) Major protocol deviation (n=19 in 19 subjects)
Assessment of masking by treating investigator (n=10) Assessment of masking by treating investigator (n=10)
Randomization envelope signed by both investigators (n=1) Treatment with the other study product (n=1)
Treatment with the other study product (n=1) Prohibited medication (NSAID/analgesic/anticoagulant) (n=3)
Prohibited medication (NSAID/analgesic/anticoagulant) (n=3) Rescue medication within 48 hours prior to visit 7 (n=5)
Rescue medication within 48 hours prior to visit 7 (n=8)
WOMAC pain score missing on at least one visit (n=2)

ITT population (n=108) ITT population (n=110)


Analysis PP population (n=83) PP population (n=91)
Without clinical effusion ITT population (n=69) Without clinical effusion ITT population (n=66)

*Two patients who discontinued prematurely also had major protocol deviations. ITT, intention to treat; NSAID, non-steroidal
anti-inflammatory drug; PP, per protocol.

Figure 1. Study population.

282 NASHA hyaluronic acid for knee osteoarthritis Arden et al. www.cmrojournal.com ! 2014 Informa UK Ltd
Current Medical Research & Opinion Volume 30, Number 2 February 2014

Table 2. WOMAC pain responder rates in study populations.

Time post-treatment Analysis NASHA responder Saline responder p-Value


population rate, % (n/N) rate, % (n/N)

2 weeks ITT 19.4 (21/108) 25.5 (28/110) 0.29


PP 19.3 (16/83) 19.8 (18/91) 0.93
4 weeks ITT 26.9 (29/108) 26.4 (29/110) 0.94
PP 28.9 (24/83) 24.2 (22/91) 0.48
6 weeks ITT 30.6 (33/108) 26.4 (29/110) 0.49
PP 34.9 (29/83) 27.5 (25/91) 0.29

ITT, intention to treat; PP, per protocol.

Table 3. Analysis of WOMAC subscale scores at baseline and 6 weeks (ITT population). Data shown are
mean  standard deviation.
Curr Med Res Opin Downloaded from informahealthcare.com by Queen's University on 01/01/15

NASHA Saline p-Value

WOMAC pain score


Baseline 9.98  2.09 9.83  2.03 –
6 weeks 7.42  3.80 7.37  3.43 –
Change from baseline at 6 weeks 2.56  3.46 2.45  3.06 0.86
WOMAC stiffness score
Baseline 4.03  1.39 3.90  1.34 –
6 weeks 3.09  1.71 3.08  1.67 –
Change from baseline at 6 weeks 0.94  1.62 0.82  1.58 0.71
WOMAC physical function score
Baseline 30.42  10.65 30.19  10.24 –
For personal use only.

6 weeks 24.16  13.62 24.60  12.66 –


Change from baseline at 6 weeks 6.26  10.81 5.59  10.58 0.77

ITT, intention to treat; WOMAC, Western Ontario and McMaster Universities Osteoarthritis Index.

NASHA than with saline among patients without clinical 45


WOMAC pain responder rate (%)

NASHA Saline
effusion in the study knee at baseline (Figure 2). Only 40
minor differences between the study groups were evident 35
in the other subgroups. 30
A total of 68 adverse events were reported among 25
44 patients in the NASHA group, compared with 20
69 adverse events among 44 patients in the saline group. 15
There were no treatment-related serious adverse events. 10
The percentage of patients reporting treatment-related
5
adverse events was higher in the NASHA group than
0
in the saline group: 15.7% (17 patients, 18 adverse Week 2 Week 4 Week 6
events) versus 5.5% (6 patients, 6 adverse events) NASHA 23.2 34.8 40.6
(p ¼ 0.0154). The nature of treatment-related adverse Saline 22.7 18.2 19.7
p-value 0.9492 0.0292 0.0084
events was as anticipated in both groups – primarily
knee pain or knee swelling. Specific treatment-related
Figure 2. WOMAC pain responder rates among patients without clinical
adverse events (defined by preferred terms) occurred effusion in the study knee (intention to treat population).
with similar frequency in the two groups, except for
‘injection site pain’ which was reported in seven patients
in the NASHA group and none in the saline group
(p ¼ 0.0066). subgroup analysis suggested that NASHA was superior in
patients without clinical knee effusion at baseline.
Overall, NASHA injections were well tolerated with no
reported treatment-related serious adverse events. There
Discussion was a higher rate of treatment-related adverse events in the
Although the current 6-week study of patients with knee NASHA group, although these were generally categorized
OA failed to demonstrate that NASHA provides a statis- as mild to moderate in severity, transient and resolved
tically significant benefit over saline injection, post-hoc without issue.

! 2014 Informa UK Ltd www.cmrojournal.com NASHA hyaluronic acid for knee osteoarthritis Arden et al. 283
Current Medical Research & Opinion Volume 30, Number 2 February 2014

Development of therapies often involves a process of anti-inflammatory effects32, it would be of interest to


clinical investigation to identify patients with the most explore further this possible mechanism of action of HA
favourable risk–benefit profile. A previous, randomized in the reduction of OA pain (possibly relating to
double-blinded saline-controlled clinical study identified synovitis).
poly-articular pain as a confounding factor in the inter- The main weakness in the reported study was the short,
pretability of HA clinical data, hence the exclusion 6 week follow-up duration. Significant improvement in
of patients with poly-articular pain from this study13. the NASHA responder rate was observed between 2 and
Other clinical factors that may predict effectiveness of 6 weeks. This is in line with a previous study of NASHA
HA injections are of great interest, and the current study for knee OA, where maximum response was seen at
suggests that clinical effusion fits this category. Previous 6 weeks13. However, assessments in that study were rela-
studies have shown that viscosupplementation is likely to tively infrequent (2, 6, 13 and 26 weeks) meaning that the
have reduced effectiveness in the presence of effusion20,21. treatment effect could potentially have peaked later than
Consequently, clinical studies investigating HA therapies 6 weeks. The maximal effect of HA has been reported to
in knee OA typically exclude patients with clinical effu- occur at around 8 weeks33, and it is possible that maximal
Curr Med Res Opin Downloaded from informahealthcare.com by Queen's University on 01/01/15

sions22–24. While care must be taken when conducting and response is delayed in a patient population that included
interpreting post-hoc subgroup analysis, interpretation is clinical effusion at baseline. Thus, a study with longer
aided when the number of sub-populations analyzed is low follow-up than 6 weeks would be valuable to provide
and based on sound clinical observations. insight into the duration of effect of NASHA in patients
To investigate further the effects of excluding patients with OA confined to the study knee. In addition, the
with poly-articular pain and clinical effusion, we per- absence of image guidance in this study introduces the
formed a pooled analysis of patients from this study and possibility that not all of the fluid was injected precisely
the previously published randomized controlled trial of as intended into the intra-articular space. The potential
NASHA13. A total of 284 patients meeting these criteria for variations in injection technique, increased by the mul-
were treated, 151 with NASHA and 133 with saline. The ticentre design of the study, may have hampered detection
For personal use only.

WOMAC pain responder rate at 6 weeks was statistically of statistically significant between-group differences.
significantly higher with NASHA than with saline (43.0% However, block randomization ensured that both treat-
vs 24.8%, p ¼ 0.0013). These data suggest that future stu- ment groups were affected equally and the inclusion of
dies of NASHA among patients without clinical effusion multiple centres increased the generalizability of the
could provide improved insight into treatment efficacy. results. Key strengths of this study are the randomized con-
The majority of knee OA patients exhibit some form of trolled design, and the provision of insight into the OA
effusion. In one magnetic resonance imaging (MRI) study patient population likely to respond to NASHA (i.e.
of 111 OA patients, 73% exhibited effusion on contrast patients with clinically active inflammation in the
enhanced images; this figure dropped to 38% for moderate knee as evidenced by overt clinical effusion are not good
to severe effusions and only 7% for the most severe grading candidates for HA therapy).
of effusion. Synovitis was found to occur in 96% of knees
with effusion and 70% of knees without effusion25. In this
respect it is notable that synovitis has been linked to pain
in OA26–29 and is often present in the absence of effusion. Conclusion
A smaller study reported a higher rate of MRI-detected We found NASHA to be clinically effective and well tol-
effusion, 93–96% in 30 OA patients. Successful synovial erated in an appropriately selected patient population.
joint volume measurement by arthrocentesis was achieved Post-hoc subgroup analysis in patients without clinical
in 60% of patients and was moderately correlated with effusions at baseline demonstrated that NASHA was stat-
MRI detection of effusion30. These studies indicate that istically and clinically significantly superior to saline at
joint effusion is a common finding in OA patients, that 6 weeks post-treatment. Further randomized controlled
some patients with an effusion detectable on MRI do not trials are required to confirm this finding and to improve
have successful arthrocentesis, and that more severe our understanding of the effects of clinical and sub-clinical
effusion detected by MRI correlates with higher joint effusion on IA therapies.
volume measurements (by arthrocentesis). It is not clear
what relationship clinical evidence of effusion has to MRI
findings or what relationship joint effusion has to pain
beyond the most serious cases of joint distension. The pres- Transparency
ence of clinical effusion may signify a more pronounced Declaration of funding
inflammatory process with the potential for increased This clinical trial was supported by Q-Med AB, Uppsala, Sweden
degradation of HA and loss of responsiveness to such ther- (study design, data collection, data analysis). Early versions of the
apy31. As IA HA therapy has been proposed to exhibit manuscript were supported by Q-Med AB and Smith & Nephew

284 NASHA hyaluronic acid for knee osteoarthritis Arden et al. www.cmrojournal.com ! 2014 Informa UK Ltd
Current Medical Research & Opinion Volume 30, Number 2 February 2014

UK Ltd through a Medical Writer (Ken Sutor whilst at 10. Edsman K, Hjelm R, Larkner H, et al. Intra-articular duration of
Fishawack Communications, 100–102 King Street, Knutsford, DurolaneTM after single injection into the rabbit knee. Cartilage
UK, WA16 6HQ). 2011;2:384-8
11. Akermark C, Adalberth T, Ericsater J, et al. Patient-perceived efficacy of non-
animal stabilized hyaluronic acid in the treatment of osteoarthritis of the knee.
Declaration of financial/other relationships
Osteoarthritis Cartilage 2004;12:S147
N.K.A. is a paid consultant for Q-Med AB and Smith & Nephew
12. Akermark C, Berg P, Bjorkman A, et al. Non-animal stabilised hyaluronic acid
Inc.; C.Å. has disclosed that he has no significant relationships in the treatment of osteoarthritis of the knee – a tolerability study. Clin Drug
with or financial interests in any commercial companies related Invest 2002;22:157-66
to this study or article; R.D.A. is a Ferring consultant, speaker, 13. Altman RD, Akermark C, Beaulieu AD, et al. Efficacy and safety of a single
Abbott consultant, Novartis consultant, Astra Zeneca speaker, intra-articular injection of non-animal stabilized hyaluronic acid (NASHA) in
Rotta consultant, Johnson & Johnson consultant, Novozyme patients with osteoarthritis of the knee. Osteoarthritis Cartilage 2004;12:
consultant, Covidien consultant and Lilly consultant; at the 642-9
time of the study M.A. was a full time employee of Q-Med AB 14. Krocker D, Matziolis G, Tuischer J, et al. Reduction of arthrosis associated
and M.G.T. was a full-time employee of Smith & Nephew knee pain through a single intra-articular injection of synthetic hyaluronic
acid. Z Rheumatol 2006;65:327-31
UK Ltd.
Curr Med Res Opin Downloaded from informahealthcare.com by Queen's University on 01/01/15

15. Berg P, Olsson U. Intra-articular injection of non-animal stabilised hyaluronic


CMRO peer reviewers may have received honoraria for
acid (NASHA) for osteoarthritis of the hip: a pilot study. Clin Exp Rheumatol
their review work. The peer reviewers on this manuscript have 2004;22:300-6
disclosed that they have no relevant financial relationships. 16. Conrozier T, Couris CM, Mathieu P, et al. Safety, efficacy and predictive
factors of efficacy of a single intra-articular injection of non-animal-stabi-
Acknowledgements lized-hyaluronic-acid in the hip joint: results of a standardized follow-up of
The Durolane Study 2 group recruited patients in Sweden: Jan patients treated for hip osteoarthritis in daily practice. Arch Orthop Trauma
Ericsäter, Malmö; Torsten Adalberth, Malmö; Johan Isacson, Surg 2009;129:843-8
Stockholm; Christian Åkermark, Stockholm; Anders 17. O’Malley KJ, Suarez-Almazor M, Aniol J, et al. Joint-specific multidimensional
Björkman, Uppsala; Per-Erik Melberg, Göteborg; UK: Nigel assessment of pain (J-MAP): factor structure, reliability, validity, and respon-
Arden, Southampton; John Dickson, Middlesbrough; Terry siveness in patients with knee osteoarthritis. J Rheumatol 2003;30:534-43
18. Schulz KF, Altman DG, Moher D. CONSORT 2010 statement: updated guide-
For personal use only.

Daymond, Sunderland; James Richardson, Oswestry; David


lines for reporting parallel group randomised trials. PLoS Med
Scott, London; and Germany: Jürgen Krämer, Bochum; Tobias 2010;7:e1000251
Schlegel, Mülheim/Ruhr (Nigel Arden was the coordinating 19. Dougados M, Leclaire P, van der Heijde D, et al. Response criteria for
investigator). Thanks to Ulf Olsson (Q-Med, AB) who provided clinical trials on osteoarthritis of the knee and hip: a report of the
critical insight during the writing of this manuscript. Osteoarthritis Research Society International Standing Committee for
Clinical Trials response criteria initiative. Osteoarthritis Cartilage
2000;8:395-403
20. Conrozier T, Mathieu P, Schott AM, et al. Factors predicting long-term efficacy
of Hylan GF-20 viscosupplementation in knee osteoarthritis. Joint Bone Spine
References 2003;70:128-33
1. Hochberg MC, Altman RD, April KT, et al. American College of Rheumatology 21. Namiki O, Toyoshima H, Morisaki N. Therapeutic effect of intra-articular
2012 recommendations for the use of nonpharmacologic and pharmacologic injection of high molecular weight hyaluronic acid on osteoarthritis of the
therapies in osteoarthritis of the hand, hip, and knee. Arthritis Care Res knee. Int J Clin Pharmacol Ther Toxicol 1982;20:501-7
2012;64:465-74 22. Berenbaum F, Grifka J, Cazzaniga S, et al. A randomised, double-blind,
2. Zhang W, Moskowitz RW, Nuki G, et al. OARSI recommendations for the controlled trial comparing two intra-articular hyaluronic acid preparations
management of hip and knee osteoarthritis, Part II: OARSI evidence- differing by their molecular weight in symptomatic knee osteoarthritis. Ann
based, expert consensus guidelines. Osteoarthritis Cartilage 2008;16: Rheum Dis 2012;71:1454-60
137-62 23. Chevalier X, Jerosch J, Goupille P, et al. Single, intra-articular treatment with
3. Zhang W, Nuki G, Moskowitz RW, et al. OARSI recommendations for the 6 ml hylan G-F 20 in patients with symptomatic primary osteoarthritis of the
management of hip and knee osteoarthritis: part III: Changes in evidence knee: a randomised, multicentre, double-blind, placebo controlled trial. Ann
following systematic cumulative update of research published through Rheum Dis 2010;69:113-19
January 2009. Osteoarthritis Cartilage 2010;18:476-99 24. Strand V, Baraf HS, Lavin PT, et al. A multicenter, randomized controlled
4. Schilling A, Corey R, Leonard M, et al. Acetaminophen: old drug, new warn- trial comparing a single intra-articular injection of Gel-200, a new cross-
ings. Cleve Clin J Med 2010;77:19-27 linked formulation of hyaluronic acid, to phosphate buffered saline for
5. Jones R, Rubin G, Berenbaum F, et al. Gastrointestinal and cardiovascular treatment of osteoarthritis of the knee. Osteoarthritis Cartilage 2012;20:
risks of nonsteroidal anti-inflammatory drugs. Am J Med 2008;121: 350-6
464-74 25. Roemer FW, Kassim Javaid M, Guermazi A, et al. Anatomical distribution of
6. Naesdal J, Brown K. NSAID-associated adverse effects and acid control aids synovitis in knee osteoarthritis and its association with joint effusion assessed
to prevent them: a review of current treatment options. Drug Saf on non-enhanced and contrast-enhanced MRI. Osteoarthritis Cartilage
2006;29:119-32 2010;18:1269-74
7. Trelle S, Reichenbach S, Wandel S, et al. Cardiovascular safety of non-ster- 26. Baker K, Grainger A, Niu J, et al. Relation of synovitis to knee pain using
oidal anti-inflammatory drugs: network meta-analysis. BMJ 2011;342:c7086 contrast-enhanced MRIs. Ann Rheum Dis 2010;69:1779-83
8. Agerup B, Berg P, Akermark C. Non-animal stabilized hyaluronic acid: a new 27. Hill CL, Hunter DJ, Niu J, et al. Synovitis detected on magnetic resonance
formulation for the treatment of osteoarthritis. BioDrugs 2005;19:23-30 imaging and its relation to pain and cartilage loss in knee osteoarthritis. Ann
9. Lindqvist U, Tolmachev V, Kairemo K, et al. Elimination of stabilised Rheum Dis 2007;66:1599-603
hyaluronan from the knee joint in healthy men. Clin Pharmacokinet 2002; 28. Ishijima M, Watari T, Naito K, et al. Relationships between biomarkers of
41:603-13 cartilage, bone, synovial metabolism and knee pain provide insights into

! 2014 Informa UK Ltd www.cmrojournal.com NASHA hyaluronic acid for knee osteoarthritis Arden et al. 285
Current Medical Research & Opinion Volume 30, Number 2 February 2014

the origins of pain in early knee osteoarthritis. Arthritis Res Ther 31. Moreland LW. Intra-articular hyaluronan (hyaluronic acid) and hylans for the
2011;13:R22 treatment of osteoarthritis: mechanisms of action. Arthritis Res Ther
29. Torres L, Dunlop DD, Peterfy C, et al. The relationship between specific tissue 2003;5:54-67
lesions and pain severity in persons with knee osteoarthritis. Osteoarthritis 32. Strauss EJ, Hart JA, Miller MD, et al. Hyaluronic acid viscosupplementation
Cartilage 2006;14:1033-40 and osteoarthritis: current uses and future directions. Am J Sports Med
30. Loeuille D, Sauliere N, Champigneulle J, et al. Comparing non-enhanced and 2009;37:1636-44
enhanced sequences in the assessment of effusion and synovitis in knee OA: 33. Bannuru RR, Natov NS, Dasi UR, et al. Therapeutic trajectory following intra-
associations with clinical, macroscopic and microscopic features. articular hyaluronic acid injection in knee osteoarthritis – meta-analysis.
Osteoarthritis Cartilage 2011;19:1433-9 Osteoarthritis Cartilage 2011;19:611-19
Curr Med Res Opin Downloaded from informahealthcare.com by Queen's University on 01/01/15
For personal use only.

286 NASHA hyaluronic acid for knee osteoarthritis Arden et al. www.cmrojournal.com ! 2014 Informa UK Ltd

You might also like