ArticlePDF Available

Anti-Tumour Necrosis Factor Therapy for Dupuytren's Disease: A Randomised Dose Response Proof of Concept Phase 2a Clinical Trial

Authors:

Abstract and Figures

Background: Dupuytren's disease is a common fibrotic condition of the hand that causes irreversible flexion contractures of the fingers, with no approved therapy for early stage disease. Our previous analysis of surgically-excised tissue defined tumour necrosis factor (TNF) as a potential therapeutic target. Here we assessed the efficacy of injecting nodules of Dupuytren's disease with a TNF inhibitor. Methods: Patients were randomised to receive adalimumab on one occasion in dose cohorts of 15 mg in 0.3 ml, 35 mg in 0.7 ml, or 40 mg in 0.4 ml, or an equivalent volume of placebo in a 3:1 ratio. Two weeks later the injected tissue was surgically excised and analysed. The primary outcome measure was levels of mRNA expression for α-smooth muscle actin (ACTA2). Secondary outcomes included levels of α-SMA and collagen proteins. The trial was registered with ClinicalTrial.gov (NCT03180957) and the EudraCT (2015-001780-40). Findings: We recruited 28 patients, 8 assigned to the 15 mg, 12 to the 35 mg and 8 to the 40 mg adalimumab cohorts. There was no change in mRNA levels for ACTA2, COL1A1, COL3A1 and CDH11. Levels of α-SMA protein expression in patients treated with 40 mg adalimumab (1.09 ± 0.09 ng per μg of total protein) were significantly lower (p = 0.006) compared to placebo treated patients (1.51 ± 0.09 ng/μg). The levels of procollagen type I protein expression were also significantly lower (p < 0.019) in the sub group treated with 40 mg adalimumab (474 ± 84 pg/μg total protein) compared with placebo (817 ± 78 pg/μg). There were two serious adverse events, both considered unrelated to the study drug. Interpretation: In this dose-ranging study, injection of 40 mg of adalimumab in 0.4 ml resulted in down regulation of the myofibroblast phenotype as evidenced by reduction in expression of α-SMA and type I procollagen proteins at 2 weeks. These data form the basis of an ongoing phase 2b clinical trial assessing the efficacy of intranodular injection of 40 mg adalimumab in 0.4 ml compared to an equivalent volume of placebo in patients with early stage Dupuytren's disease. Funding: Health Innovation Challenge Fund (Wellcome Trust and Department of Health) and 180 Therapeutics LP.
Content may be subject to copyright.
Research Paper
Anti-Tumour Necrosis Factor Therapy for Dupuytren's Disease:
A Randomised Dose Response Proof of Concept Phase 2a Clinical Trial
Jagdeep Nanchahal
a,
, Catherine Ball
a
, Dominique Davidson
b
,LynnWilliams
a
, William Sones
c
,
Fiona E. McCann
a
,MarisaCabrita
a
, Jennifer Swettenham
a
,NeilJ.Cahoon
b
, Bethan Copsey
c
, E. Anne Francis
a
,
Peter C. Taylor
a
, Joanna Black
c
, Vicki S. Barber
c
, Susan Dutton
c
,MarcFeldmann
a
, Sarah E. Lamb
c
a
Kennedy Institute of Rheumatology, Nufeld Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, Oxford, UK
b
Edinburgh Department of Plastic Surgery, St John's Hospital, Livingston, UK
c
Oxford Clinical Trials Research Unit, Centre for Statistics in Medicine, Nufeld Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, Oxford, UK
abstractarticle info
Article history:
Received 26 April 2018
Received in revised form 8 June 2018
Accepted 19 June 2018
Available online 6 July 2018
Background: Dupuytren's disease is a common brotic condition of the hand that causes irreversible exion con-
tractures of the ngers, with no approved therapy for early stage disease. Our previous analysis of surgically-
excised tissue dened tumour necrosis factor (TNF) as a potential therapeutic target. Here we assessed the
efcacy of injecting nodules of Dupuytren's disease with a TNF inhibitor.
Methods: Patients were randomised to receive adalimumab on one occasion in dose cohorts of 15 mg in 0.3 ml,
35 mg in 0.7 ml, or 40 mg in 0.4 ml, or an equivalent volumeof placebo in a 3:1 ratio. Two weekslater the injected
tissue was surgically excised and analysed. The primary outcome measure was levels of mRNA expression for α-
smooth muscle actin (ACTA2). Secondary outcomesincluded levels of α-SMAand collagen proteins.The trial was
registered with ClinicalTrial.gov (NCT03180957) and the EudraCT (2015-001780-40).
Findings:We recruited 28 patients, 8 assigned to the 15 mg, 12 to the 35 mg and 8 to the 40 mg adalimumab cohorts.
There was no change in mRNA levels for ACTA2, COL1A1, COL3A1 and CDH11. Levels of α-SMA protein expression in
patients treated with 40 mg adalimumab (1.09 ± 0.09 ng per μg of total protein) were signicantly lower (p=
0.006) compared to placebo treatedpatients(1.51±0.09ng/μg). The levels of procollagen type I protein expression
were also signicantly lower (pb0.019) in the sub group treated with 40 mg adalimumab (474 ± 84 pg/μgtotal
protein) compared with placebo (817 ± 78 pg/μg). There were two serious adverse events, both considered unre-
lated to the study drug.
Interpretation: In this dose-ranging study, injection of 40 mg of adalimumab in 0.4 ml resultedindownregulationof
the myobroblast phenotype as evidenced by reduction in expression of α-SMA and type I procollagen proteins at
2 weeks. These data form the basis of an ongoing phase 2b clinical trial assessing the efcacy of intranodular injec-
tion of 40 mg adalimumab in 0.4 ml compared to an equivalent volume of placebo in patients with early stage
Dupuytren's disease.
Funding: Health Innovation Challenge Fund (Wellcome Trust and Department of Health) and 180 Therapeutics LP.
© 2018 The Authors. Published by Elsevier B.V. This is an open access article under the CC BY license
(http://creativecommons.org/licenses/by/4.0/).
Keywords:
Dupuyten's disease
Anti-TNF
Fibrosis
Adalimumab
Myobroblast
1. Introduction
Dupuytren's disease (DD) is a common brotic disease conned to
the hand that affects approximately 4% of the general UK and US popu-
lations [1]. The early stages of the disease are manifest as nodules that
are typically quiescent for a period and thenbecome active, progressing
to cords and exion deformities of the ngers in approximately 50% of
patients [2] and result in loss of hand function [3]. Whilst the mainstay
of treatment remainssurgical excision (fasciectomy) of the diseased tis-
sue or cords [4], approximately 40% of patients in the USA are treated by
disruption of the cords using collagenase or needle fasciotomy [5].
Generally patients undergo these treatments when digits are exed to
30° or more and hand function is impaired [6]. The recurrence rate
following surgery is 21% within 5 years [7] and these individuals may
require more extensive surgery involving excision of the diseased tissue
and overlying skin (dermofasciectomy). Post-operatively, some pa-
tients require prolonged hand therapy and splintage. Complications
occur in approximately 20% of patients undergoing surgery [8]. Alterna-
tive, less invasive techniques to disrupt the cords with a needle orcolla-
genase digestion are associated with rapid recovery of hand function
with minimal therapy. However, recurrence rates are high, affecting
EBioMedicine 33 (2018) 282288
Corresponding authorat: Kennedy Institute of Rheumatology, Nufeld Department of
Orthopaedics, Rh eumatology and Musculoskeletal Sc iences, Universi ty of Oxford,
Roosevelt Drive, Oxford OX3 7FY, UK.
E-mail address: jagdeep.nanchahal@kennedy.ox.ac.uk (J. Nanchahal).
https://doi.org/10.1016/j.ebiom.2018.06.022
2352-3964/© 2018 The Authors. Published by Elsevier B.V. This is an open access article under the CC BY license (http://creativecommons.org/licenses/by/4.0/).
Contents lists available at ScienceDirect
EBioMedicine
journal homepage: www.ebiomedicine.com
85% of patients treated with percutaneous needle aponeurotomy [7]
and 32% of those treated with collagenase [9] at 5 years. The complica-
tion rate is 20% following needle aponeurotomy [8]andover70%after
collagenase injection, the majority being minor and mostly transient
[10].
The ideal therapy would be directed towards patients with early
stage disease to prevent progression to development of cords and sub-
sequent exion contractures of the digits. Our systematic review [11]
highlighted the lack of robust evidence for treatments proposed for
early stage DD for which there is currently no approved therapy. Studies
reporting the efcacy of intranodular injection of steroids or radiother-
apy are limited by a lack of quality, with no blinding or randomisation
and the use of subjective outcome measures [11]. Fifty percent of pa-
tients receiving steroid therapy developed transient subcutaneous atro-
phy or depigmentation. Approximately 2030% of patients receiving
radiotherapy developed long-term adverse effects, including dry skin,
desquamation, skin atrophy, telangiectasia, erythema, and altered heat
and pain sensation. A recent trial reported that injection of collagenase
resulted in nodules becoming smaller and softer, with approximately
50% of patients experiencing bruising and pain [12]. Therefore, there is
a need to develop an effective therapy to retard progression of early
DD and also prevent the development of recurrent disease following
surgery, needle fasciotomy or collagenase injection in patients with
established nger contractures.
The cell responsible for deposition of the excessive collagenous
matrix and contraction in all brotic conditions, including DD, is the
myobroblast [13,14], which is characterised by the expression of α-
smooth muscle actin (α-SMA) [15]. Unlike the brotic diseases of
visceral organs such as the kidney, lung and liver, tissue from patients
with DD is readily accessible, allowing identication of potential novel
therapeutic targets [16]. Using surgically excised tissue from patients
we found that the myobroblasts in DD are aggregated in nodules in
the vicinity of the affected joints, and nodules were absent in patients
with more advanced stage disease [17]. Interspersed through the nod-
ule are immune cells, including macrophages, T cells and mast cells,
and the nodular cells secrete a variety of cytokines [18]. Comparison
of the effects of each of these cytokines showed that only tumour necro-
sis factor (TNF) converted palmar broblasts from patients withDD into
myobroblasts at the low concentrations seen ex vivo, but not non-
palmar broblasts from the same patients or palmar broblasts from
normal individuals. In contrast, TGF-βindiscriminately converts all -
broblasts into myobroblasts [18]. This is important as the brotic pro-
cess seen in DD is limited to the palm of genetically susceptible
individuals. Genome-wide association studies have highlighted the
role of Wnt signalling in DD [19,20] and we found that TNF acted via
the Wnt signalling pathway only in palmar dermal broblasts from pa-
tients with DD [18]. Myobroblasts from DD showed a dose-dependent
reduction in contractility on treatment with anti-TNF, with a concomi-
tant reduction in expression of α-SMA [18]. All the clinically approved
anti-TNF agents assessed were effective in down regulating DD
myobroblast contractility in vitro, with the two fully human IgG mole-
cules, adalimumab and golimumab being the most efcacious at the
doses tested [18].
Here we report the effects of injection of escalating doses of
adalimumab or corresponding volume of placebo directly into the nod-
ules of patients who then underwent surgery two weeks later. Markers
of myobroblast phenotype and collagen production were assessed in
the excised samples.
2. Methods
2.1. Study Design and Patients
Repurposing anti-TNF for Dupuytren's disease (RIDD) is a two-part
phase 2 randomised, double-blinded placebo-controlled dose response
study to assess the efcacy of local injection of adalimumab in patients
with DD. The protocol was reviewed by the South Central Oxford B
Research Ethics Committee (Reference number 15/SC/0259) and the
Medicine and Healthcare products Regulatory Authority (EudraCT no:
2015-001780-40) and has been published [21] (details of subsequent
amendments in appendix). The phase 2a dose escalation study reported
here was performed at a single centre in the UK at the Edinburgh De-
partment of Plastic Surgery at St John's Hospital, NHS Lothian. Patients
referredto the hand surgery service atSt John's Hospitalby their general
practitioner with a diagnosis of DD were screened for entry to the trial.
Eligibility criteria included no prior treatment for Dupuytren's disease to
the affected hand, a clinically distinct nodule of DD, suitability for injec-
tion of adalimumab and fasciectomy. All potential participants were
screened for TB, HIV, hepatitis B and C using serological testing and
chest X-ray in accordance with local standard procedures for anti-TNF
screening.
During the study a new preparation of adalimumab became avail-
able with 40 mg formulated in 0.4 ml. The lower volume and absence
of excipients including citrate was expected to result in reduced pain
and improve participant acceptability. Therefore, the trial design was
modied to include a cohort at a dose of 40 mg using this preparation.
The 40 mg in 0.4 ml formulation is currently only available in a pre-
lled single use syringe and so only the full syringe dose of 40 mg
could be administered with this formulation. Therefore, the trial design
was modied to include a cohort of 40 mg in 0.4 ml. Since publication,
the protocol has been amended to introduce an endpoint to assess
whether adalimumab affects the healing of the surgical incisions or
subsequent scarring. This visual assessment of the surgical wounds
was carried out by blinded review of the hand photographs taken at
baseline and then at 2 and 4 weeks post-surgery.
2.2. Procedures
Patients were randomised (3:1) to receive either adalimumab or sa-
line in 3 dose cohorts (15 mg in 0.3 ml, 35 mg in 0.7 ml), both using the
40 mg in 0.8 ml preparation, or 40 mg in 0.4ml using the more recently
introduced formulation, or an equivalent volume of placebo (normal sa-
line) on one occasion by intra-nodular injection two weeks (±3 days)
prior to scheduled surgery. Nodule hardness was measured using a du-
rometer (RX-1800-00, Rex Gauge Company, Illinois, USA) and nodule
size was assessed using an ultrasound scan before injection and again
before surgery. Blood was collected at baseline and again immediately
before surgery, and the serum stored at -80 °C prior to analysis. ELISA
kits from Promonitor were used according to the manufacturer's in-
structions to measure serum levels of adalimumab (Cat. No. 728552)
and anti-adalimumab (Cat. No. 728533). All samples were measured
in duplicate and repeated on 3 plates using a FLUOstar Omega Spectro-
photometer (BMG Labtech) and MARSsoftware. Pain related to the
adalimumab injection was rated by the participant and the injection
site assessed by visual inspection. Following surgery, the excised
Dupuytren's tissue was transported to the lab, where the nodule was
dissected to retain a central piece for histological examination whilst
the remainder was frozen and pulverised. The resulting powder was
split in two for protein or RNA extraction and stored at 80 °C. RNA
was extracted and following the generation of cDNA, absolute levels of
ACTA2, COL1A1, COL3A1and CDH11 were determined using thestandard
curve method (appendix). Procollagen 1A1 protein levels were deter-
mined by DuoSet ELISA reagents (DY6220-05, R&D systems, Oxon,
UK) in triplicate following the manufacturer's instructions. α-SMA
protein levels were determined using a custom developed MSD
®
plate
(appendix 2). Tissue samples for histology were xed in 4%
paraformaldehyde, longitudinally bisected, embedded in parafn wax
and 7 μm sections obtained from the cut surface. Sequential sections
were stained with hematoxylin-eosin, mouse antiα-SMA antibody
(Abcam 7817) or a mouse isotype control. Antibodies were detected
using biotinylated anti-mouse antibody and avidin/biotin/HRP complex
reagent (Vectastain ABC, Vector Lab, UK). Patients completed their
283J. Nanchahal et al. / EBioMedicine 33 (2018) 282288
standard care following surgery, returning at 2 weeks (±1 week) for as-
sessmentof the wound, change of dressing and commencement of hand
therapy. Final clinical assessment was at 12 weeks (±4 weeks), when a
photograph was obtained for nal blinded assessment of wound healing
and scarring.
2.3. Outcomes
The primary outcome was mRNA expression of ACTA2 (α-SMA). Sec-
ondary outcomes comprised mRNA expression for COL1A1, COL3A1 and
CDH11, and protein levels of α-SMA. Protein levels of type I procollagen,
type III procollagen, acid and pepsin soluble and insoluble collagen,
alongside nodule hardness and size of the nodule on ultrasound scan
were also assessed. Adverse events were assessed using visual inspec-
tion of the injection site and site of surgery, photographs and laboratory
reports. Tertiary outcomes comprised circulating levels of adalimumab,
antibodies to adalimumab and participant injection experience
recorded as pain during injection using a 5-level Likert scale (1 =
Not at all), and immediately after injection also using a Likert scale
(110, 1 = no pain).
2.4. Randomisation and Masking
Participants were randomised for treatment with placebo or
adalimumab using a 1:3 ratio with a block size of four. The dose cohort
was determined by the progression of the trial, with initial patients allo-
cated to the lowest dose cohort (15 mg adalimumab) and doses esca-
lated (35 mg then 40 mg adalimumab) following a review of patient
wound healing two weeks after surgery. The volume of saline solution
for the placebo treatment matched the relevant volume associated
with the dose cohort to ensure blinding. The allocation log, generated
by the trial statistician, using the statistical software STATA version
13.0, was stored securely within the hospital pharmacy. Due to the dis-
tinctive packaging of the 40 mg in 0.4 ml preparation of adalimumab in
pre-lled syringes, blinding of the person administering the injection
was not feasible and therefore this healthcare professional was inde-
pendent and not involved with routine patient care, recruitment, out-
come assessment, surgery or follow-up. A shield prevented patients in
this dose cohort from seeing the syringe. For all cohorts, participants,
healthcare and laboratory professionals involved in recruitment, sur-
gery, follow-up and outcome assessment were blinded to treatment
allocation.
2.5. Statistical Analysis
This study was designed as a phase 2a clinical trial to enable
estimation of potential dose and efcacy of adalimumab in inhibiting
progression of DD. As no previous studies have explored the effect of
adalimumab on in vivo expression of ACTA2 mRNA, sample size was de-
termined using in vitro ndings and inated to account for the non-
clinical nature of the research upon which estimation was performed.
Descriptive statistics, complemented using exploratory data analyses,
were used to assess the demographics across intervention groups for
each dose based on an intention-to-treat population. Additional
analyses were performed using either linear mixed modelling or nested
analysis of variance to account for trial structure, which included a
random-effects component to allow for the values from three plates
and duplicates per participant. Where appropriate, Tukey's HSD adjust-
ment for multiplicity was applied when assessing contrasts. For the
basis of most contrasts the patients allocated to placebo were pooled.
The validity of samples for circulating levels of adalimumab could not
be veried for three patients. To address this, a per-protocol approach
was adopted and the corresponding patients were excluded from
analysis.
2.6. Role of the Funding Source
The study was funded by the Health Innovation Challenge Fund
(Wellcome Trust and Department of Health). Funding for purchase of
the adalimumab was provided by 180 Therapeutics LLP. The funders
had no involvement in study design or data analyses. The study was
sponsored by the University of Oxford. Raw unblinded data were
analysed by the trial statisticians (WS and SD) and subsequently made
available to the remainder of the trial team. The corresponding author
drafted the manuscript in conjunction with all the other authors and
all share the responsibility for submission for publication.
3. Results
3.1. Patient Demographics
Between November 2015 and November 2016, 85 participants were
screened and 28 were randomised tosuccessive dose escalation cohorts,
8 to the 15 mg adalimumab cohort, 12 to the 35 mg adalimumab cohort
and 8 to the 40 mg adalimumab cohort. In each cohort, patients were
randomised in a ratio of 3:1 to receive either adalimumab or an equiva-
lent volume of placebo (saline) (Fig. 1). Baseline characteristics were
similar between different cohorts (Table 1).
3.2. Outcomes
There was no difference between any of the groups, including pla-
cebo, in mRNA levels for genes that encode ACTA2,COL1A1,COL3A1 or
CDH11 (Fig. 2). Levels of α-SMA protein expression in patients treated
with 40 mg adalimumab (1.09 ± 0.09 ng per μgoftotalprotein)were
statistically signicantly lower(p= 0.006) compared toplacebo treated
patients (1.51 ± 0.09 ng per μg of total protein) and compared to those
treated with 15 mg (1.60 ± 0.09 ng per μg of total protein; pb0.001) or
35 mg (1.44 ± 0.08 ng per μg of total protein; p=0.024)adalimumab,
whilst 35 mg and 15 mg adalimumab cohorts showed no difference
compared to placebo (Fig. 3). Qualitative assessment of immunohisto-
chemical staining for α-SMA showed reduced intensity for the re-
sponders in the 40 mg cohort compared to placebo (appendix,
Supplementary Fig. 1).
The levels of procollagen type I protein expression were signicantly
lower (p= 0.019) in the subgroup treated with 40 mg adalimumab
(474 ± 84 pg/μg total protein) compared with placebo (817 ±
78 pg/μg total protein) (Fig. 4). Pro-collagen type I levels were below
the minimum limit for quantication of 250 pg/μg in three patients
injected 35 mg adalimumab and two injected with 40 mg adalimumab.
Tissue fromnodules surgically excised two weeksafter injection was
also assessed for levels of procollagen type III and collagen using the
Biocolor (UK) Sircol Soluble and Insoluble Collagen Assays. There were
no differences between treatment groups and placebo for either assay.
Plasma levels of adalimumab and antibodies to adalimumab were
assayed prior to intra-nodular injection of adalimumab and repeated
2 weeks (±3 days) after injection,prior to surgery. Prior to nodule injec-
tion, circulating blood adalimumab levels were below the minimum de-
tectable limit for all patients. At two weeks after injection, patients
within the placebo treatment group had undetectable levels of blood
adalimumab whilst all patients within adalimumab treatment groups
demonstrated detectable levels (appendix, Supplementary Table 1).
Two weeks after treatment, no antibodies to adalimumab were detected
amongst placebo treated patients, demonstrating assay specicity and
the absence of cross reactivity. Antibodies were detected in one patient
treated with 15 mg adalimumab, 5 patients treated with 35 mg
adalimumab and two patients treated with 40 mg adalimumab. How-
ever, antibody levels only exceeded the minimum threshold for clinical
signicance [22] of 12 AU/ml in two of the three repeat measurements
(mean±SD, 12.4±0.63 AU/ml) in one patient treated with 35 mg
adalimumab.
284 J. Nanchahal et al. / EBioMedicine 33 (2018) 282288
Ultrasound scans performed prior to injection and two weeks later
prior to surgery showed no signicant changes in any of the dose
cohorts in nodule size based on the area of the nodule, the length of
the perimeter and the feret (maximum length across the nodule)
(appendix, Supplementary Fig. 2). Similarly, there was no change in
nodule hardness measured using a durometer in any of the dose cohorts
when comparing before and two weeks after injection (appendix,
Supplementary Fig. 3).
All but one patient reported pain during the injection, with higher
levels of pain experienced with larger volumes and with the more dilute
formulation of adalimumab. The Likert scores immediately after injec-
tion showed that high volume of injection (0.7 ml) was associated
with more pain than lower volumes (0.30.4 ml) and that the 40 mg
in 0.4 ml adalimumab formulation was associated with lower pain
scores (appendix, Supplementary Fig. 4).
3.3. Adverse Events
Two patients suffered serious adverse events (SAE) after injection.
One patient fell two months before injection of placebo whilst running
and presented 5 days after surgery for DD with impaired motor function
due to bilateral subdural haematomas that were treated non-
operatively. He made a full recovery with spontaneous resolution of
Table 1
Baseline characteristics for treatment groups. Ageof onset of Dupuytren's disease and age
on entry into trial shown by treatment group. Patients receiving placebo are pooled.
Treatment Mean SD Min Max Median n
Age upon onset (years)
15 mg adalimumab 47.0 10.5 30 60 48.5 6
35 mg adalimumab 53.8 11.9 28 67 57.0 9
40 mg adalimumab 53.8 5.6 45 60 55.0 6
Sodium chloride 0.9% 56.1 8.5 42 69 57.0 7
Age upon trial entry
15 mg adalimumab 57.2 9.1 48 71 53.0 6
35 mg adalimumab 63.7 9.9 48 78 64.0 9
40 mg adalimumab 63.3 5.7 56 69 65.0 6
Sodium chloride 0.9% 62.9 8.3 46 72 63.0 7
Total
Age upon onset 52.9 9.8 28 69 56.0 28
Age upon trial entry 62.0 8.5 46 78 63.0 28
Assessed for eligibility (n = 85)
Excluded (n=57)
Not meeting inclusion criteria (n=13)
Declined to participate (n=34)
Screened ineligible (n=7)
Screened declined/not recorded (n=3)
Withdrew consent (n=0)
Allocated to adalimumab injection (n = 6)
Received adalimumab (n = 6)
Randomised to
15 mg (n = 8)
Allocated to saline injection (n = 2)
Received saline (n = 2)
Randomised to
35 mg (n = 12)
Allocated to adalimumab injection (n = 9)
Received adalimumab (n = 9)
Randomised to
40 mg (n = 8)
Randomised (n=28)
Allocated to saline injection (n = 3)
Received saline (n = 3)
Allocated to adalimumab injection (n = 6)
Received adalimumab (n = 6)
Allocated to saline injection (n = 2)
Received saline (n = 2)
Fig. 1. Trial prole.
285J. Nanchahal et al. / EBioMedicine 33 (2018) 282288
the haematomas. A patient with type 2 diabetes who received an injec-
tion of 35 mg of adalimumab into a nodule situated over the proximal
phalanx ofthe little nger presented four days aftersurgical fasciectomy
with a suture abscess at the distal palmar crease in the axis of the ring
nger. He was initially treated with oral antibiotics by his general prac-
titioner but due to lack of improvement after 5 days he wasadmitted to
hospital for i.v. antibiotics. Inspection of the wounds revealederythema
around the sutures and a small amount of pus was expressed following
suture removal. He was discharged home the following day on oral
antibiotics and the wound healed uneventfully. Blinded assessment of
photographs of scars obtained 12 weeks (±4 weeks) after surgery
Fig. 2. Box and whiskers plotof log RNA concentration by treatment received. The gureis divided into separate plots for each gene assessed. The box represents the inter-quartile range
(IQR) and whiskers extend to 1.5 relevant IQR (Tukey boxplot). The x-axisshows individual patients, grouped by treatment received, with the three repeatmeasures performedfor each
patient represented by points stacked within the same column.
Fig. 3. Box and whiskers plot of α-SMA protein concentration by treatment received. The
box represents the inter-quartile range (IQR), the horizontal line represents the median
and whiskers extend to 1.5 relevant IQR. The x-axis shows individual patients, grouped
by treatment received. Samples from each patient were analysed in duplicate on three
separate plates and the values for each plate are shown as circles, triangles or squares.
Fig. 4. Box and whiskers plot of pro-collagen protein concentration by treatmentreceived.
The box represents the inter-quarti le range (IQR), the horizontal line represents the
median and whiskers extend to 1.5 relevant IQR (Tukey boxplot). The x-axis shows
individual pati ents, grouped by tr eatment received, with the three repeat measures
performed for each patient represented by points stacked within the same column. The
plot includes pro-collagen concentrations reported be low the minimum detectable
range as zero, 3 of 9 in 35 mg and 2 of 6 in 40 mg adalimumab cohorts.
286 J. Nanchahal et al. / EBioMedicine 33 (2018) 282288
showed well-healed mature scars in all patients except the patientwho
developed the post-operative wound infection, where the scars were
more prominent. Both events were considered by the local investigator
and an independent reviewer not to be related to the investigational
medicinal product and the second SAE was considered to be related to
the surgical procedure.
Swelling at the injection site was only apparent in 3 patients receiving
35 mg adalimumab and two receiving 0.7 ml saline. Transient redness
was noted at the injection site in 7 patients distributed across the differ-
ent treatment groups. No patient experienced blistering, haematoma,
bruising, local itching or signs of nerve injury. No patient reported any ad-
verse events one week after injection.
4. Discussion
Our results show that two weeks following administration of 40 mg of
adalimumab in 0.4 ml into Dupuytren's nodules there was down regula-
tion of the myobroblast phenotype as evidenced by lower expression of
α-SMA and pro-collagen type I proteins. These ndings represent the
clinical translation of our in vitro data based on human tissue where we
showed that tumour necrosis factor (TNF) selectively converts precursor
palmar broblasts from Dupuytren's patients to myobroblasts via the
Wnt signalling pathway, and anti-TNF is inhibitory [18]. Like all brotic
conditions, Dupuytren's disease is characterised by the deposition of ex-
cessive collagenous extracellular matrix which is remodelled and
contracted by α-SMA-expressing myobroblasts [15], that aggregate in
nodules [17]. Collagen is synthesised and secreted in the precursor state
as procollagen and the N- and C-terminal globular domains are cleaved
at the plasma membrane to produce tropocollagen that self-aggregates
to form brils, which grow and are eventually cross-linked by lysyl oxi-
dase to form insoluble collagen [23]. The soluble Sircol assay detects all
the uncross-linked forms and the insoluble assay only detects the cross-
linked collagens. The short half-life of procollagen due to its rapid conver-
sion to brillar collagen may explain the reduction in type I procollagen
we observed at the two-week primary endpoint and it is possible that
soluble and insoluble collagens may also change but at later time points.
Although type III collagen comprises 3549% of the total [24]in
Dupuytren's nodules, which represent the earlier stages of disease, we
were unable to detect any changes in type III procollagen due to the
poor sensitivity of the only commercially available assay, with levels fall-
ing below the threshold of detection in half of samples in all treatment
groups. The downregulation of both procollagen and the contractile pro-
tein α-SMA by local inhibition of tumour necrosis factor (TNF) provides
the rst successful proof of concept for targeted treatment of the patho-
logical mechanisms underlying this common localised brotic disease
of the hand that affects 4% of the general UK and US populations [1].
There was nochange in the levels mRNA levels for genes that encode
for ACTA2,COL1A1,COL3A1 and CDH11. The most likely explanation for
the down regulation in protein expression for α-SMA and type I
procollagen without a concomitant change in the respective mRNA is
that expression of these proteins is predominantly regulated at the
post-transcriptional level. We have previously shown to be the case
for α-SMA in Dupuytren's myobroblasts [25], but did not select α-
SMA protein levels as the primary outcome measure as the assay for
the protein was optimised over the course of the trial. Collagen 1 ex-
pression is also mainly regulated post translationally in brotic condi-
tions [26]. Another potential but less likely explanation would be
different half-lives of mRNA and collagen proteins of hours and days re-
spectively [26], which would potentially allow mRNA levels to recover
at 2 weeks whilst corresponding protein levels remained suppressed.
The downregulation of α-SMA and procollagen type I only occurred
in patients where 40 mg of adalimumab in 0.4 ml was injected directly
into the nodule, but not following administration of 35 mg in 0.7 ml.
This could be explained by some of the adalimumab in the more dilute
preparation extravasating out of the nodule and hence not available to
act at high concentrations locally on the aggregates of myobroblasts
and immune cells. An alternative but less likely explanation is that the
excipients in the more dilute preparation of adalimumab rendered it
less efcacious for this application. Our data would suggest that only
high local concentrations of adalimumab are effective in down regulat-
ing the myobroblast phenotype in DD and it is unlikely adalimumab or
other anti-TNF drugs would be efcacious if administered systemically.
This would be consistent with the low-grade inammation that is con-
ned to the nodules of DD [18]. The half-life of adalimumab is 2 weeks
[27], and trough levels at this time for patients on regular systemic ther-
apy of 40 mg every 2 weeks averaged ~5μg/ml in patients with rheuma-
toid arthritis and 610μg/ml in patients with psoriatic arthropathy not
treated with concomitant methotrexate [27]. This would suggest that
the kinetics of absorption of adalimumab when injected into the nod-
ules of DD may be similar to when the drug is administered subcutane-
ously. We would postulate that formulations that retard dissipation of
adalimumab from the site of injection are likely to be more effective
and may need to be administered less frequently for the treatment of
localised brotic conditions characterised by low-grade inammation
such as DD. Neutralising antibodies to adalimumab are likely to result
in reduced efcacy [28]. Only one patient, who received 35 mg in
0.7 ml, had antibody levels (12.4±0.63 AU/ml) just above the threshold
of signicance of 12Au/ml [22], which are unlikely to be of clinical sig-
nicance. Neutralising antibodies may occur more commonly after re-
peated administration of adalimumab, especially in patients who are
not on concomitant methotrexate.
Whilst almost all patients reported pain during injection, pain scores
immediately after injection were higher in participants receiving volumes
of either placebo or adalimumab N0.30.4 ml and in those injected with
the formulation of adalimumab comprising 40 mg in 0.8 ml of carrier
that was used for the 15 mg and 35 mg cohorts. This formulation differs
signicantly from the more concentrated form of 40 mg in 0.4 ml in
that it contains a variety of excipients [27], including citrate, which is as-
sociated with higher pain scores when injected subcutaneously [29]. The
40 mg in 0.4 ml formulation was not available commercially when the
trial opened to recruitment and therefore the study was initially based
on dose cohorts using the 40 mg in 0.8 ml formulation. Transient swelling
at the injection site was apparent in some patients receiving 0.7 ml of ei-
ther placebo or adalimumab, most likely due to extravasation of the
injected material outside the connes of the nodule. The safety prole
of adalimumab is well-known and the commonest adverse events in
patients with autoimmune disorders on long-term therapy are related
to infection [30]. One patient in our study, who suffered from type 2
diabetes mellitus received 35 mg of adalimumab developed a wound
infection 4 days following surgery that resolved following removal of
sutures and antibiotics. The infection was considered to be unrelated to
the adalimumab injection as it was a localised suture abscess away
from the site of the injected nodule. Wound infection has been reported
to occur in approximately 3.6% of patients undergoing Dupuytren's
fasciectomy [8].
Unsurprisingly, there was no difference in nodule hardness or sizeas
assessed by tonometry and ultrasound scanrespectively at 2 weeks post
injection. This trial enabled us to optimisethe methods for obtaining the
data and analysing them in preparation for follow up studies.
5. Conclusions
This phase 2a randomised trial shows that a single intranodular
injection of 40 mg adalimumab in 0.4 ml in patients with Dupuytren's
disease is safe and leads to down regulation of the myobroblast
phenotype as evidenced by reduced expression of α-SMA and type I
procollagen proteins. Having dened the most efcacious dose and
preparation and based on these positive proof of concept data we are
now proceeding with a phase 2b trial in 138 patients with early
stage Dupuytren's disease randomised 1:1 to receive 4 injections of
adalimumab or placebo at 3 month intervals and followed for a total
of 18 months from baseline [21]. Our study also illustrates the utility
287J. Nanchahal et al. / EBioMedicine 33 (2018) 282288
of using early stage brotic human tissue to elucidate novel therapeutic
targets [18] that can be translated to the clinic.
Conicts of Interest
Mrs. Ball, Dr. Davidson, Dr. Williams, Dr. Sones, Dr. McCann, Ms.
Cabrita, Dr. Swettenham, Dr. Cahoon, Dr. Copsey, Dr. Francis, Dr. Black,
Dr. Barber, Mrs. Dutton report grants from Wellcome Trust, grants
from Department of Health, other from 180 Therapeutics LP, during
the conduct of the study.
Professor Taylor reports grants from Wellcome Trust, grants from
Department of Health, other from 180 Therapeutics LP, during the con-
duct of the study; grants from UCB, personal fees from UCB, AbbVie,
Pzer, outside the submitted work.
Professor Lamb reports grants from Wellcome Trust, grants from
Department of Health, during the conduct of the study; Professor
Sarah Lamb reports grants from NIHR Health Technology Assessment
Programme during the conduct of this study.
Professor Nanchahal and Professor Feldmann report grants from
Wellcome Trust, grants from Department of Health, other from 180
Therapeutics LP, during the conduct of the study; In addition, Professor
Nanchahal has a patent PCT/EP2011/069147 issued to 180 Therapeutics,
and with Professor Feldmann a patent PCT/US2017/049691 pending, a
patent PCT/US2017/026382 pending, a patent 16759325.0 EP2017
pending, and a patent 16759326.8 EP2017 pending.
Funding Sources
We would like to acknowledge funding from the Health Innovation
Challenge Fund (HICF) (HICF-R8-433), a parallel funding partnership
between the Wellcome Trust and the Department of Health, 180 Thera-
peutics, and the National Institute for Health Research (NIHR) Oxford
Biomedical Research Centre (BRC). The views expressed are those of
the authors and not necessarily those of the NHS, the NIHR or the
Department of Health.
Author Contributions
JN, SL, MF, CB, SD and PT conceived and designed the study. DD and
NC recruited the patients. CB, DDand NC collected the clinical data. LW,
FM and MC performed the laboratory analyses. WS, SD and BC per-
formed the statistical analyses. JS, VB, JB, EAF and SL managed the trial.
Appendix A. Supplementary Data
Supplementary data to this article can be found online at https://doi.
org/10.1016/j.ebiom.2018.06.022.
References
[1] Hindocha S, McGroutherDA, Bayat A. Epidemiological evaluation of Dupuytren's dis-
ease incidence and prevalence rates in relati on to etiology. Hand (N Y) 2009;4:
25669.
[2] Reilly RM, Stern PJ, Goldfarb CA. A retrospective revie w of the management o f
Dupuytren's nodules. J Hand Surg 2005;30:10148.
[3] Engstrand C, Krevers B, Kvist J. Factors affecting functional recovery after surgery
and hand therapy in patients with Dupuytren's disease. J Hand Ther 2015;28:
255259; quiz 260.
[4] Davis TR. Surgical treatment of primary Dupuytren's contractures of the ngers in
the UK: surgeons' preferences and research priorities. J Hand Surg Eur Vol 2013;
38:835.
[5] Zhao JZ, Hadley S, Floyd E,Earp BE, Blazar PE. The impact of collagenase Clostridium
histolyticum introduction on Dupuytren treatment patterns in the United States.
J Hand Surg 2016;41:9638.
[6] Rayan GM. Dupuytren's disease: anatomy, pathology, presentation, and treatment.
Instr Course Lect 2007;56:10111.
[7] van Rijssen AL, ter Linden H, Werker PM. Five-year results of a randomized clinical
trial on treatment in Dupuytren's disease: percutaneous needle fasciotomy versus
limited fasciectomy. Plast Reconstr Surg 2012;129:46977.
[8] Crean SM, Gerber RA, Le Graverand MP, Boyd DM, Cappelleri JC. The efcacy and
safety of fasciectomy and fasciotomy for Dupuytren's contracture in European pa-
tients: a structu red review of pu blished studies. J Hand Surg Eur Vol 2011;36:
396407.
[9] Peimer CA, Wilbrand S,Gerber RA, Chapman D, Szczypa PP. Safety and tolerability of
collagenase Clostridium histolyticum and fasciectomy for Dupuytren's contracture. J
Hand Surg Eur Vol 2015;40:1419.
[10] Hurst LC, Badala mente MA, Hentz VR, et al. Injecta ble collagenase clostridium
histolyticum for Dupuytren's contracture. N Engl J Med 2009;361:96879.
[11] Ball C, Izadi D, Verjee LS, Chan J, Nanchahal J. Systematic review of non-surgical
treatments for early dupuytren's disease. BMC Musculoskelet Disord 2016;17:345.
[12] CostasB, Coleman S, Kaufman G, James R, Cohen B,Gaston RG. Efcacy and safety of
collagenase clostridium histolyticum for Dupuytren disease nodules: a randomized
controlled trial. BMC Musculoskelet Disord 2017;18:374.
[13] Darby IA, Zakuan N, Billet F, Desmouliere A. The myobroblast, a key cell in normal
and pathological tissue repair. Cell Mol Life Sci 2016;73:114557.
[14] Wynn TA, Ramalingam TR. Mechanisms of brosis: therapeutic translation for -
brotic disease. Nat Med 2012;18:102840.
[15] Hinz B, Phan SH, Thannickal VJ, Galli A, B ochaton-Piall at ML, Gabbiani G. Th e
myobroblast: one function, multiple origins. Am J Pathol 2007;170:180716.
[16] Nanchahal J, Hinz B. Strategies to overcome the hurdles to treat brosis, a major
unmet clinical need. Proc Natl Acad Sci U S A 2016;113:72913.
[17] Verjee LS, Midwood K, Davidson D, Essex D, Sandison A, Nanchahal J. Myobroblast
distribution in Dupuytren's cords: correlation with digital contracture. J Hand Surg
2009;34:178594.
[18] Verjee LS, Verhoekx JS, Chan JK, et al. Unraveling the signaling pathways promoting
brosis inDupuytren's diseasereveals TNF as a therapeutic target.Proc Natl Acad Sci
U S A 2013;110:E9 2837.
[19] Dolmans GH, Werker PM, Hennies HC, et al. Wnt signaling and Dupuytren's disease.
N Engl J Med 2011;365:30717.
[20] Ng M, Thakkar D, Southam L, et al. A genome-wide association study of dupuytren
disease reveals 17 additional variants implicated in brosis. Am J Hum Genet
2017;101:41727.
[21] Nanchahal J, Ball C, Swettenham J, et al. Study protocol: a multi-Centre, doubleblind,
randomised, placebo-controlled, parallel group, phase II trial (RIDD) to determine
the efcacy of intra-nodular injection of anti-TNF to control disease progression in
early Dupuytren's disease, with an embedded dose response study. Wellcome
Open Res 2017. https://wellcomeopenresearch.org/articles/2-37/v2.
[22] Bartelds GM, Krieckaert CL, Nurmohamed MT, et al. Development of antidrug anti-
bodies against adalimumab and association with disease activity and treatment fail-
ure during long-term follow-up. JAMA 2011;305:14608.
[23] Canty EG, Kadler KE. Procollagen trafcking, processing and brillogenesis. J Cell Sci
2005;118:134153.
[24] Lam WL, Rawlins JM, Karoo RO, Naylor I, Sharpe DT. Re-visiting Luck's classication:
a histological analysis of Dupuytren's disease. J Hand Surg Eur Vol 2010;35:3127.
[25] Verjee LS, Midwood K, Davidson D, Eastwood M, Nanchahal J. Post-transcriptional
regulation of alpha-smooth muscle actin determines the contractile phenotype of
Dupuytren's nodular cells. J Cell Physiol 2010;224:68190.
[26] Stefanovic B. RNA protein interactions governing expression of the most abundant
protein in human body, type I collagen. Wiley Interdiscip Rev RNA 2013;4:53545.
[27] Abbvie. http://www.rxabbvie.com/pdf/humira.pdf;2018.
[28] ChenDY, Chen YM, Tsai WC, et al. Signicantassociations of antidrug antibodylevels
with serum drug trough levels and the rapeutic respon se of adalimumab and
etanercept treatment in rheumatoid arthritis. Ann Rheum Dis 2015;74:e16.
[29] Laursen T, Hansen B, FiskerS. Pain perception aftersubcutaneous injections of media
containing different buffers. Basic Clin Pharmacol Toxicol 2006;98:21821.
[30] Burmester GR, Panaccione R, Gordon KB, McIlraith MJ, Lacerda AP. Adalimumab:
long-termsafety in 23 458 patients fromglobal clinical trialsin rheumatoid arthritis,
juvenile idiopathic arthritis, ankylosing spondylitis, psoriatic arthritis, psoriasis and
Crohn's disease. Ann Rheum Dis 2013;72:51724.
288 J. Nanchahal et al. / EBioMedicine 33 (2018) 282288
Commentary
Cytokine Targeted Therapy for Dupuytren's Disease
Latha Satish
Research Department, Shriners Hospitals for Children, Cincinnati, OH-45229, USA
Department of Pathology and Laboratory Medicine, University of Cincinnati, OH-45229, USA
Dupuytren's disease (DD) is a complex broproliferative disease of
the hand and a difcult condition to live with for the patients and a puz-
zling disease for the treating physicians. It has been morethan 180 years
since Baron Dupuytren was credited with discovering the disease, yet
there is no denitive treatment to combat DD. Varying numbers with
regards to prevalence rate are reported but a recent systematic review
estimated 12% those aged 55 years are affected, rising to 29% in those
aged 75 years in the general population in Western countries [1]. DD
is a benign condition and early stage disease manifests itself as pits or
nodules in the palm that develop into cords [2]. Non-invasive treat-
ments for early stage disease are of limited efcacy [3] and the mainstay
of treatment for late stage disease for years has been surgery. Recently,
collagenase Clostridium histolyticum (CCH) injection has gained popu-
larity as an alternative to surgery for treating DD. However, it is associ-
ated with a high rate of adverse effects [4] and patient satisfaction
decreases over time asthe disease recurs[5]. Cu rrent research is focused
on targeting factors (mainly cytokines) that are responsible for mediat-
ing matrix production in DD, specically collagen. Several growth fac-
tors mainly TGF-β1, PDGF, IGF, CTGF, bFGF and the pro-inammatory
cytokine TNF are found to play a prominent role in the progression of
DD. TNF has been shown to act via the Wnt signaling pathway to pro-
mote contraction and probrotic signaling in DD cells. In vitro studies
also showed that neutralizing antibodies to TNF downregulates
myobroblast activity [6]. In EBioMedicine, Nanchahal and colleagues
identied TNF as a potential target for clinical translation to treat DD [7].
The study reports a phase 2a double-blind, randomized placebo-
controlled clinical trial on the efcacy of injecting nodules of DD with
adalimumab, a TNF inhibitor [7]. The authors should be applauded for
choosing a local delivery route via intra-nodular injection to assess the
efcacy of a biologic, which might become routine in the future to
avoid the necessity for surgical procedure. In this trial, the authors
chose to inject the nodules two weeks prior to the scheduled surgery
to test three different doses of adalimumab, which are 15 mg in 0.3 ml
(n=6;placebon= 2), 35 mg in 0.7 ml (n=9;placebon=3)or
40 mg in 0.4 ml (n = 6; placebo n = 2). Surgically excised tissues
were subjected to mRNA and protein analyses. The primary outcome
measure was to determine the levels of mRNA expression for α-SMA.
Secondary outcomes were to determine the mRNA expression for
collagen types I, III, and cadherin 11, as well as levels of α-SMA and col-
lagen proteins. The authors found no changes in the mRNA expression
for all genes assayed. Expression of α-SMA protein was reduced in pa-
tients who received 40 mg of adalimumab compared to those injected
with placebo, or 35 mg or 15 mg of adalimumab. These data are in con-
cordance with the previous in vitro nding that inhibiting TNF levels can
downregulate the myobroblast phenotype [6]. Previous reports sug-
gest an increased proportion of type III collagen in earlier lesions,
which changes to a greater proportion of type I collagen at later stages
of the disease [8]. In the present study, the authors reported that TNF in-
hibition decreased the protein expression of procollagen type I but no
differences were noted in procollagen type III levels due to the low sen-
sitivity of the assay for procollagen type III [7]. TNF inhibition also did
not reduce nodule size or hardness, which is not unexpected as the in-
jection was administered only on one occasion.
The study also has a few limitations, which hopefully be addressed
by the authors in their upcoming trial. An interesting nding would
have been if the levels of the growth factors mainly TGF-β1andPDGF,
along with TNF in the excised tissue was reported, which would have
added more strength on the utility of adalimumab for DD. Another in-
teresting observation would have been if authors had reported the
changes in the expression level of the ECM protein namely bronectin
a known contributor in DD pathogenesis along with type I and type III
collagens.
Overall, with this trial, the authors haveestablished safety and deter-
mined the effective dose and volume (40 mg in 0.4 ml) of adalimumab
to proceed to a phase 2b clinical trial with a larger cohort of patients
with early-stage DD to investigate the efcacy of intra-nodular injec-
tions every 3 months over a 12-month period. The ongoing phase 2b
clinical trial should allow the authors to ascertain whether multiple in-
jections can decrease nodule size and hardness. If a signicant decrease
in nodule size and hardness is noticed, it indirectly reects that there
might have been a signicant decrease in myobroblast formation and
collagen accumulation. The authors should also consider a futureclinical
trial of patients with the advanced form of the disease to investigate the
potential of adalimumab in preventing recurrence.
Disclosure
The author declared no conicts of interest.
EBioMedicine 34 (2018) 1415
DOI of original article: https://doi.org/10.1016/j.ebiom.2018.06.022.
Corresponding author at: ShrinersHospitals for Children, Research Department, 3229
Burnet Avenue, Cincinnati, OH 45229, USA.
E-mail addresses: lsatish@shrinenet.org,satishla@uc.edu (L. Satish).
Contents lists available at ScienceDirect
EBioMedicine
journal homepage: www.ebiomedicine.com
https://doi.org/10.1016/j.ebiom.2018.07.016
2352-3964/© 2018 The Author. Published by Elsevier B.V. This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).
References
[1] Lanting R, Broekstra DC, Werker PM, van den Heuvel ER. A systematic review and
meta-analysis on the prevalence of Dupuytren disease in the general population of
Western countries. Plast Reconstr Surg 2014;133:593603.
[2] Warwick D. Dupuytren's disease: my personal view. J Hand Surg Eur 2017;42E(7):
66572.
[3] Ball C, Izadi D, Verjee LS, Chan J, Nanchahal J. Systematic review of non-surgical treat-
ments for early dupuytren's disease. BMC Musculoskelet Disord 2016 Aug 15;17(1):
345.
[4] Peimer CA, Wilbrand S, Gerber RA, Chapman D, Szczypa PP. Safety and tolerability of
collagenase clostridium histolyticum and fasciectomy of dupuytren's contracture. J
Hand Surg Eur 2014;40(2):1419.
[5] BradleyJ, Warwick D. Patient satisfaction with collagenase. J Hand Surg [Am] 2016;41
(6):68997.
[6] Verjee LS, Verhoekx JS, Chan JK, Krausgruber T, Nicolaidou V, Izadi D, Davidson D,
Feldmann M, Midwood KS, Nanchahal J. Unraveling the signaling pathways promot-
ing brosis in Dupuytren's disease reveals TNF as a therapeutic target. Proc Natl Acad
Sci U S A 2013;110:E92837.
[7] Nanchahal J, Ball C , Davidson D, et al . Anti-tumour n ecrosis factor therapy for
Dupuytren's disease: a randomised dose response proof of concept phase 2a clinical
trial. EBioMedicine 2018. https://doi.org/10.1016/j.ebiom.2018.06.022.
[8] Lam WL, Rawlins JM, Karoo RO, Naylor I, Sharpe DT. Re-visiting Luck's classication: a
histological analysis of Dupuytren's disease. J Hand Surg Eur 2010;35:3127.
15L. Satish / EBioMedicine 34 (2018) 1415
showed well-healed mature scars in all patients except the patientwho
developed the post-operative wound infection, where the scars were
more prominent. Both events were considered by the local investigator
and an independent reviewer not to be related to the investigational
medicinal product and the second SAE was considered to be related to
the surgical procedure.
Swelling at the injection site was only apparent in 3 patients receiving
35 mg adalimumab and two receiving 0.7 ml saline. Transient redness
was noted at the injection site in 7 patients distributed across the differ-
ent treatment groups. No patient experienced blistering, haematoma,
bruising, local itching or signs of nerve injury. No patient reported any ad-
verse events one week after injection.
4. Discussion
Our results show that two weeks following administration of 40 mg of
adalimumab in 0.4 ml into Dupuytren's nodules there was down regula-
tion of the myobroblast phenotype as evidenced by lower expression of
α-SMA and pro-collagen type I proteins. These ndings represent the
clinical translation of our in vitro data based on human tissue where we
showed that tumour necrosis factor (TNF) selectively converts precursor
palmar broblasts from Dupuytren's patients to myobroblasts via the
Wnt signalling pathway, and anti-TNF is inhibitory [18]. Like all brotic
conditions, Dupuytren's disease is characterised by the deposition of ex-
cessive collagenous extracellular matrix which is remodelled and
contracted by α-SMA-expressing myobroblasts [15], that aggregate in
nodules [17]. Collagen is synthesised and secreted in the precursor state
as procollagen and the N- and C-terminal globular domains are cleaved
at the plasma membrane to produce tropocollagen that self-aggregates
to form brils, which grow and are eventually cross-linked by lysyl oxi-
dase to form insoluble collagen [23]. The soluble Sircol assay detects all
the uncross-linked forms and the insoluble assay only detects the cross-
linked collagens. The short half-life of procollagen due to its rapid conver-
sion to brillar collagen may explain the reduction in type I procollagen
we observed at the two-week primary endpoint and it is possible that
soluble and insoluble collagens may also change but at later time points.
Although type III collagen comprises 3549% of the total [24]in
Dupuytren's nodules, which represent the earlier stages of disease, we
were unable to detect any changes in type III procollagen due to the
poor sensitivity of the only commercially available assay, with levels fall-
ing below the threshold of detection in half of samples in all treatment
groups. The downregulation of both procollagen and the contractile pro-
tein α-SMA by local inhibition of tumour necrosis factor (TNF) provides
the rst successful proof of concept for targeted treatment of the patho-
logical mechanisms underlying this common localised brotic disease
of the hand that affects 4% of the general UK and US populations [1].
There was nochange in the levels mRNA levels for genes that encode
for ACTA2,COL1A1,COL3A1 and CDH11. The most likely explanation for
the down regulation in protein expression for α-SMA and type I
procollagen without a concomitant change in the respective mRNA is
that expression of these proteins is predominantly regulated at the
post-transcriptional level. We have previously shown to be the case
for α-SMA in Dupuytren's myobroblasts [25], but did not select α-
SMA protein levels as the primary outcome measure as the assay for
the protein was optimised over the course of the trial. Collagen 1 ex-
pression is also mainly regulated post translationally in brotic condi-
tions [26]. Another potential but less likely explanation would be
different half-lives of mRNA and collagen proteins of hours and days re-
spectively [26], which would potentially allow mRNA levels to recover
at 2 weeks whilst corresponding protein levels remained suppressed.
The downregulation of α-SMA and procollagen type I only occurred
in patients where 40 mg of adalimumab in 0.4 ml was injected directly
into the nodule, but not following administration of 35 mg in 0.7 ml.
This could be explained by some of the adalimumab in the more dilute
preparation extravasating out of the nodule and hence not available to
act at high concentrations locally on the aggregates of myobroblasts
and immune cells. An alternative but less likely explanation is that the
excipients in the more dilute preparation of adalimumab rendered it
less efcacious for this application. Our data would suggest that only
high local concentrations of adalimumab are effective in down regulat-
ing the myobroblast phenotype in DD and it is unlikely adalimumab or
other anti-TNF drugs would be efcacious if administered systemically.
This would be consistent with the low-grade inammation that is con-
ned to the nodules of DD [18]. The half-life of adalimumab is 2 weeks
[27], and trough levels at this time for patients on regular systemic ther-
apy of 40 mg every 2 weeks averaged ~5μg/ml in patients with rheuma-
toid arthritis and 610μg/ml in patients with psoriatic arthropathy not
treated with concomitant methotrexate [27]. This would suggest that
the kinetics of absorption of adalimumab when injected into the nod-
ules of DD may be similar to when the drug is administered subcutane-
ously. We would postulate that formulations that retard dissipation of
adalimumab from the site of injection are likely to be more effective
and may need to be administered less frequently for the treatment of
localised brotic conditions characterised by low-grade inammation
such as DD. Neutralising antibodies to adalimumab are likely to result
in reduced efcacy [28]. Only one patient, who received 35 mg in
0.7 ml, had antibody levels (12.4±0.63 AU/ml) just above the threshold
of signicance of 12Au/ml [22], which are unlikely to be of clinical sig-
nicance. Neutralising antibodies may occur more commonly after re-
peated administration of adalimumab, especially in patients who are
not on concomitant methotrexate.
Whilst almost all patients reported pain during injection, pain scores
immediately after injection were higher in participants receiving volumes
of either placebo or adalimumab N0.30.4 ml and in those injected with
the formulation of adalimumab comprising 40 mg in 0.8 ml of carrier
that was used for the 15 mg and 35 mg cohorts. This formulation differs
signicantly from the more concentrated form of 40 mg in 0.4 ml in
that it contains a variety of excipients [27], including citrate, which is as-
sociated with higher pain scores when injected subcutaneously [29]. The
40 mg in 0.4 ml formulation was not available commercially when the
trial opened to recruitment and therefore the study was initially based
on dose cohorts using the 40 mg in 0.8 ml formulation. Transient swelling
at the injection site was apparent in some patients receiving 0.7 ml of ei-
ther placebo or adalimumab, most likely due to extravasation of the
injected material outside the connes of the nodule. The safety prole
of adalimumab is well-known and the commonest adverse events in
patients with autoimmune disorders on long-term therapy are related
to infection [30]. One patient in our study, who suffered from type 2
diabetes mellitus received 35 mg of adalimumab developed a wound
infection 4 days following surgery that resolved following removal of
sutures and antibiotics. The infection was considered to be unrelated to
the adalimumab injection as it was a localised suture abscess away
from the site of the injected nodule. Wound infection has been reported
to occur in approximately 3.6% of patients undergoing Dupuytren's
fasciectomy [8].
Unsurprisingly, there was no difference in nodule hardness or sizeas
assessed by tonometry and ultrasound scanrespectively at 2 weeks post
injection. This trial enabled us to optimisethe methods for obtaining the
data and analysing them in preparation for follow up studies.
5. Conclusions
This phase 2a randomised trial shows that a single intranodular
injection of 40 mg adalimumab in 0.4 ml in patients with Dupuytren's
disease is safe and leads to down regulation of the myobroblast
phenotype as evidenced by reduced expression of α-SMA and type I
procollagen proteins. Having dened the most efcacious dose and
preparation and based on these positive proof of concept data we are
now proceeding with a phase 2b trial in 138 patients with early
stage Dupuytren's disease randomised 1:1 to receive 4 injections of
adalimumab or placebo at 3 month intervals and followed for a total
of 18 months from baseline [21]. Our study also illustrates the utility
287J. Nanchahal et al. / EBioMedicine 33 (2018) 282288
of using early stage brotic human tissue to elucidate novel therapeutic
targets [18] that can be translated to the clinic.
Conicts of Interest
Mrs. Ball, Dr. Davidson, Dr. Williams, Dr. Sones, Dr. McCann, Ms.
Cabrita, Dr. Swettenham, Dr. Cahoon, Dr. Copsey, Dr. Francis, Dr. Black,
Dr. Barber, Mrs. Dutton report grants from Wellcome Trust, grants
from Department of Health, other from 180 Therapeutics LP, during
the conduct of the study.
Professor Taylor reports grants from Wellcome Trust, grants from
Department of Health, other from 180 Therapeutics LP, during the con-
duct of the study; grants from UCB, personal fees from UCB, AbbVie,
Pzer, outside the submitted work.
Professor Lamb reports grants from Wellcome Trust, grants from
Department of Health, during the conduct of the study; Professor
Sarah Lamb reports grants from NIHR Health Technology Assessment
Programme during the conduct of this study.
Professor Nanchahal and Professor Feldmann report grants from
Wellcome Trust, grants from Department of Health, other from 180
Therapeutics LP, during the conduct of the study; In addition, Professor
Nanchahal has a patent PCT/EP2011/069147 issued to 180 Therapeutics,
and with Professor Feldmann a patent PCT/US2017/049691 pending, a
patent PCT/US2017/026382 pending, a patent 16759325.0 EP2017
pending, and a patent 16759326.8 EP2017 pending.
Funding Sources
We would like to acknowledge funding from the Health Innovation
Challenge Fund (HICF) (HICF-R8-433), a parallel funding partnership
between the Wellcome Trust and the Department of Health, 180 Thera-
peutics, and the National Institute for Health Research (NIHR) Oxford
Biomedical Research Centre (BRC). The views expressed are those of
the authors and not necessarily those of the NHS, the NIHR or the
Department of Health.
Author Contributions
JN, SL, MF, CB, SD and PT conceived and designed the study. DD and
NC recruited the patients. CB, DDand NC collected the clinical data. LW,
FM and MC performed the laboratory analyses. WS, SD and BC per-
formed the statistical analyses. JS, VB, JB, EAF and SL managed the trial.
Appendix A. Supplementary Data
Supplementary data to this article can be found online at https://doi.
org/10.1016/j.ebiom.2018.06.022.
References
[1] Hindocha S, McGroutherDA, Bayat A. Epidemiological evaluation of Dupuytren's dis-
ease incidence and prevalence rates in relati on to etiology. Hand (N Y) 2009;4:
25669.
[2] Reilly RM, Stern PJ, Goldfarb CA. A retrospective revie w of the management o f
Dupuytren's nodules. J Hand Surg 2005;30:10148.
[3] Engstrand C, Krevers B, Kvist J. Factors affecting functional recovery after surgery
and hand therapy in patients with Dupuytren's disease. J Hand Ther 2015;28:
255259; quiz 260.
[4] Davis TR. Surgical treatment of primary Dupuytren's contractures of the ngers in
the UK: surgeons' preferences and research priorities. J Hand Surg Eur Vol 2013;
38:835.
[5] Zhao JZ, Hadley S, Floyd E,Earp BE, Blazar PE. The impact of collagenase Clostridium
histolyticum introduction on Dupuytren treatment patterns in the United States.
J Hand Surg 2016;41:9638.
[6] Rayan GM. Dupuytren's disease: anatomy, pathology, presentation, and treatment.
Instr Course Lect 2007;56:10111.
[7] van Rijssen AL, ter Linden H, Werker PM. Five-year results of a randomized clinical
trial on treatment in Dupuytren's disease: percutaneous needle fasciotomy versus
limited fasciectomy. Plast Reconstr Surg 2012;129:46977.
[8] Crean SM, Gerber RA, Le Graverand MP, Boyd DM, Cappelleri JC. The efcacy and
safety of fasciectomy and fasciotomy for Dupuytren's contracture in European pa-
tients: a structu red review of pu blished studies. J Hand Surg Eur Vol 2011;36:
396407.
[9] Peimer CA, Wilbrand S,Gerber RA, Chapman D, Szczypa PP. Safety and tolerability of
collagenase Clostridium histolyticum and fasciectomy for Dupuytren's contracture. J
Hand Surg Eur Vol 2015;40:1419.
[10] Hurst LC, Badala mente MA, Hentz VR, et al. Injecta ble collagenase clostridium
histolyticum for Dupuytren's contracture. N Engl J Med 2009;361:96879.
[11] Ball C, Izadi D, Verjee LS, Chan J, Nanchahal J. Systematic review of non-surgical
treatments for early dupuytren's disease. BMC Musculoskelet Disord 2016;17:345.
[12] CostasB, Coleman S, Kaufman G, James R, Cohen B,Gaston RG. Efcacy and safety of
collagenase clostridium histolyticum for Dupuytren disease nodules: a randomized
controlled trial. BMC Musculoskelet Disord 2017;18:374.
[13] Darby IA, Zakuan N, Billet F, Desmouliere A. The myobroblast, a key cell in normal
and pathological tissue repair. Cell Mol Life Sci 2016;73:114557.
[14] Wynn TA, Ramalingam TR. Mechanisms of brosis: therapeutic translation for -
brotic disease. Nat Med 2012;18:102840.
[15] Hinz B, Phan SH, Thannickal VJ, Galli A, B ochaton-Piall at ML, Gabbiani G. Th e
myobroblast: one function, multiple origins. Am J Pathol 2007;170:180716.
[16] Nanchahal J, Hinz B. Strategies to overcome the hurdles to treat brosis, a major
unmet clinical need. Proc Natl Acad Sci U S A 2016;113:72913.
[17] Verjee LS, Midwood K, Davidson D, Essex D, Sandison A, Nanchahal J. Myobroblast
distribution in Dupuytren's cords: correlation with digital contracture. J Hand Surg
2009;34:178594.
[18] Verjee LS, Verhoekx JS, Chan JK, et al. Unraveling the signaling pathways promoting
brosis inDupuytren's diseasereveals TNF as a therapeutic target.Proc Natl Acad Sci
U S A 2013;110:E9 2837.
[19] Dolmans GH, Werker PM, Hennies HC, et al. Wnt signaling and Dupuytren's disease.
N Engl J Med 2011;365:30717.
[20] Ng M, Thakkar D, Southam L, et al. A genome-wide association study of dupuytren
disease reveals 17 additional variants implicated in brosis. Am J Hum Genet
2017;101:41727.
[21] Nanchahal J, Ball C, Swettenham J, et al. Study protocol: a multi-Centre, doubleblind,
randomised, placebo-controlled, parallel group, phase II trial (RIDD) to determine
the efcacy of intra-nodular injection of anti-TNF to control disease progression in
early Dupuytren's disease, with an embedded dose response study. Wellcome
Open Res 2017. https://wellcomeopenresearch.org/articles/2-37/v2.
[22] Bartelds GM, Krieckaert CL, Nurmohamed MT, et al. Development of antidrug anti-
bodies against adalimumab and association with disease activity and treatment fail-
ure during long-term follow-up. JAMA 2011;305:14608.
[23] Canty EG, Kadler KE. Procollagen trafcking, processing and brillogenesis. J Cell Sci
2005;118:134153.
[24] Lam WL, Rawlins JM, Karoo RO, Naylor I, Sharpe DT. Re-visiting Luck's classication:
a histological analysis of Dupuytren's disease. J Hand Surg Eur Vol 2010;35:3127.
[25] Verjee LS, Midwood K, Davidson D, Eastwood M, Nanchahal J. Post-transcriptional
regulation of alpha-smooth muscle actin determines the contractile phenotype of
Dupuytren's nodular cells. J Cell Physiol 2010;224:68190.
[26] Stefanovic B. RNA protein interactions governing expression of the most abundant
protein in human body, type I collagen. Wiley Interdiscip Rev RNA 2013;4:53545.
[27] Abbvie. http://www.rxabbvie.com/pdf/humira.pdf;2018.
[28] ChenDY, Chen YM, Tsai WC, et al. Signicantassociations of antidrug antibodylevels
with serum drug trough levels and the rapeutic respon se of adalimumab and
etanercept treatment in rheumatoid arthritis. Ann Rheum Dis 2015;74:e16.
[29] Laursen T, Hansen B, FiskerS. Pain perception aftersubcutaneous injections of media
containing different buffers. Basic Clin Pharmacol Toxicol 2006;98:21821.
[30] Burmester GR, Panaccione R, Gordon KB, McIlraith MJ, Lacerda AP. Adalimumab:
long-termsafety in 23 458 patients fromglobal clinical trialsin rheumatoid arthritis,
juvenile idiopathic arthritis, ankylosing spondylitis, psoriatic arthritis, psoriasis and
Crohn's disease. Ann Rheum Dis 2013;72:51724.
288 J. Nanchahal et al. / EBioMedicine 33 (2018) 282288
... 17 In a dose ranging phase 2a study, we found that intranodular injection of 40 mg adalimumab in 0·4 mL resulted in down-regulation of the myofibroblast phenotype as characterised by reduced expression of alpha-smooth muscle actin (α-SMA) and procollagen type I proteins 2 weeks after injection. 18 Here, we report the outcome of a randomised, phase 2b trial comparing four injections of adulimumab or placebo given once every 3 months in participants with early-stage Dupuytren's disease. ...
... Mean age of participants was 59·7 years (SD 10·0). Overall, baseline characteristics were similar between the two groups ( [18][19][20]. The joint adjacent to the treated nodule was considered to be the affected joint as assessed by a masked observer from baseline photographs. ...
... This builds on our previous phase 2a dose-ranging study, where we found that this preparation of adalimumab downregulated the myofibroblast phenotype as evidenced by reduced concentrations of α-SMA and procollagen type I proteins. 18 Adalimumab is usually administered every 2 weeks for systemic inflammatory disorders. Our decision to administer four doses at 3 month intervals was based on the results of an end user survey of 40 participants, 20 with early-stage and 20 with late-stage Dupuytren's disease, regarding the number of injections they would find acceptable per annum. ...
Article
Full-text available
Background Dupuytren's disease is a common fibrotic condition that causes the fingers to flex irreversibly into the palm. Treatments for late-stage disease all have limitations, and there is no approved treatment for early-stage disease. We identified tumour necrosis factor as a therapeutic target in Dupuytren's disease, and in a dose ranging trial found 40 mg adalimumab in 0·4 mL to be most efficacious. Here we aimed to assess the effects of intranodular injection of adalimumab in early-stage disease. Methods In this phase 2b, randomised, double-blind, placebo-controlled trial adults with early-stage Dupuytren's disease and an established clinically distinct nodule with a clear history of progression in the preceding 6 months were recruited from two clinical centres in the UK and were randomly assigned 1:1 to receive four injections of adalimumab or saline every 3 months. Participants and assessors were masked. The primary outcome was nodule hardness measured with a durometer at 12 months. Data were analysed by linear mixed effects regression models in the intention-to-treat population with multiple imputation for missing primary outcome data. The trial is registered at the ISRCTN registry, ISRCTN 27786905 and is complete. Findings Between Feb 17, 2017, and Jan 11, 2019, 284 participants were screened in the UK and 140 were enrolled. 47 (34%) participants were female and 93 (66%) were male. Mean age of participants was 59·7 years (SD 10·0). Primary outcome data were available from 113 participants. Nodule hardness was lower (−4·6 AU [95% CI −7·1 to −2·2], p=0·0002) in the adalimumab compared with the saline group at 12 months. There were no related serious adverse events; the most common adverse events were minor injection site reactions. Interpretation Intranodular injections of adalimumab in participants with early-stage Dupuytren's disease resulted in softening and reduction in size of the nodules. Longer follow-up would be required to assess the effect of tumour necrosis factor inhibition on disease progression, extension deficit and hand function. Funding Health Innovation Challenge Fund (Wellcome Trust, Department of Health) and 180 Life Sciences.
... steroid injections or radiotherapy) are used to treat earlystage DD, none have been compared against other interventions in randomized controlled trials (RCTs). 5 We identified tumour necrosis factor (TNF) as a potential therapeutic target, 6,7 and the RIDD phase 2a a doseranging trial found that injecting adalimumab (40 mg in 0.4 mL) downregulated myofibroblasts, 8 the effector cells in DD. The RIDD phase 2b RCT found that adalimumab injections resulted in softening and reduction in size of early-stage progressive DD nodules, which continued to decrease further for nine months after the final injection. ...
... 9 Furthermore, when the nodules affected the proximal interphalangeal joint, placebo group participants developed greater flexion deformity. 9 Taken together with evidence that anti-TNF downregulates myofibroblast activity in vitro 6,7 and in vivo, 8 our data suggest that intranodular injections of adalimumab are likely to control disease progression. 9 Further follow-up would be needed to test the assumption that patients who are quiescent after the first course of treatment will remain quiescent following retreatment. ...
Article
Aims: To estimate the potential cost-effectiveness of adalimumab compared with standard care alone for the treatment of early-stage Dupuytren’s disease (DD) and the value of further research from an NHS perspective. Methods: We used data from the Repurposing anti-TNF for Dupuytren’s disease (RIDD) randomized controlled trial of intranodular adalimumab injections in patients with early-stage progressive DD. RIDD found that intranodular adalimumab injections reduced nodule hardness and size in patients with early-stage DD, indicating the potential to control disease progression. A within-trial cost-utility analysis compared four adalimumab injections with no further treatment against standard care alone, taking a 12-month time horizon and using prospective data on EuroQol five-dimension five-level questionnaire (EQ-5D-5L) and resource use from the RIDD trial. We also developed a patient-level simulation model similar to a Markov model to extrapolate trial outcomes over a lifetime using data from the RIDD trial and a literature review. This also evaluated repeated courses of adalimumab each time the nodule reactivated (every three years) in patients who initially responded. Results: The within-trial economic evaluation found that adalimumab plus standard care cost £503,410 per quality-adjusted life year (QALY) gained versus standard care alone over a 12-month time horizon. The model-based extrapolation suggested that, over a lifetime, repeated courses of adalimumab could cost £14,593 (95% confidence interval £7,534 to £42,698) per QALY gained versus standard care alone. If the NHS was willing to pay £20,000/QALY gained, there is a 77% probability that adalimumab with retreatment is the best value for money. Conclusion: Repeated courses of adalimumab are likely to be a cost-effective treatment for progressive early-stage DD. The value of perfect parameter information that would eliminate all uncertainty around the parameters estimated in RIDD and the duration of quiescence was estimated to be £105 per patient or £272 million for all 2,584,411 prevalent cases in the UK.
... The excised nodules were then analysed for markers of myofibroblast activity. Only 40 mg of adalimumab in 0.4 mL was found to lead to significant downregulation of a-smooth muscle actin and procollagen type I proteins; 15 mg of adalimumab in 0.3 mL and 35 mg in 0.7 mL were not efficacious (Nanchahal et al., 2018). We noted that some of the material extravasated out of the nodule into the subcutaneous tissues when we injected a volume of 0.7 mL, suggesting that high local doses would be the most efficacious. ...
... Taken together, the in vitro (Izadi et al., 2019;Verjee et al., 2013) and phase 2a trial data (Nanchahal et al., 2018) show that anti-TNF downregulates the phenotype of myofibroblasts from Dupuytren's nodules. The phase 2b clinical trial data suggest that intranodular injections of adalimumab may be efficacious in delaying or preventing the progression of early-stage Dupuyten's disease although follow-up over approximately 10 years would be required to confirm this (Nanchahal et al., 2022). ...
Article
Full-text available
Current treatments for Dupuytren's disease are limited to late-stage disease when patients have developed flexion contractures and have impaired hand function. They all have limitations, including the risk of recurrence and complications. The use of treatments for early-stage disease, such as intralesional steroid injections or radiotherapy which lack a clear biological basis or evidence of effectiveness based robust randomized, double blind, placebo-controlled trials, highlights the desire of patients to access treatments before they develop significant flexion contractures. A detailed understanding of the cellular landscape and molecular signalling in nodules of early-stage disease would permit the identification of potential therapeutic targets. This approach led to the identification of tumour necrosis factor (TNF) as a target. A phase 2a clinical trial identified 40 mg in 0.4 mL adalimumab as the most efficacious dose and a subsequent randomized, double blind, placebo-controlled phase 2b trial showed that four intranodular injections at 3-month intervals resulted in decrease in nodule hardness and size on ultrasound scan at 12 months, and both parameters continued to decrease further at 18 months, 9 months after the final injection. This type of approach provides clinicians with a robust evidence base for advising their patients.
... Teljesgenomasszociációs vizsgálattal végzett kutatások identifikálták a betegség kialakulásában szerepet játszó géneket, melyek a szöveti differenciálódást szabályozó Wnt jelátviteli út kóros működéséért felelősek, és a patológiás extracellulárismátrix-képződés hátterében állnak [11,12]. Az autoimmun hátterű betegségek gyakran hasonló mechanizmusú gyulladásos folyamatok következményeként jönnek létre [13], ezért nem meglepő, hogy a biológiai terápiás kezelések (anti-TNFα-terápia) mind a gyulladásos bélbetegségek, mind a Dupuytren-kontraktúra kezelésében eredményesek lehetnek [13,14]. ...
... A betegséget megelőzni jelen ismereteink szerint nem lehet, napjaink intenzív kutatásainak ellenére oki terápia még nem áll rendelkezésünkre [14]. Kezelése alapvetően sebészi, a leggyakrabban végzett műtét a parciális aponeurectomia. ...
Article
Full-text available
Introduction: Limited fasciectomy is the gold-standard treatment in Dupuytren's surgery. The anatomical position of digital nerves can be altered by Dupuytren's tissue resulting in a difficult dissection and localization, with a relatively high risk of iatrogenic nerve injury. This risk could be decreased by using intraoperative neural marking to facilitate locating the potentially displaced nerves. We recently demonstrated in an animal model that in vivo nerve staining with methylene blue is a suitable method to mark nerves without damaging them. Objective: We aimed to test the efficacy of our methylene blue nerve staining technique developed in a rat sciatic nerve model on human cadaveric digital nerves. Method: First, we performed epineural staining using 40 µl 1 : 80 diluted methylene blue solution on four human cadaver digital nerves fixed with formalin. In the second experiment, we stained six cadaver digital nerves without previous fixation. To increase the length of the stained segments, we used 200 µl solution on two nerves. Results: The epineural nerve labeling was not successful on formalin-fixed tissues. However, nerves without fixation were successfully stained with methylene blue. Forty µl methylene blue solution marked a 13 mm long segment, while 200 µl stained a 18 mm long segment. Conclusion: The epineural methylene blue nerve staining is limited on formalin-fixed digital nerves due tissue shrinkage. Non-fixed nerves with preserved histological structure can be stained in an 18 mm long segment. Further studies are necessary to determine the technique's value in hand surgery by testing digital nerves surrounded by Dupuytren's and scar tissues. Orv Hetil. 2022; 163(46): 1834-1839.
... Management of Dupuytren's contracture includes observation, non-operative management, and operative management (10). Non-operative management includes physiotherapy, radiation, steroids, anti-tumor necrosis factor agents and clostridium histolyticum collagenase (11)(12)(13)(14)(15). ...
... Dupuytren's disease (DD) is a common fibrotic hand condition affecting 12% of 55-year-olds and 29% of 75-year-olds in Western populations. 1 Early-stage DD presents as nodules on the palmar aspect of the hand that progress to form cords. The cords of late-stage DD cause curling of the finger joints (flexion deformity), impairing hand function and quality of life. 2 Treatment options for late-stage DD include surgical excision, needle fasciotomy, and collagenase injections. ...
Article
Full-text available
Aims: To estimate the potential cost-effectiveness of adalimumab compared with standard care alone for the treatment of early-stage Dupuytren's disease (DD) and the value of further research from an NHS perspective. Methods: We used data from the Repurposing anti-TNF for Dupuytren's disease (RIDD) randomized controlled trial of intranodular adalimumab injections in patients with early-stage progressive DD. RIDD found that intranodular adalimumab injections reduced nodule hardness and size in patients with early-stage DD, indicating the potential to control disease progression. A within-trial cost-utility analysis compared four adalimumab injections with no further treatment against standard care alone, taking a 12-month time horizon and using prospective data on EuroQol five-dimension five-level questionnaire (EQ-5D-5L) and resource use from the RIDD trial. We also developed a patient-level simulation model similar to a Markov model to extrapolate trial outcomes over a lifetime using data from the RIDD trial and a literature review. This also evaluated repeated courses of adalimumab each time the nodule reactivated (every three years) in patients who initially responded. Results: The within-trial economic evaluation found that adalimumab plus standard care cost £503,410 per quality-adjusted life year (QALY) gained versus standard care alone over a 12-month time horizon. The model-based extrapolation suggested that, over a lifetime, repeated courses of adalimumab could cost £14,593 (95% confidence interval £7,534 to £42,698) per QALY gained versus standard care alone. If the NHS was willing to pay £20,000/QALY gained, there is a 77% probability that adalimumab with retreatment is the best value for money. Conclusion: Repeated courses of adalimumab are likely to be a cost-effective treatment for progressive early-stage DD. The value of perfect parameter information that would eliminate all uncertainty around the parameters estimated in RIDD and the duration of quiescence was estimated to be £105 per patient or £272 million for all 2,584,411 prevalent cases in the UK. Cite this article: Bone Jt Open 2022;3(11):898-906.
Article
Objectives: The Anti-Freaze-F trial will assess the feasibility of conducting a large randomised controlled trial to assess whether intra-articular injection of anti-TNF (adalimumab) can reduce pain and improve function in people with pain predominant early stage frozen shoulder. Methods and analysis: We are conducting a multi-centre, randomised feasibility study, with an embedded qualitative sub-study. We will recruit adults ≥18 years with a new episode of shoulder pain attributable to early stage frozen shoulder, recruited from at least five UK NHS musculoskeletal and related physiotherapy services. Participants (n=84) will be randomised (centralised computer generated 1:1 allocation) to receive either: 1) intra-articular injection of anti-TNF (adalimumab 160mg) or 2) placebo injection (saline [0.9% sodium chloride]), both under ultrasound guidance. A second injection of the allocated treatment (adalimumab 80mg) or equivalent volume of placebo will be administered 2-3 weeks later. All participants will receive a physiotherapy advice leaflet providing education and advice about frozen shoulder and pain management. The primary feasibility objectives are: 1) the ability to screen and identify potential participants with pain predominant early stage frozen shoulder; 2) willingness of eligible participants to consent and be randomised to intervention; 3) practicalities of delivering the intervention, including time to first injection and number of participants receiving second injection; 4) standard deviation of the Shoulder Pain and Disability Index (SPADI) score and attrition rate at 3 months from baseline in order to estimate the sample size for a definitive trial. We will also assess follow up rates and viability of patient-reported outcome measures and range of shoulder motion for a definitive trial. Research Ethics Committee approval (REC 21/NE/0214). Trial registration number: ISRCTN 27075727; EudraCT number: 2021-003509-23; ClinicalTrials.gov NCT05299242.
Article
Dupuytren disease is a benign, progressive fibroproliferative disorder of the hands. To date, only one pharmacotherapy (clostridial collagenase) has been approved for use in Dupuytren disease. There is a great need for additional nonsurgical methods that can be used to either avoid the risks of invasive treatments or help minimize recurrence rates following treatment. A number of nonsurgical modalities have been discussed in the past and continue to appear in discussions among hand surgeons, despite highly variable and often poor or no long-term clinical data. This article reviews many of the pharmacotherapies discussed in the treatment of Dupuytren disease and novel therapies used in inflammation and fibrosis that offer potential treatment options.
Article
Talipes equinovarus congenitus (clubfoot) is frequently defined as a stiff, contracted deformity, but few studies have described the tissue from the point of view of the extracellular matrix, and none have quantified its mechanical properties. Several researchers have observed that clubfoot exhibits signs of fibrosis in the medial side of the deformity that are absent in the lateral side. Our study aims to quantify the differences between the medial and lateral side tissue obtained from relapsed clubfoot during surgery in terms of the morphological and mechanical properties of the tissue. Combining methods of optical and atomic force microscopy, our study revealed that the medial side has a higher Young’s modulus, contains more collagen and less adipose tissue and that the collagen fibers propagate at a higher frequency of the crimp pattern after surgical dissection of the tissue. Our study offers a multi-correlative approach that thoroughly investigates the relapsed clubfoot tissue.
Article
We review the biology of Dupuytren's disease (DD), a common localised fibrotic disorder of the hand. The disease develops through a complex interplay of genetic and environmental factors, and epigenetic signalling. The early-stage disease nodules comprise a complex milieu of stromal and immune cells which interact to promote disease development. Recently, inhibition of tumour necrosis factor (TNF) locally resulted in softening and a decrease in nodule size, potentially controlling disease progression. Unlike fibrotic disorders of the visceral organs, the easy access to tissue in DD patients enables dissection of the cellular landscape and molecular signalling pathways. In addition, the study of DD may have wider benefits in enhancing our understanding of less-accessible fibrotic tissues.
Article
Full-text available
Individuals with Dupuytren disease (DD) are commonly seen by physicians and surgeons across multiple specialties. It is an increasingly common and disabling fibroproliferative disorder of the palmar fascia, which leads to flexion contractures of the digits, and is associated with other tissue-specific fibroses. DD affects between 5% and 25% of people of European descent and is the most common inherited disease of connective tissue. We undertook the largest GWAS to date in individuals with a surgically validated diagnosis of DD from the UK, with replication in British, Dutch, and German individuals. We validated association at all nine previously described signals and discovered 17 additional variants with p ≤ 5 × 10⁻⁸. As a proof of principle, we demonstrated correlation of the high-risk genotype at the statistically most strongly associated variant with decreased secretion of the soluble WNT-antagonist SFRP4, in surgical specimen-derived DD myofibroblasts. These results highlight important pathways involved in the pathogenesis of fibrosis, including WNT signaling, extracellular matrix modulation, and inflammation. In addition, many associated loci contain genes that were hitherto unrecognized as playing a role in fibrosis, opening up new avenues of research that may lead to novel treatments for DD and fibrosis more generally. DD represents an ideal human model disease for fibrosis research.
Article
Full-text available
Background To determine the safety and efficacy of collagenase clostridium histolyticum (CCH) injection for the treatment of palmar Dupuytren disease nodules. Methods In this 8-week, double-blind trial, palpable palmar nodules on one hand of adults with Dupuytren disease were selected for treatment. Patients were randomly assigned using an interactive web response system to receive a dose of 0.25 mg, 0.40 mg, or 0.60 mg (1:1:1 ratio) and then allocated to active treatment (CCH) or placebo (4:1 ratio). All patients and investigators were blinded to treatment. One injection was made in the selected nodule on Day 1. Caliper measurements of nodule length and width were performed at screening and at Weeks 4 and 8. Investigator-reported nodular consistency and hardness were evaluated at baseline and Weeks 1, 4, and 8. Investigator-rated patient improvement (1 [very much improved] to 7 [very much worse]) and patient satisfaction were assessed at study end. ResultsIn the efficacy population (n = 74), percentage changes in area were significantly greater with CCH 0.40 mg (−80.1%, P = 0.0002) and CCH 0.60 mg (−78.2%, P = 0.0003), but not CCH 0.25 mg (−58.3%, P = 0.079), versus placebo (−42.2%) at post-treatment Week 8. Mean change in nodular consistency and hardness were significantly improved with CCH versus placebo at Weeks 4 and 8 (P ≤ 0.0139 for all). At Week 8, investigator global assessment of improvement was significantly greater with CCH 0.40 mg and 0.60 mg (P ≤ 0.0014) but not statistically significant with CCH 0.25 mg versus placebo (P = 0.13). Most patients were “very satisfied” or “quite satisfied” with CCH 0.40 mg and 0.60 mg. Contusion/bruising (50.0% to 59.1%) was the most common adverse event with CCH treatment. Conclusion In patients with Dupuytren disease, a single CCH injection significantly improved palmar nodule size and hardness. The safety of CCH was similar to that observed previously in patients with Dupuytren contracture. Trial registrationClinicalTrials.gov identifier: NCT02193828. Date of trial registration: July 2, 2014 to December 5, 2014
Article
Full-text available
Dupuytren's disease is a common fibrotic condition of the hand affecting 4% of the population and causes the fingers to curl irreversibly into the palm. It has a strong familial tendency, there is no approved treatment for early stage disease, and patients with established digital contractures are most commonly treated by surgery. This is associated with prolonged recovery, and less invasive techniques have high recurrence rates.The myofibroblasts, the cells responsible for the excessive matrix deposition and contraction, are aggregated in nodules. Using excised diseased and control human tissue, we found that immune cells interspersed amongst the myofibroblasts secrete cytokines. Of these, only tumour necrosis factor (TNF) promoted the development of myofibroblasts. The clinically approved anti-TNF agents led to inhibition of the myofibroblast phenotype in vitro. This clinical trial is designed to assess the efficacy of the anti-TNF agent adalimumab on participants with early disease. The first part is a dose-ranging study where nodules of participants already scheduled for surgery will be injected with either placebo (saline) or varying doses of adalimumab. The excised tissue will then be analysed for markers of myofibroblast activity.The second part of the study will recruit participants with early stage disease. They will be randomised 1: 1 to receive either adalimumab or placebo at 3 month intervals over 1 year and will then be followed for a further 6 months. Outcome measures will include nodule hardness, size and disease progression. The trial will also determine the cost-effectiveness of adalimumb treatment for this group of participants.
Article
Full-text available
Background Dupuytren’s disease is a common fibrotic disorder of the palm characterized by the development of progressive flexion deformities in the digits, leading to significant functional impairment. Surgical excision remains the most common treatment. However, this is only indicated in patients with established contractures rather than those with early disease. Early disease is generally characterized by the presence of palmar nodules with limited or no contracture of the fingers. The ideal treatment would be directed at patients with early progressive disease to prevent future deterioration. Various non-surgical treatment modalities have been described but there is currently no systematic assessment of the role and efficacy of these treatments in patients with early disease. Methods Using a PICOS analysis we reviewed publications of studies of patients with early disease who had received physical therapies, pharmacological treatment, or radiotherapy. Following PRISMA guidelines titles and abstract were screened using predefined criteria to identify those reporting outcomes specifically relating to the treatment of early disease. In the absence of a definition of early disease studies were included if early DD was described clinically, with digital contractures not exceeding 30°, Tubiana grades N to 1, and which reported identifiable data. Studies were excluded if data for early DD patients could not be extracted for analysis. ResultsIn this systematic review, 26 studies were identified and analyzed to evaluate the effect of pharmacological therapy (n = 11), physical therapy (n = 5) and radiotherapy (n = 10) on early Dupuytren’s disease. The studies comprised 20 case series, 1 cohort study with the remainder reporting case studies. All publications were graded level of evidence 4 or 5 assessed using the Oxford Centre for Evidence Based Medicine grading. Narrative descriptions of the data are presented. Conclusions Physical therapies were the most robustly assessed, using objective measures but the studies were under powered, providing insufficient evidence of efficacy. Intralesional steroid injection and radiotherapy appeared to lead to softening of nodules and to retard disease progression but lacked rigorous evaluation and studies were poorly designed. There is an urgent need for adequately powered double blinded randomized trials for this common disorder which affects 4 % of the population. Trial registrationThe protocol was registered (CRD42015008986 16 November 2015) with the PROSPERO international prospective register of systematic reviews.
Article
Full-text available
Myofibroblasts are characterized by their expression of α-smooth muscle actin, their enhanced contractility when compared to normal fibroblasts and their increased synthetic activity of extracellular matrix proteins. Myofibroblasts play an important role in normal tissue repair processes, particularly in the skin where they were first described. During normal tissue repair, they appear transiently and are then lost via apoptosis. However, the chronic presence and continued activity of myofibroblasts characterize many fibrotic pathologies, in the skin and internal organs including the liver, kidney and lung. More recently, it has become clear that myofibroblasts also play a role in many types of cancer as stromal or cancer-associated myofibroblast. The fact that myofibroblasts are now known to be key players in many pathologies makes understanding their functions, origin and the regulation of their differentiation important to enable them to be regulated in normal physiology and targeted in fibrosis, scarring and cancer.
Article
Dupuytren's disease is a heterogenous condition for which a palette of treatment options is required. Randomized control trial evidence is sparse; design challenges, such as validated outcome measures, blinding, equipoise, funding and assessment of recurrence, may limit further data accrual. Recurrence has different significance with different treatments and so rates are not directly comparable. The risk of any treatment is a function of both the chance of a complication and the clinical sequelae of that complication. The patient must be intimately involved in choosing treatment and is often trading rapid recovery for a higher chance of recurrence. Health economies are strained and as custodians of healthcare, surgeons should consider whether many patients even need treatment. To minimize the chance of complex, hazardous and expensive revision surgery, a low threshold for primary skin grafting should be applied, especially for those who are young, have dense disease or vulnerable genes.
Article
Purpose: The U.S. Food and Drug Administration approved the use of collagenase Clostridium histolyticum (CCH) in the United States in February 2010. This study addresses the impact of that approval on the number of Dupuytren contracture (DC) encounters and treatment patterns in the United States. Methods: Using the Intercontinental Marketing Services Health Office-Based Medical Claims database, we identified the monthly number of DC encounters and DC procedures between January 2007 and December 2013. Collagenase Clostridium histolyticum usage data from March 2010 to December 2013 was derived from the U.S. CCH manufacturer's data warehouse. Using the combined data, the yearly increasing trends in DC encounters and treatment volume were compared before and after the introduction of CCH. Time trends in the relative procedure frequencies were then examined. Finally, the presence of seasonal variation was tested for in each treatment type. Results: Dupuytren contracture encounters increased on average by 19,015 per year between 2007 and 2009, whereas between 2011 and 2013, DC encounters increased on average by 34,940 per year. In terms of absolute procedure counts, the surgery trend line began decreasing in 2010 with the release of CCH. Meanwhile, CCH continuously increased between 2010 and 2013, and needle aponeurotomy (NA) remained relatively stable. By the year 2013, minimally invasive techniques (NA and CCH) comprised 39% of all treatment, compared with only 14% in 2007. Lastly, there was a statistically significant seasonal increase in the number of surgical procedures during the wintertime but no seasonal variation in NA or CCH. Conclusions: After the introduction of CCH, the number of Dupuytren encounters increased at a greater annual rate. The introduction and growth of CCH coincided with a decrease in surgery. The number of NA procedures remained steady throughout the study period. The number of open surgery cases followed a predictable seasonal variation with more procedures during the winter months, but this seasonal variation was not seen with less invasive techniques. Type of study/level of evidence: Economic/Decision Analysis II.
Article
Purpose: To establish patient satisfaction after collagenase clostridium histolyticum (CCH) injection. Methods: In a cross-sectional study, 213 patients who had been treated for Dupuytren disease with CCH were reviewed between 37 and 1421 days after injection. Results: A total of 73% of the patients were very satisfied or satisfied, and 21% were dissatisfied; 75% would probably or definitely have CCH again, whereas 17% probably or definitely would not. We found that satisfaction and willingness to undergo a second treatment decreased over time and had a negative relationship with recurrence. Dissatisfaction was greater in those with a poor initial outcome but not in those with an initial complication. Of 212 patients, 78 had previously experienced surgery for Dupuytren disease of whom 71% would prefer CCH to surgery and 15% the converse. Satisfaction shows a relationship with function as measured by both QuickDASH and the Southampton Dupuytren Scoring Scheme. Conclusions: Patient satisfaction with CCH is generally high but deteriorates over time as the disease recurs. To manage patient expectation, this issue should be made explicit to patients in the consent process. Clinical relevance: Overall satisfaction with CCH is high, with initial satisfaction rates especially good. Forewarning of complications and recurrence can help maintain satisfaction levels.