Advertisement
Current Concepts|Articles in Press

Pharmacotherapies in Dupuytren Disease: Current and Novel Strategies

  • Alex G. Lambi
    Correspondence
    Corresponding author: Alex G. Lambi, MD, PhD, Department of Orthopedics & Rehabilitation, University of New Mexico School of Medicine, MSC08 4720 1 UNM, Albuquerque, NM 87131.
    Affiliations
    Department of Orthopedics and Rehabilitation, University of New Mexico School of Medicine, Albuquerque, NM
    Search for articles by this author
  • Steven N. Popoff
    Affiliations
    Department of Orthopaedic Surgery, Lewis Katz School of Medicine at Temple University, Philadelphia, PA

    Department of Biomedical Education and Data Science, Lewis Katz School of Medicine at Temple University, Philadelphia, PA
    Search for articles by this author
  • Prosper Benhaim
    Affiliations
    Department of Orthopaedic Surgery, University of California Los Angeles, Los Angeles, CA
    Search for articles by this author
  • Mary F. Barbe
    Affiliations
    Department of Biomedical Education and Data Science, Lewis Katz School of Medicine at Temple University, Philadelphia, PA

    Center for Translational Medicine, Lewis Katz School of Medicine at Temple University, Philadelphia, PA
    Search for articles by this author
Open AccessPublished:March 17, 2023DOI:https://doi.org/10.1016/j.jhsa.2023.02.003
      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.

      Key words

      Our understanding of the mechanisms behind fibroproliferative disorders continues to expand. As a result, newer potential pharmacotherapies in treating diseases such as idiopathic pulmonary fibrosis, scleroderma, Peyronie disease, and Dupuytren disease (DD) are being identified. There has also been renewed interest in repurposing medications already approved by the US Food and Drug Administration (FDA) for the treatment of fibrotic disorders.
      • Karatzas E.
      • Kakouri A.C.
      • Kolios G.
      • Delis A.
      • Spyrou G.M.
      Fibrotic expression profile analysis reveals repurposed drugs with potential anti-fibrotic mode of action.
      • Rosenbloom J.
      • Mendoza F.A.
      • Jimenez S.A.
      Strategies for anti-fibrotic therapies.
      • Tai Y.
      • Woods E.L.
      • Dally J.
      • et al.
      Myofibroblasts: function, formation, and scope of molecular therapies for skin fibrosis.
      Despite this, the mainstay of treatment for patients with DD is largely surgical. The two most common nonsurgical treatments are local collagenase injections, for enzymatic degradation, and local corticosteroid injections, to reduce inflammatory processes. There is a need for additional pharmacotherapeutic options that are directed toward halting early disease progression or following treatment to prevent recurrence.
      The purpose of this article is to provide a summary of currently used and potential new pharmacotherapies in DD. A brief description of the pathophysiology, key signaling cascades, and natural history of the disease are included to set the stage for the need for additional nonsurgical treatments. This article covers many of the medications that have been considered for a repurposed use in DD over the years. Several newer monoclonal antibodies already in use are discussed for their potential antifibrotic and anti-inflammatory role.

      Natural History

      The typical patient presenting for symptomatic DD demonstrates contracture of the ulnar digits of the hand, with the long and ring fingers most often affected. The Hueston table-top test, considered positive when a patient can no longer place their hand and fingers flat on a table, can serve as an indication for intervention. Specific degrees of flexion contraction of 30° at the MCP joint and 15° at the PIP joint also serve as thresholds for the provision of an intervention.
      • Mella J.R.
      • Guo L.
      • Hung V.
      Dupuytren’s contracture: an evidence based review.
      As contractures progress, impairment of hand function ensues, with 53° of MCP joint contracture and 77° of PIP joint contracture indicative of critical impairments in hand function.
      • Raymond A.
      • Parry M.
      • Amirfeyz R.
      Critical angles of deformity in Dupuytren’s contracture of the little and ring fingers.
      Current interventional strategies include fasciectomy, percutaneous needle fasciotomy (PNF), and enzymatic digestion. Fasciectomy techniques, including limited fasciectomy and dermatofasciectomy, remain the gold standard, since they have demonstrated high clinical efficacy and low recurrence rates.
      • Mella J.R.
      • Guo L.
      • Hung V.
      Dupuytren’s contracture: an evidence based review.
      ,
      • Denkler K.A.
      • Vaughn C.J.
      • Dolan E.L.
      • Hansen S.L.
      Evidence-based medicine: options for Dupuytren’s contracture: incise, excise, and dissolve.
      ,
      • Shih B.
      • Bayat A.
      Scientific understanding and clinical management of Dupuytren disease.
      Nevertheless, because of their high complication rate as a consequence of their invasiveness, attention has been paid to minimally invasive techniques, such as PNF and enzymatic digestion.

      Pathophysiology

      DD is a benign fibroproliferative disorder that affects the palmar fascia of the hand and digits. Its clinical course can involve progressive and symptomatic contractures of the hand and digits, leading to decreased hand function and diminishing quality of life.
      • Shih B.
      • Bayat A.
      Scientific understanding and clinical management of Dupuytren disease.
      The natural history of DD can be divided into 3 histologic stages, as initially described by Luck.
      • Luck J.V.
      Dupuytren’s contracture; a new concept of the pathogenesis correlated with surgical management.
      Stage I, the proliferative phase, is classically characterized by nodule formation within the palmar fascia as well as increased fibroblast activity. Myofibroblasts comprise the majority of cells in the nodule in this phase. Stage II, the involutional phase, is noted by marked nodular thickening and an increase in underlying type III collagen synthesis that becomes oriented along the lines of tension within the palm. Early joint contracture can be seen during this phase. Stage III, the residual phase, is characterized by a large disappearance of myofibroblasts and the replacement of type III collagen with type I collagen (Fig. 1).
      • Zhang A.Y.
      • Kargel J.S.
      The basic science of Dupuytren disease.
      Figure thumbnail gr1
      Figure 1Pathophysiology of Dupuytren contracture. Dupuytren disease progression exists on histologic, cellular, and clinical levels. Resting tissue of the unaffected hand contains quiescent fibroblasts. Upon tissue injury or stress, fibroblasts are activated, proliferate, and differentiate into mature myofibroblasts (proliferative stage). With continued TGF-β stimulation and Dupuytren risk factors, disease progresses. Nodule formation occurs as myofibroblasts differentiate and produce extracellular matrix (ECM), with collagen III:I ratio predominating (Involutional stage). The ECM collagen ratio changes to I:III, increased collagen crosslinking occurs, and cellularity decreases as cords form and contraction ensues (residual stage). TGF, transforming growth factor.
      Disease progression varies between individuals and can be influenced by established risk factors, such as alcohol intake, smoking, manual labor, diabetes, anticonvulsant drugs, metabolic factors, and genetic predisposition.
      • Zhang A.Y.
      • Kargel J.S.
      The basic science of Dupuytren disease.
      • Godtfredsen N.S.
      • Lucht H.
      • Prescott E.
      • Sørensen T.I.
      • Grønbaek M.
      A prospective study linked both alcohol and tobacco to Dupuytren’s disease.
      • Burke F.D.
      • Proud G.
      • Lawson I.J.
      • McGeoch K.L.
      • Miles J.N.
      An assessment of the effects of exposure to vibration, smoking, alcohol and diabetes on the prevalence of Dupuytren’s disease in 97,537 miners.
      • Burge P.
      • Hoy G.
      • Regan P.
      • Milne R.
      Smoking, alcohol and the risk of Dupuytren’s contracture.
      • Liss G.M.
      • Stock S.R.
      Can Dupuytren’s contracture be work-related?: review of the evidence.
      • Gudmundsson K.G.
      • Arngrímsson R.
      • Sigfússon N.
      • Björnsson A.
      • Jónsson T.
      Epidemiology of Dupuytren’s disease: clinical, serological, and social assessment. The Reykjavik Study.
      • Noble J.
      • Heathcote J.G.
      • Cohen H.
      Diabetes mellitus in the aetiology of Dupuytren’s disease.
      • Dibenedetti D.B.
      • Nguyen D.
      • Zografos L.
      • Ziemiecki R.
      • Zhou X.
      Prevalence, incidence, and treatments of Dupuytren’s disease in the United States: results from a population-based study.
      • Rydberg M.
      • Zimmerman M.
      • Löfgren J.P.
      • et al.
      Metabolic factors and the risk of Dupuytren’s disease: data from 30,000 individuals followed for over 20 years.
      Despite significant research, the underlying genesis of DD has not been clearly elucidated. DD nodules are thought to originate from or near the palmar fascia via mechanisms that include trauma to the palmar fascia, altered immune responses, and/or the presence of oxygen-free radicals.
      • Shih B.
      • Bayat A.
      Scientific understanding and clinical management of Dupuytren disease.
      Additionally, the amount and composition of subcutaneous palmar fat may play a role in the progression and recurrence of DD, as lower levels of subcutaneous fat tissue have been noted in individuals with DD.
      • Rabinowitz J.L.
      • Ostermann Jr., L.
      • Bora F.W.
      • Staeffen J.
      Lipid composition and de novo lipid biosynthesis of human palmar fat in Dupuytren’s disease.
      • Bergenudd H.
      • Lindgärde F.
      • Nilsson B.E.
      Prevalence of Dupuytren’s contracture and its correlation with degenerative changes of the hands and feet and with criteria of general health.
      • Shih B.
      • Brown J.J.
      • Armstrong D.J.
      • Lindau T.
      • Bayat A.
      Differential gene expression analysis of subcutaneous fat, fascia, and skin overlying a Dupuytren’s disease nodule in comparison to control tissue.
      Although the specific mechanisms and triggers for DD development are still yet to be fully elucidated, it is well established that the cell type responsible for DD progression is the myofibroblast. Derived from fibroblasts, the myofibroblast is characterized by the co-expression of high levels of α-smooth muscle actin (α-SMA) and platelet-derived growth factor (PDGF).
      • Hinz B.
      Formation and function of the myofibroblast during tissue repair.
      The clinical contractures seen in DD most likely occur on a cellular level through a contractile apparatus of the myofibroblast containing bundles of actin microfilaments and associated contractile proteins (eg, nonmuscle myosin). Intracellular actin bundles terminate on the myofibroblast surface in the fibronexus, an adhesion complex that incorporates transmembrane integrin proteins to link the actin with extracellular matrix proteins, such as fibronectin fibrils, and adjacent myofibroblasts.
      • Tomasek J.J.
      • Schultz R.J.
      • Haaksma C.J.
      Extracellular matrix-cytoskeletal connections at the surface of the specialized contractile fibroblast (myofibroblast) in Dupuytren disease.
      • Tomasek J.J.
      • Gabbiani G.
      • Hinz B.
      • Chaponnier C.
      • Brown R.A.
      Myofibroblasts and mechano-regulation of connective tissue remodelling.
      • Singer II,
      • Kawka D.W.
      • Kazazis D.M.
      • Clark R.A.
      In vivo co-distribution of fibronectin and actin fibers in granulation tissue: immunofluorescence and electron microscope studies of the fibronexus at the myofibroblast surface.
      • Burridge K.
      • Chrzanowska-Wodnicka M.
      Focal adhesions, contractility, and signaling.
      • Dugina V.
      • Fontao L.
      • Chaponnier C.
      • Vasiliev J.
      • Gabbiani G.
      Focal adhesion features during myofibroblastic differentiation are controlled by intracellular and extracellular factors.
      Extensive research has been performed to better understand the modulators of fibroblasts and myofibroblasts in DD development. Transforming growth factor-β (TGF-β) signaling has been highlighted as critical in DD development.
      • Krause C.
      • Kloen P.
      Concurrent inhibition of TGF-beta and mitogen driven signaling cascades in Dupuytren’s disease - non-surgical treatment strategies from a signaling point of view.
      Its specific role in myofibroblast function, DD progression, and potential in treatment is discussed below.

      Transforming Growth Factor-Signaling In Dupuytren Disease Development

      Transforming growth factor-β has been implicated in DD development and progression. Three mammalian isoforms of TGF-β exist: TGF-β1, TGF-β2, and TGF-β3. All 3 isoforms have been identified in DD disease nodules, palmar fascia, and cord tissue.
      • Berndt A.
      • Kosmehl H.
      • Mandel U.
      • et al.
      TGF beta and bFGF synthesis and localization in Dupuytren’s disease (nodular palmar fibromatosis) relative to cellular activity, myofibroblast phenotype and oncofetal variants of fibronectin.
      ,
      • Baird K.S.
      • Crossan J.F.
      • Ralston S.H.
      Abnormal growth factor and cytokine expression in Dupuytren’s contracture.
      TGF-β signaling is upregulated in DD and has been shown to be expressed in fibroblasts and myofibroblasts in all 3 histologic stages of DD progression.
      • Berndt A.
      • Kosmehl H.
      • Mandel U.
      • et al.
      TGF beta and bFGF synthesis and localization in Dupuytren’s disease (nodular palmar fibromatosis) relative to cellular activity, myofibroblast phenotype and oncofetal variants of fibronectin.
      • Baird K.S.
      • Crossan J.F.
      • Ralston S.H.
      Abnormal growth factor and cytokine expression in Dupuytren’s contracture.
      • Badalamente M.A.
      • Sampson S.P.
      • Hurst L.C.
      • Dowd A.
      • Miyasaka K.
      The role of transforming growth factor beta in Dupuytren’s disease.
      • Bisson M.A.
      • McGrouther D.A.
      • Mudera V.
      • Grobbelaar A.O.
      The different characteristics of Dupuytren’s disease fibroblasts derived from either nodule or cord: expression of alpha-smooth muscle actin and the response to stimulation by TGF-beta1.
      In fibroblasts derived from either DD affected or unaffected tissues, TGF-β upregulates α-SMA expression and induces differentiation of a quiescent fibroblast to a contracting myofibroblast.
      • Zhang A.Y.
      • Kargel J.S.
      The basic science of Dupuytren disease.
      ,
      • Bisson M.A.
      • McGrouther D.A.
      • Mudera V.
      • Grobbelaar A.O.
      The different characteristics of Dupuytren’s disease fibroblasts derived from either nodule or cord: expression of alpha-smooth muscle actin and the response to stimulation by TGF-beta1.
      • Verjee L.S.
      • Verhoekx J.S.
      • Chan J.K.
      • et al.
      Unraveling the signaling pathways promoting fibrosis in Dupuytren’s disease reveals TNF as a therapeutic target.
      • Krause C.
      • Kloen P.
      • Ten Dijke P.
      Elevated transforming growth factor beta and mitogen-activated protein kinase pathways mediate fibrotic traits of Dupuytren’s disease fibroblasts.
      The addition of TGF-β in culture models leads to increased contracture of DD fibroblasts.
      • Tse R.
      • Howard J.
      • Wu Y.
      • Gan B.S.
      Enhanced Dupuytren’s disease fibroblast populated collagen lattice contraction is independent of endogenous active TGF-beta2.
      Furthermore, when TGF-β signaling is blocked in DD cells in vitro, a dose-dependent decrease in contractility with concomitant decreases in α-SMA and Col1 gene expression and α-SMA protein level are seen.
      • Verjee L.S.
      • Verhoekx J.S.
      • Chan J.K.
      • et al.
      Unraveling the signaling pathways promoting fibrosis in Dupuytren’s disease reveals TNF as a therapeutic target.
      Therefore, the ability to block the profibrotic effects of TGF-β signaling in DD is an area ripe for research and clinical potential. Several therapeutic options discussed later in this article target the TGF-β pathway.

      Challenges In Studying New Therapies In Dupuytren Disease

      To date no single, reliable animal model has been created to study the pathophysiology of DD or the disease response to therapeutics. As a result, researchers have had to rely on a few ways to study the potential efficacy of therapeutics. The first is through in vitro studies using fibroblasts isolated from Dupuytren nodules or cords in comparison to fascia overlying the carpal tunnel or the transverse carpal ligament.
      • Verjee L.S.
      • Verhoekx J.S.
      • Chan J.K.
      • et al.
      Unraveling the signaling pathways promoting fibrosis in Dupuytren’s disease reveals TNF as a therapeutic target.
      Key limitations to this approach include the fact that the palmar fascia overlying the carpal tunnel is rarely involved in DD, and the transverse carpal ligament never.
      • Satish L.
      • LaFramboise W.A.
      • Johnson S.
      • et al.
      Fibroblasts from phenotypically normal palmar fascia exhibit molecular profiles highly similar to fibroblasts from active disease in Dupuytren’s contracture.
      Furthermore, because of the paucity of cells isolated from tissue, many experiments expand their cell population through passage 5 prior to performing experiments. However, prior work has shown that by this passage the phenotypes and normal human dermal fibroblasts and mature myofibroblasts tend to converge.
      • Rehman S.
      • Xu Y.
      • Dunn W.B.
      • et al.
      Dupuytren’s disease metabolite analyses reveals alterations following initial short-term fibroblast culturing.
      ,
      • Verjee L.S.
      • 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.
      The other challenge in studying DD at the clinical level is our reliance on only clinical findings to measure therapeutic efficacy. A noninvasive test to measure the therapeutic effect on Dupuytren tissues in real time is sorely needed. Imaging modalities for monitoring other fibrotic disorders, chiefly idiopathic pulmonary fibrosis, are well described.
      • Gotway M.B.
      • Freemer M.M.
      • King Jr., T.E.
      Challenges in pulmonary fibrosis. 1: Use of high resolution CT scanning of the lung for the evaluation of patients with idiopathic interstitial pneumonias.
      Noninvasive tests that could be used to study DD are being investigated for other musculoskeletal fibroses. These include modalities such as nuclear magnetic resonance, to assess thickened tissue layers, and ultrasound shear-wave elastography, to assess tissue stiffness.
      • Martins-Bach A.B.
      • Bachasson D.
      • Araujo E.C.A.
      • et al.
      Non-invasive assessment of skeletal muscle fibrosis in mice using nuclear magnetic resonance imaging and ultrasound shear wave elastography.

      Current Pharmacotherapies Used In Dupuytren Disease Treatment

      Enzymatic digestion with collagenase

      To date, the only approved pharmacologic therapy that has shown sustained efficacy in treating DD is clostridial collagenase. The underlying mechanism by which Clostridium histolyticum collagenases produce their effect is through the degradation of the collagen found in DD contracture. In 2010, the US FDA approved C. histolyticum for injectable use under the name Xiaflex (Auxilium Pharmaceuticals, Inc.).
      • Denkler K.A.
      • Vaughn C.J.
      • Dolan E.L.
      • Hansen S.L.
      Evidence-based medicine: options for Dupuytren’s contracture: incise, excise, and dissolve.
      ,
      • Murphy A.
      • Lalonde D.H.
      • Eaton C.
      • et al.
      Minimally invasive options in Dupuytren’s contracture: aponeurotomy, enzymes, stretching, and fat grafting.
      ,
      • Badalamente M.A.
      • Hurst L.C.
      Development of collagenase treatment for Dupuytren disease.
      Xiaflex constitutes 2 purified collagenases (AUX-I and AUX-II) that preferentially degrade collagen types I and III found in DD cords, while sparing collagen types IV and VI that are predominant in vascular basement membranes and perineurium.
      • Badalamente M.A.
      • Hurst L.C.
      Development of collagenase treatment for Dupuytren disease.
      Treatment takes place over 2 stages, with the first including injection of the diseased DD cord and the second consisting of cord rupture via manual manipulation. Success has been seen in the treatment of MCP and PIP joint contracture, with higher success rates seen in reducing contracture of MCP joints (to within 5° of full extension) than for PIP joints. However, limited data exist for the use of collagenase in early DD as the safety and efficacy data included in the original submission to the FDA for approval were for flexion deformities >20°, in either MCP or PIP joints.
      • Freshwater M.F.
      What were the adverse events for Dupuytren’s patients treated with Xiaflex who had contractures less than 20°?.
      Published recurrence rates following collagenase treatment vary widely, with the most cited rate as 35% when defined as a worsening of previously treated contracture >20°.
      • Scherman P.
      • Jenmalm P.
      • Dahlin L.B.
      Three-year recurrence of Dupuytren’s contracture after needle fasciotomy and collagenase injection: a two-centre randomized controlled trial.
      With respect to recurrence rates, enzymatic treatment performs similarly to PNF when used in PIP joints and potentially outperforms PNF when used in MCP joints.
      • Skov S.T.
      • Bisgaard T.
      • Søndergaard P.
      • Lange J.
      Injectable collagenase versus percutaneous needle fasciotomy for Dupuytren contracture in proximal interphalangeal joints: A randomized controlled trial.

      Corticosteroid administration

      Corticosteroids, such as injectable triamcinolone, are a common treatment choice for patients with DD.
      • Ketchum L.D.
      The rationale for treating the nodule in Dupuytren’s disease.
      • Zachariae L.
      • Zachariae F.
      Hydrocortisone acetate in the treatment of Dupuytren’s contraction and allied conditions.
      • Ketchum L.D.
      • Donahue T.K.
      The injection of nodules of Dupuytren’s disease with triamcinolone acetonide.
      • Coste F.
      • Weissenbach R.
      [Treatment of Dupuytren’s disease by local injections of hydrocortisone].
      Corticosteroids have been shown to decrease rates of cell proliferation in DD nodules and in DD cells cultured in vitro,
      • Meek R.M.
      • McLellan S.
      • Reilly J.
      • Crossan J.F.
      The effect of steroids on Dupuytren’s disease: role of programmed cell death.
      as well as modify disease progression in patients.
      • Zachariae L.
      • Zachariae F.
      Hydrocortisone acetate in the treatment of Dupuytren’s contraction and allied conditions.
      • Ketchum L.D.
      • Donahue T.K.
      The injection of nodules of Dupuytren’s disease with triamcinolone acetonide.
      • Coste F.
      • Weissenbach R.
      [Treatment of Dupuytren’s disease by local injections of hydrocortisone].
      Triamcinolone administration leads to inhibition of TGF-β1 expression and fibroblast apoptosis.
      • Denkler K.A.
      • Vaughn C.J.
      • Dolan E.L.
      • Hansen S.L.
      Evidence-based medicine: options for Dupuytren’s contracture: incise, excise, and dissolve.
      Triamcinolone has also been shown to potentiate the activity of collagenase in vitro.
      • Ketchum L.D.
      • Robinson D.W.
      • Masters F.W.
      The degradation of mature collagen: a laboratory study.
      To our knowledge, no clinical studies have been performed examining the effect of triamcinolone as part of treatment with collagenase. It has been shown that short-term improvements in flexion deformity occur when triamcinolone injection is used in combination with PNF.
      • McMillan C.
      • Binhammer P.
      Steroid injection and needle aponeurotomy for Dupuytren contracture: a randomized, controlled study.
      However, long-term studies are needed to examine whether these effects result in significant long-term recurrence reduction.

      Repurposed Pharmacotherapies Proposed For Dupuytren Disease

      To date, several other pharmacotherapies have been proposed for off-label use in treating DD (Table 1). None have demonstrated either decreased severity or recurrence in long-term clinical trials.
      • Ball C.
      • Izadi D.
      • Verjee L.S.
      • Chan J.
      • Nanchahal J.
      Systematic review of non-surgical treatments for early Dupuytren’s disease.
      For the sake of understanding the rationale for their use they will be discussed here briefly.
      Table 1Repurposed Pharmacotherapies Proposed for Dupuytren Disease
      MedicationBasic Mechanism of DrugProposed Mechanism in DDIn Vitro Results in DD ModelIn Vivo Results in Patients with DDRef.
      Anti-inflammatory
      Corticosteroid
      • Nonspecific reduction in collagen synthesis, ECM composition, and proinflammatory mediators
        • Berman B.
        • Maderal A.
        • Raphael B.
        Keloids and hypertrophic scars: pathophysiology, classification, and treatment.
      • Decreased fibroblast activity, density, and maturation
        • Berman B.
        • Maderal A.
        • Raphael B.
        Keloids and hypertrophic scars: pathophysiology, classification, and treatment.
      • Inhibition of TGF-β1 expression and fibroblast apoptosis
        • Meek R.M.
        • McLellan S.
        • Reilly J.
        • Crossan J.F.
        The effect of steroids on Dupuytren’s disease: role of programmed cell death.
      • Potentiate activity of collagenase
        • Ketchum L.D.
        • Robinson D.W.
        • Masters F.W.
        The degradation of mature collagen: a laboratory study.
      • Zachariae L.
      • Zachariae F.
      Hydrocortisone acetate in the treatment of Dupuytren’s contraction and allied conditions.
      • Ketchum L.D.
      • Donahue T.K.
      The injection of nodules of Dupuytren’s disease with triamcinolone acetonide.
      • Coste F.
      • Weissenbach R.
      [Treatment of Dupuytren’s disease by local injections of hydrocortisone].
      • Meek R.M.
      • McLellan S.
      • Reilly J.
      • Crossan J.F.
      The effect of steroids on Dupuytren’s disease: role of programmed cell death.
      • Ketchum L.D.
      • Robinson D.W.
      • Masters F.W.
      The degradation of mature collagen: a laboratory study.
      • McMillan C.
      • Binhammer P.
      Steroid injection and needle aponeurotomy for Dupuytren contracture: a randomized, controlled study.
      ,
      • Berman B.
      • Maderal A.
      • Raphael B.
      Keloids and hypertrophic scars: pathophysiology, classification, and treatment.
      Celecoxib
      • COX-2 inhibition, downregulation of proinflammatory mediators
        • Davies N.M.
        • McLachlan A.J.
        • Day R.O.
        • Williams K.M.
        Clinical pharmacokinetics and pharmacodynamics of celecoxib: a selective cyclo-oxygenase-2 inhibitor.
      • Decreased myofibroblast differentiation through TGF-β signaling pathways
        • Salib C.G.
        • Reina N.
        • Trousdale W.H.
        • et al.
        Inhibition of COX-2 pathway as a potential prophylaxis against arthrofibrogenesis in a rabbit model of joint contracture.
        ,
        • Chen H.
        • Qian Z.
        • Zhang S.
        • et al.
        Silencing COX-2 blocks PDK1/TRAF4-induced AKT activation to inhibit fibrogenesis during skeletal muscle atrophy.
      • Not tested
      • Not tested
      • Davies N.M.
      • McLachlan A.J.
      • Day R.O.
      • Williams K.M.
      Clinical pharmacokinetics and pharmacodynamics of celecoxib: a selective cyclo-oxygenase-2 inhibitor.
      • Salib C.G.
      • Reina N.
      • Trousdale W.H.
      • et al.
      Inhibition of COX-2 pathway as a potential prophylaxis against arthrofibrogenesis in a rabbit model of joint contracture.
      • Chen H.
      • Qian Z.
      • Zhang S.
      • et al.
      Silencing COX-2 blocks PDK1/TRAF4-induced AKT activation to inhibit fibrogenesis during skeletal muscle atrophy.
      Interferon (IFN)-γ, α2b
      • Inhibit cell growth, immunomodulation
        • Baron S.
        • Tyring S.K.
        • Fleischmann W.R.
        • et al.
        The interferons. Mechanisms of action and clinical applications.
      • Decreased fibroblast proliferation, myofibroblast differentiation, and collagen production
        • Berman B.
        • Maderal A.
        • Raphael B.
        Keloids and hypertrophic scars: pathophysiology, classification, and treatment.
        • Pittet B.
        • Rubbia-Brandt L.
        • Desmoulière A.
        • et al.
        Effect of gamma-interferon on the clinical and biologic evolution of hypertrophic scars and Dupuytren’s disease: an open pilot study.
      • Decreased fibroblast proliferation and α-SMA expression
        • Pittet B.
        • Rubbia-Brandt L.
        • Desmoulière A.
        • et al.
        Effect of gamma-interferon on the clinical and biologic evolution of hypertrophic scars and Dupuytren’s disease: an open pilot study.
      • Decreased fibroblast contraction
        • Sanders J.L.
        • Dodd C.
        • Ghahary A.
        • Scott P.G.
        • Tredget E.E.
        The effect of interferon-alpha2b on an in vitro model Dupuytren’s contracture.
      • Decreased early DD nodule size
        • Pittet B.
        • Rubbia-Brandt L.
        • Desmoulière A.
        • et al.
        Effect of gamma-interferon on the clinical and biologic evolution of hypertrophic scars and Dupuytren’s disease: an open pilot study.
      • Berman B.
      • Maderal A.
      • Raphael B.
      Keloids and hypertrophic scars: pathophysiology, classification, and treatment.
      • Baron S.
      • Tyring S.K.
      • Fleischmann W.R.
      • et al.
      The interferons. Mechanisms of action and clinical applications.
      • Pittet B.
      • Rubbia-Brandt L.
      • Desmoulière A.
      • et al.
      Effect of gamma-interferon on the clinical and biologic evolution of hypertrophic scars and Dupuytren’s disease: an open pilot study.
      • Sanders J.L.
      • Dodd C.
      • Ghahary A.
      • Scott P.G.
      • Tredget E.E.
      The effect of interferon-alpha2b on an in vitro model Dupuytren’s contracture.
      Antimitotic
      5-Fluorouracil
      • Inhibit fibroblast proliferation
      • Inhibit TGF-β-induced expression of collagen
        • Berman B.
        • Maderal A.
        • Raphael B.
        Keloids and hypertrophic scars: pathophysiology, classification, and treatment.
      • Inhibition of myofibroblast proliferation and differentiation
        • Jemec B.
        • Linge C.
        • Grobbelaar A.O.
        • Smith P.J.
        • Sanders R.
        • McGrouther D.A.
        The effect of 5-fluorouracil on Dupuytren fibroblast proliferation and differentiation.
      • No clinical benefit when topically applied intraoperatively after limited fasciectomy
        • Bulstrode N.W.
        • Bisson M.
        • Jemec B.
        • Pratt A.L.
        • McGrouther D.A.
        • Grobbelaar A.O.
        A prospective randomised clinical trial of the intra-operative use of 5-fluorouracil on the outcome of Dupuytren’s disease.
        • Berman B.
        • Maderal A.
        • Raphael B.
        Keloids and hypertrophic scars: pathophysiology, classification, and treatment.
        ,
        • Werker P.M.N.
        • Degreef I.
        Alternative and adjunctive treatments for Dupuytren disease.
        • Jemec B.
        • Linge C.
        • Grobbelaar A.O.
        • Smith P.J.
        • Sanders R.
        • McGrouther D.A.
        The effect of 5-fluorouracil on Dupuytren fibroblast proliferation and differentiation.
        • Bulstrode N.W.
        • Bisson M.
        • Jemec B.
        • Pratt A.L.
        • McGrouther D.A.
        • Grobbelaar A.O.
        A prospective randomised clinical trial of the intra-operative use of 5-fluorouracil on the outcome of Dupuytren’s disease.
      Colchicine
      • Disrupts cytoskeletal functions by inhibiting β-tubulin polymerization
      • Inhibits cell proliferation by blocking mitosis
      • Decrease collagen synthesis
      • Increase collagenase activity
        • Rojkind M.
        • Uribe M.
        • Kershenobich D.
        Colchicine and the treatment of liver cirrhosis.
        ,
        • Diegelmann R.F.
        • Peterkofsky B.
        Inhibition of collagen secretion from bone and cultured fibroblasts by microtubular disruptive drugs.
      • Not tested
      • No effect on DD contracture when administered orally in Peyronie disease pts
        • Akkus E.
        • Carrier S.
        • Rehman J.
        • Breza J.
        • Kadioglu A.
        • Lue T.F.
        Is colchicine effective in Peyronie’s disease? A pilot study.
      Anti-Oxidant
      Vitamin E
      • Decreases reactive oxygen species
      • Decrease myofibroblast differentiation
        • Guy C.D.
        • Suzuki A.
        • Abdelmalek M.F.
        • Burchette J.L.
        • Diehl A.M.
        • NASH C.R.N.
        Treatment response in the PIVENS trial is associated with decreased Hedgehog pathway activity.
      • Not tested
        • Guy C.D.
        • Suzuki A.
        • Abdelmalek M.F.
        • Burchette J.L.
        • Diehl A.M.
        • NASH C.R.N.
        Treatment response in the PIVENS trial is associated with decreased Hedgehog pathway activity.
        • Richards H.J.
        Dupuytren’s contracture treated with vitamin E.
        • Steinberg C.L.
        Tocopherols in treatment of primary fibrositis; including Dupuytren’s contracture, periarthritis of the shoulders, and Peyronie’s disease.
      N-acetyl-L-cysteine (NAC)
      • Decreases reactive oxygen species
      • Downregulates TGF-β signaling
      • Decreased production of α-SMA and collagen
        • Meurer S.K.
        • Lahme B.
        • Tihaa L.
        • Weiskirchen R.
        • Gressner A.M.
        N-acetyl-L-cysteine suppresses TGF-beta signaling at distinct molecular steps: the biochemical and biological efficacy of a multifunctional, antifibrotic drug.
        ,
        • Kopp J.
        • Seyhan H.
        • Müller B.
        • et al.
        N-acetyl-L-cysteine abrogates fibrogenic properties of fibroblasts isolated from Dupuytren’s disease by blunting TGF-beta signalling.
      • Not tested
      • Not tested
        • Meurer S.K.
        • Lahme B.
        • Tihaa L.
        • Weiskirchen R.
        • Gressner A.M.
        N-acetyl-L-cysteine suppresses TGF-beta signaling at distinct molecular steps: the biochemical and biological efficacy of a multifunctional, antifibrotic drug.
        ,
        • Kopp J.
        • Seyhan H.
        • Müller B.
        • et al.
        N-acetyl-L-cysteine abrogates fibrogenic properties of fibroblasts isolated from Dupuytren’s disease by blunting TGF-beta signalling.
      Antihypertensive
      ACE inhibitors and angiotensin II antagonists
      • Disrupt renin-angiotensin-aldosterone system
        • Timmermans P.B.
        • Wong P.C.
        • Chiu A.T.
        • et al.
        Angiotensin II receptors and angiotensin II receptor antagonists.
      • Decrease TGF-β and collagen production
        • Yu L.
        • Border W.A.
        • Anderson I.
        • McCourt M.
        • Huang Y.
        • Noble N.A.
        Combining TGF-beta inhibition and angiotensin II blockade results in enhanced antifibrotic effect.
        ,
        • Zimman O.A.
        • Toblli J.
        • Stella I.
        • Ferder M.
        • Ferder L.
        • Inserra F.
        The effects of angiotensin-converting-enzyme inhibitors on the fibrous envelope around mammary implants.
      • Increased angiotensin II receptors in DD tissue
        • Stephen C.
        • Touil L.
        • Vaiude P.
        • Singh J.
        • McKirdy S.
        Angiotensin receptors in Dupuytren’s disease: a target for pharmacological treatment?.
      • Not tested
        • Timmermans P.B.
        • Wong P.C.
        • Chiu A.T.
        • et al.
        Angiotensin II receptors and angiotensin II receptor antagonists.
        • Yu L.
        • Border W.A.
        • Anderson I.
        • McCourt M.
        • Huang Y.
        • Noble N.A.
        Combining TGF-beta inhibition and angiotensin II blockade results in enhanced antifibrotic effect.
        • Zimman O.A.
        • Toblli J.
        • Stella I.
        • Ferder M.
        • Ferder L.
        • Inserra F.
        The effects of angiotensin-converting-enzyme inhibitors on the fibrous envelope around mammary implants.
        • Stephen C.
        • Touil L.
        • Vaiude P.
        • Singh J.
        • McKirdy S.
        Angiotensin receptors in Dupuytren’s disease: a target for pharmacological treatment?.
      Verapamil
      • Block voltage-gated calcium channels in cardiac nodes and vessel lining smooth muscle
      • Decrease myofibroblast-mediated contracture
        • Lee R.C.
        • Doong H.
        • Jellema A.F.
        The response of burn scars to intralesional verapamil. Report of five cases.
      • Not tested
      Vasoactive
      Phosphodiesterase inhibitors
      • Inhibits degradation of cyclic GMP by PDE5
        • Gonzalez-Cadavid N.F.
        • Rajfer J.
        Treatment of Peyronie’s disease with PDE5 inhibitors: an antifibrotic strategy.
      • Prevent TGF-β1–induced collagen formation and myofibroblast differentiation
        • Dunkern T.R.
        • Feurstein D.
        • Rossi G.A.
        • Sabatini F.
        • Hatzelmann A.
        Inhibition of TGF-beta induced lung fibroblast to myofibroblast conversion by phosphodiesterase inhibiting drugs and activators of soluble guanylyl cyclase.
        ,
        • Ferrini M.G.
        • Kovanecz I.
        • Nolazco G.
        • Rajfer J.
        • Gonzalez-Cadavid N.F.
        Effects of long-term vardenafil treatment on the development of fibrotic plaques in a rat model of Peyronie’s disease.
      • Not tested
      • Not tested
        • Gonzalez-Cadavid N.F.
        • Rajfer J.
        Treatment of Peyronie’s disease with PDE5 inhibitors: an antifibrotic strategy.
        • Dunkern T.R.
        • Feurstein D.
        • Rossi G.A.
        • Sabatini F.
        • Hatzelmann A.
        Inhibition of TGF-beta induced lung fibroblast to myofibroblast conversion by phosphodiesterase inhibiting drugs and activators of soluble guanylyl cyclase.
        • Ferrini M.G.
        • Kovanecz I.
        • Nolazco G.
        • Rajfer J.
        • Gonzalez-Cadavid N.F.
        Effects of long-term vardenafil treatment on the development of fibrotic plaques in a rat model of Peyronie’s disease.
      Nitric oxide donors
      • Increases release of nitric oxide
        • Gonzalez-Cadavid N.F.
        • Rajfer J.
        Treatment of Peyronie’s disease with PDE5 inhibitors: an antifibrotic strategy.
      • Inhibits TGF-β signaling, collagen synthesis, myofibroblast differentiation
        • Ferrini M.G.
        • Vernet D.
        • Magee T.R.
        • et al.
        Antifibrotic role of inducible nitric oxide synthase.
        ,
        • Vernet D.
        • Ferrini M.G.
        • Valente E.G.
        • et al.
        Effect of nitric oxide on the differentiation of fibroblasts into myofibroblasts in the Peyronie’s fibrotic plaque and in its rat model.
      • Not tested
      • Not tested
      Endocrine
      Tamoxifen
      • Partial agonist of estrogen receptors
      • Modulate TGF-β signaling, decrease fibroblast proliferation and collagen production
        • Chau D.
        • Mancoll J.S.
        • Lee S.
        • et al.
        Tamoxifen downregulates TGF-beta production in keloid fibroblasts.
      • Decreased TGF-β expression in DD fibroblasts, decreased fibroblast contraction
        • Kuhn M.A.
        • Wang X.
        • Payne W.G.
        • Ko F.
        • Robson M.C.
        Tamoxifen decreases fibroblast function and downregulates TGF(beta2) in Dupuytren’s affected palmar fascia.
      • Short-term improvements after limited fasciectomy, effect lost within 2 years
        • Degreef I.
        • Tejpar S.
        • Sciot R.
        • De Smet L.
        High-dosage tamoxifen as neoadjuvant treatment in minimally invasive surgery for Dupuytren disease in patients with a strong predisposition toward fibrosis: a randomized controlled trial.
        • Chau D.
        • Mancoll J.S.
        • Lee S.
        • et al.
        Tamoxifen downregulates TGF-beta production in keloid fibroblasts.
        • Kuhn M.A.
        • Wang X.
        • Payne W.G.
        • Ko F.
        • Robson M.C.
        Tamoxifen decreases fibroblast function and downregulates TGF(beta2) in Dupuytren’s affected palmar fascia.
        • Degreef I.
        • Tejpar S.
        • Sciot R.
        • De Smet L.
        High-dosage tamoxifen as neoadjuvant treatment in minimally invasive surgery for Dupuytren disease in patients with a strong predisposition toward fibrosis: a randomized controlled trial.
        • Lorizio W.
        • Wu A.H.
        • Beattie M.S.
        • et al.
        Clinical and biomarker predictors of side effects from tamoxifen.
      Metformin
      • Phosphorylate AMP-activated protein kinase, regulating intracellular energy balance
        • Martin-Montalvo A.
        • Mercken E.M.
        • Mitchell S.J.
        • et al.
        Metformin improves healthspan and lifespan in mice.
      • Reduces TGF-β-induced ECM production in fibroblasts
        • Park I.H.
        • Um J.Y.
        • Hong S.M.
        • et al.
        Metformin reduces TGF-beta1-induced extracellular matrix production in nasal polyp-derived fibroblasts.
      • Decreased TGF-β-induced contraction of DD fibroblasts
        • Baeri A.
        • Levraut M.
        • Diazzi S.
        • et al.
        A role for metformin in the treatment of Dupuytren disease?.
      • Not tested
      ACE, angiotensin-converting enzyme; AMP, adenosine monophosphate; COX, carboxylase; DD, Dupuytren disease; ECM, extracellular matrix; GMP, guanosine monophosphate; LPA, lysophosphatidic acid; PDE5, phosphodiesterase 5; PNF, percutaneous needle fasciotomy; Ref., reference; SMA, smooth muscle actin; TGF, transforming growth factor.
      Multiple classes of anti-inflammatory and antimitotic medications, in addition to corticosteroids, have been proposed for DD. The nonsteroidal anti-inflammatory (NSAID) celecoxib is being investigated for a role in patients with a high risk of recurrence, while naproxen may have a benefit in reducing postoperative swelling following fasciectomy in patients with DD, although data thus far have been limited and not shown to effect a significant clinical difference.
      • Husby T.
      • Haugstvedt J.R.
      • Fyllingen G.
      • Skoglund L.A.
      Acute postoperative swelling after hand surgery: an exploratory, double-blind, randomised study with paracetamol, naproxen, and placebo.
      Interferons, both γ and α2b, have the ability to decrease mechanisms behind DD contracture in vitro.
      • Pittet B.
      • Rubbia-Brandt L.
      • Desmoulière A.
      • et al.
      Effect of gamma-interferon on the clinical and biologic evolution of hypertrophic scars and Dupuytren’s disease: an open pilot study.
      ,
      • Sanders J.L.
      • Dodd C.
      • Ghahary A.
      • Scott P.G.
      • Tredget E.E.
      The effect of interferon-alpha2b on an in vitro model Dupuytren’s contracture.
      A small pilot study demonstrated the potential to decrease the size of early DD nodules when injected intralesionally.
      • Pittet B.
      • Rubbia-Brandt L.
      • Desmoulière A.
      • et al.
      Effect of gamma-interferon on the clinical and biologic evolution of hypertrophic scars and Dupuytren’s disease: an open pilot study.
      However, no further studies appear in the literature to expand this work. 5-Fluorouracil (5-FU) demonstrated inhibitory effects on Dupuytren myofibroblasts in vitro,
      • Jemec B.
      • Linge C.
      • Grobbelaar A.O.
      • Smith P.J.
      • Sanders R.
      • McGrouther D.A.
      The effect of 5-fluorouracil on Dupuytren fibroblast proliferation and differentiation.
      but showed no beneficial clinical effect when used topically.
      • Bulstrode N.W.
      • Bisson M.
      • Jemec B.
      • Pratt A.L.
      • McGrouther D.A.
      • Grobbelaar A.O.
      A prospective randomised clinical trial of the intra-operative use of 5-fluorouracil on the outcome of Dupuytren’s disease.
      Systemic colchicine has been reported to improve the severity of penile contractures in Peyronie disease, although without improving concomitant DD contractures.
      • Akkus E.
      • Carrier S.
      • Rehman J.
      • Breza J.
      • Kadioglu A.
      • Lue T.F.
      Is colchicine effective in Peyronie’s disease? A pilot study.
      The anti-oxidants vitamin E and N-acetyl-L-cysteine (NAC) have been investigated for potential roles in DD owing to their ability to abrogate fibrogenesis in vitro.
      • Meurer S.K.
      • Lahme B.
      • Tihaa L.
      • Weiskirchen R.
      • Gressner A.M.
      N-acetyl-L-cysteine suppresses TGF-beta signaling at distinct molecular steps: the biochemical and biological efficacy of a multifunctional, antifibrotic drug.
      • Simon A.R.
      • Rai U.
      • Fanburg B.L.
      • Cochran B.H.
      Activation of the JAK-STAT pathway by reactive oxygen species.
      Vitamin E supplementation was initially described in the 1940s as a potential substitute for surgical therapy in DD.
      • Thomson G.R.
      Treatment of Dupuytren’s contracture with vitamin E.
      Its utility was refuted by subsequent studies as patients continued to progress despite supplementation.
      • Richards H.J.
      Dupuytren’s contracture treated with vitamin E.
      ,
      • Steinberg C.L.
      Tocopherols in treatment of primary fibrositis; including Dupuytren’s contracture, periarthritis of the shoulders, and Peyronie’s disease.
      NAC has been shown to play a potential role in fibroblast maturation in vitro, although it has not been explored as a therapeutic in patients.
      • Kopp J.
      • Seyhan H.
      • Müller B.
      • et al.
      N-acetyl-L-cysteine abrogates fibrogenic properties of fibroblasts isolated from Dupuytren’s disease by blunting TGF-beta signalling.
      Various antihypertensive and vasoactive medications have been described for DD. Angiotensin-converting enzyme (ACE) inhibitors and angiotensin II antagonists were proposed owing to their effect in decreasing fibrotic responses in vitro and in animal models.
      • Yu L.
      • Border W.A.
      • Anderson I.
      • McCourt M.
      • Huang Y.
      • Noble N.A.
      Combining TGF-beta inhibition and angiotensin II blockade results in enhanced antifibrotic effect.
      ,
      • Zimman O.A.
      • Toblli J.
      • Stella I.
      • Ferder M.
      • Ferder L.
      • Inserra F.
      The effects of angiotensin-converting-enzyme inhibitors on the fibrous envelope around mammary implants.
      More recently, Dupuytren tissue has been shown to express angiotensin II receptors.
      • Stephen C.
      • Touil L.
      • Vaiude P.
      • Singh J.
      • McKirdy S.
      Angiotensin receptors in Dupuytren’s disease: a target for pharmacological treatment?.
      Calcium-channel blockers (eg, verapamil) have been described owing to potential effects on myofibroblast-mediated contracture and the potential to decrease scarring in patients with burns.
      • Lee R.C.
      • Doong H.
      • Jellema A.F.
      The response of burn scars to intralesional verapamil. Report of five cases.
      ,
      • Rayan G.M.
      • Parizi M.
      • Tomasek J.J.
      Pharmacologic regulation of Dupuytren’s fibroblast contraction in vitro.
      Phosphodiesterase inhibitors (eg, sildenafil) have been proposed as they improve fibrosis via plaque development in animal models with Peyronie disease, another localized fibrotic process.
      • Gonzalez-Cadavid N.F.
      • Rajfer J.
      Treatment of Peyronie’s disease with PDE5 inhibitors: an antifibrotic strategy.
      • Dunkern T.R.
      • Feurstein D.
      • Rossi G.A.
      • Sabatini F.
      • Hatzelmann A.
      Inhibition of TGF-beta induced lung fibroblast to myofibroblast conversion by phosphodiesterase inhibiting drugs and activators of soluble guanylyl cyclase.
      • Ferrini M.G.
      • Kovanecz I.
      • Nolazco G.
      • Rajfer J.
      • Gonzalez-Cadavid N.F.
      Effects of long-term vardenafil treatment on the development of fibrotic plaques in a rat model of Peyronie’s disease.
      Nitric oxide donors (eg, molsidomine) decrease lung fibrosis and Peyronie disease progression in animal models, likely due to the inhibitory effect nitric oxide has on myofibroblast differentiation and function.
      • Gonzalez-Cadavid N.F.
      • Rajfer J.
      Treatment of Peyronie’s disease with PDE5 inhibitors: an antifibrotic strategy.
      ,
      • Ferrini M.G.
      • Vernet D.
      • Magee T.R.
      • et al.
      Antifibrotic role of inducible nitric oxide synthase.
      • Vernet D.
      • Ferrini M.G.
      • Valente E.G.
      • et al.
      Effect of nitric oxide on the differentiation of fibroblasts into myofibroblasts in the Peyronie’s fibrotic plaque and in its rat model.
      • Kilic T.
      • Parlakpinar H.
      • Polat A.
      • et al.
      Protective and therapeutic effect of molsidomine on bleomycin-induced lung fibrosis in rats.
      Neither phosphodiesterase inhibitors nor nitric oxide donors have been tested in models of DD.
      The synthetic nonsteroidal antiestrogen, tamoxifen, modulates TGF-β production, signaling, and fibroblast contractility in vitro.
      • Chau D.
      • Mancoll J.S.
      • Lee S.
      • et al.
      Tamoxifen downregulates TGF-beta production in keloid fibroblasts.
      • Kuhn M.A.
      • Wang X.
      • Payne W.G.
      • Ko F.
      • Robson M.C.
      Tamoxifen decreases fibroblast function and downregulates TGF(beta2) in Dupuytren’s affected palmar fascia.
      • Lorizio W.
      • Wu A.H.
      • Beattie M.S.
      • et al.
      Clinical and biomarker predictors of side effects from tamoxifen.
      It also results in short-term improvements in patients with DD undergoing limited fasciectomy. However, the gain is lost by two years after treatment and the side effect profile was poorly tolerated.
      • Degreef I.
      • Tejpar S.
      • Sciot R.
      • De Smet L.
      High-dosage tamoxifen as neoadjuvant treatment in minimally invasive surgery for Dupuytren disease in patients with a strong predisposition toward fibrosis: a randomized controlled trial.
      Recently, metformin was proposed as a potential treatment for DD because of its ability to prevent TGF-β–mediated induction of fibroblasts in vitro.
      • Martin-Montalvo A.
      • Mercken E.M.
      • Mitchell S.J.
      • et al.
      Metformin improves healthspan and lifespan in mice.
      ,
      • Park I.H.
      • Um J.Y.
      • Hong S.M.
      • et al.
      Metformin reduces TGF-beta1-induced extracellular matrix production in nasal polyp-derived fibroblasts.
      While these results were shown using fibroblasts isolated from samples of patients with DD, no clinical trials demonstrating an effect of metformin in DD have been performed.
      • Baeri A.
      • Levraut M.
      • Diazzi S.
      • et al.
      A role for metformin in the treatment of Dupuytren disease?.

      Currently Approved Therapies Targeting Inflammation And Fibrosis

      Several pharmacotherapies with current approval for their anti-inflammatory or antifibrotic effects are under investigation for use in DD (Table 2). The rationale for their use and results to date are discussed below.
      Table 2Currently Approved Pharmacotherapies in Fibrosis and Inflammation
      MedicationBasic Mechanism of DrugProposed Mechanism in DDIn Vitro Results in DD modelIn Vivo Results in Patients with DDRef.
      TNF inhibitors (eg, adalimumab)
      • Monoclonal antibody targeting and inactivating TNF-α
      • Decrease TNF-driven fibroblast differentiation and contraction
        • Verjee L.S.
        • Verhoekx J.S.
        • Chan J.K.
        • et al.
        Unraveling the signaling pathways promoting fibrosis in Dupuytren’s disease reveals TNF as a therapeutic target.
      • Inhibition of myofibroblast contraction
      • Reduced α-SMA expression
        • Verjee L.S.
        • Verhoekx J.S.
        • Chan J.K.
        • et al.
        Unraveling the signaling pathways promoting fibrosis in Dupuytren’s disease reveals TNF as a therapeutic target.
      • Down regulation of myofibroblast phenotype in DD nodules
        • Nanchahal J.
        • Ball C.
        • Davidson D.
        • et al.
        Anti-tumour necrosis factor therapy for Dupuytren’s disease: A randomised dose response proof of concept phase 2a clinical trial.
        ,
        • Nanchahal J.
        • Ball C.
        • Swettenham J.
        • et al.
        Study protocol: A multi-centre, double blind, randomised, placebo-controlled, parallel group, phase II trial (RIDD) to determine the efficacy of intra-nodular injection of anti-TNF to control disease progression in early Dupuytren’s disease, with an embedded dose response study.
      • DD nodule softening and size reduction at 1 year
        • Nanchahal J.
        • Ball C.
        • Rombach I.
        • et al.
        Anti-tumour necrosis factor therapy for early-stage Dupuytren’s disease (RIDD): a phase 2b, randomised, double-blind, placebo-controlled trial.
        • Verjee L.S.
        • Verhoekx J.S.
        • Chan J.K.
        • et al.
        Unraveling the signaling pathways promoting fibrosis in Dupuytren’s disease reveals TNF as a therapeutic target.
        ,
        • Nanchahal J.
        • Ball C.
        • Davidson D.
        • et al.
        Anti-tumour necrosis factor therapy for Dupuytren’s disease: A randomised dose response proof of concept phase 2a clinical trial.
        • Nanchahal J.
        • Ball C.
        • Swettenham J.
        • et al.
        Study protocol: A multi-centre, double blind, randomised, placebo-controlled, parallel group, phase II trial (RIDD) to determine the efficacy of intra-nodular injection of anti-TNF to control disease progression in early Dupuytren’s disease, with an embedded dose response study.
        • Nanchahal J.
        • Ball C.
        • Rombach I.
        • et al.
        Anti-tumour necrosis factor therapy for early-stage Dupuytren’s disease (RIDD): a phase 2b, randomised, double-blind, placebo-controlled trial.
      Nintedanib
      • Tyrosine kinase inhibitor targeting profibrogenesis signaling (VEGF, FGF, PDGF)
        • Richeldi L.
        • Costabel U.
        • Selman M.
        • et al.
        Efficacy of a tyrosine kinase inhibitor in idiopathic pulmonary fibrosis.
      • Decrease fibroblast differentiation by PDGF- and FGF-signaling
        • Krause C.
        • Kloen P.
        • Ten Dijke P.
        Elevated transforming growth factor beta and mitogen-activated protein kinase pathways mediate fibrotic traits of Dupuytren’s disease fibroblasts.
        ,
        • Lappi D.A.
        • Martineau D.
        • Maher P.A.
        • et al.
        Basic fibroblast growth factor in cells derived from Dupuytren’s contracture: synthesis, presence, and implications for treatment of the disease.
      • Not tested
      • Not tested
        • Krause C.
        • Kloen P.
        • Ten Dijke P.
        Elevated transforming growth factor beta and mitogen-activated protein kinase pathways mediate fibrotic traits of Dupuytren’s disease fibroblasts.
        ,
        • Richeldi L.
        • Costabel U.
        • Selman M.
        • et al.
        Efficacy of a tyrosine kinase inhibitor in idiopathic pulmonary fibrosis.
        ,
        • Lappi D.A.
        • Martineau D.
        • Maher P.A.
        • et al.
        Basic fibroblast growth factor in cells derived from Dupuytren’s contracture: synthesis, presence, and implications for treatment of the disease.
      Pirfenidone
      • Inhibits TGF-β production and TGF-β-mediated fibroblast function
        • Glassberg M.K.
        Overview of idiopathic pulmonary fibrosis, evidence-based guidelines, and recent developments in the treatment landscape.
        ,
        • Noble P.W.
        • Albera C.
        • Bradford W.Z.
        • et al.
        Pirfenidone in patients with idiopathic pulmonary fibrosis (CAPACITY): two randomised trials.
      • Decrease TGF-β-mediated fibroblast function and differentiation
        • Glassberg M.K.
        Overview of idiopathic pulmonary fibrosis, evidence-based guidelines, and recent developments in the treatment landscape.
        ,
        • Noble P.W.
        • Albera C.
        • Bradford W.Z.
        • et al.
        Pirfenidone in patients with idiopathic pulmonary fibrosis (CAPACITY): two randomised trials.
      • Decreased fibroblast proliferation, myofibroblast differentiation, and matrix production
        • Zhou C.
        • Liu F.
        • Gallo P.H.
        • Baratz M.E.
        • Kathju S.
        • Satish L.
        Anti-fibrotic action of pirfenidone in Dupuytren’s disease-derived fibroblasts.
        ,
        • Zhou C.
        • Zeldin Y.
        • Baratz M.E.
        • Kathju S.
        • Satish L.
        Investigating the effects of pirfenidone on TGF-beta1 stimulated non-SMAD signaling pathways in Dupuytren’s disease -derived fibroblasts.
      • Not tested
      • Intradermal formulation being investigated for DD
        • Panigrahi S.
        • Barry A.
        • Multner S.
        • et al.
        Pirfenidone as a potential antifibrotic injectable for Dupuytren’s disease.
        • Glassberg M.K.
        Overview of idiopathic pulmonary fibrosis, evidence-based guidelines, and recent developments in the treatment landscape.
        • Noble P.W.
        • Albera C.
        • Bradford W.Z.
        • et al.
        Pirfenidone in patients with idiopathic pulmonary fibrosis (CAPACITY): two randomised trials.
        • Zhou C.
        • Liu F.
        • Gallo P.H.
        • Baratz M.E.
        • Kathju S.
        • Satish L.
        Anti-fibrotic action of pirfenidone in Dupuytren’s disease-derived fibroblasts.
        • Zhou C.
        • Zeldin Y.
        • Baratz M.E.
        • Kathju S.
        • Satish L.
        Investigating the effects of pirfenidone on TGF-beta1 stimulated non-SMAD signaling pathways in Dupuytren’s disease -derived fibroblasts.
        • Panigrahi S.
        • Barry A.
        • Multner S.
        • et al.
        Pirfenidone as a potential antifibrotic injectable for Dupuytren’s disease.
      Tocilizumab
      • Monoclonal antibody targeting IL-6 receptor, preventing binding of IL-6
      • Decrease IL-6–mediated myofibroblast development
        • Cardoneanu A.
        • Burlui A.M.
        • Macovei L.A.
        • Bratoiu I.
        • Richter P.
        • Rezus E.
        Targeting systemic sclerosis from pathogenic mechanisms to clinical manifestations: why IL-6?.
      • No effects on DD cells
        • Verjee L.S.
        • Verhoekx J.S.
        • Chan J.K.
        • et al.
        Unraveling the signaling pathways promoting fibrosis in Dupuytren’s disease reveals TNF as a therapeutic target.
      • Not tested
        • Verjee L.S.
        • Verhoekx J.S.
        • Chan J.K.
        • et al.
        Unraveling the signaling pathways promoting fibrosis in Dupuytren’s disease reveals TNF as a therapeutic target.
        ,
        • Cardoneanu A.
        • Burlui A.M.
        • Macovei L.A.
        • Bratoiu I.
        • Richter P.
        • Rezus E.
        Targeting systemic sclerosis from pathogenic mechanisms to clinical manifestations: why IL-6?.
      Rituximab
      • Monoclonal antibody targeting B-cell surface protein CD20
        • Smith M.R.
        Rituximab (monoclonal anti-CD20 antibody): mechanisms of action and resistance.
      • Decrease autoantibody contribution to DD
        • Brenner P.
        • Sachse C.
        • Reichert B.
        • Berger A.
        [Expression of various monoclonal antibodies in nodules and band stage in Dupuytren’s disease].
        • Menzel E.J.
        • Piza H.
        • Zielinski C.
        • Endler A.T.
        • Steffen C.
        • Millesi H.
        Collagen types and anticollagen-antibodies in Dupuytren’s disease.
        • Pereira R.S.
        • Black C.M.
        • Turner S.M.
        • Spencer J.D.
        Antibodies to collagen types I–VI in Dupuytren’s contracture.
      • Not tested
      • Not tested
        • Smith M.R.
        Rituximab (monoclonal anti-CD20 antibody): mechanisms of action and resistance.
        • Brenner P.
        • Sachse C.
        • Reichert B.
        • Berger A.
        [Expression of various monoclonal antibodies in nodules and band stage in Dupuytren’s disease].
        • Menzel E.J.
        • Piza H.
        • Zielinski C.
        • Endler A.T.
        • Steffen C.
        • Millesi H.
        Collagen types and anticollagen-antibodies in Dupuytren’s disease.
        • Pereira R.S.
        • Black C.M.
        • Turner S.M.
        • Spencer J.D.
        Antibodies to collagen types I–VI in Dupuytren’s contracture.
      CD20, B-lymphocyte antigen CD20; DD, Dupuytren disease; FGF, fibroblast growth factor; IL, interleukin; PDGF, platelet-derived growth factor; Ref., reference; SMA, smooth muscle actin; TGF, transforming growth factor; TNF, tumor necrosis factor; VEGF, vascular endothelial growth factor.

      Tumor necrosis factor inhibition

      Tumor necrosis factor (TNF) is known to play a role in the development and maintenance of the myofibroblast phenotype in DD nodules (Table 2). This has been demonstrated in vitro where the addition of TNF, but not other known proinflammatory cytokines (interleukin [IL]-6 and IL-1β), to fibroblasts from samples of patients with DD promoted their differentiation into myofibroblasts.
      • Verjee L.S.
      • Verhoekx J.S.
      • Chan J.K.
      • et al.
      Unraveling the signaling pathways promoting fibrosis in Dupuytren’s disease reveals TNF as a therapeutic target.
      TNF blockade has been performed on DD cells in vitro using the FDA-approved anti-TNF agents, adalimumab and golimumab. Both agents effectively inhibit myofibroblast contraction.
      • Verjee L.S.
      • Verhoekx J.S.
      • Chan J.K.
      • et al.
      Unraveling the signaling pathways promoting fibrosis in Dupuytren’s disease reveals TNF as a therapeutic target.
      These studies have since been corroborated in a proof-of-concept clinical trial. TNF blockade was performed by injection of adalimumab into DD nodules, followed by surgical excision and evaluation. Nodules demonstrated down regulation of the myofibroblast phenotype.
      • Nanchahal J.
      • Ball C.
      • Davidson D.
      • et al.
      Anti-tumour necrosis factor therapy for Dupuytren’s disease: A randomised dose response proof of concept phase 2a clinical trial.
      ,
      • Nanchahal J.
      • Ball C.
      • Swettenham J.
      • et al.
      Study protocol: A multi-centre, double blind, randomised, placebo-controlled, parallel group, phase II trial (RIDD) to determine the efficacy of intra-nodular injection of anti-TNF to control disease progression in early Dupuytren’s disease, with an embedded dose response study.
      Most recently, adalimumab injection in early-stage DD resulted in nodule softening and size reduction at one year.
      • Nanchahal J.
      • Ball C.
      • Rombach I.
      • et al.
      Anti-tumour necrosis factor therapy for early-stage Dupuytren’s disease (RIDD): a phase 2b, randomised, double-blind, placebo-controlled trial.
      Further studies are necessary to assess the long-term utility of TNF blockade in the treatment of DD.

      Nintedanib

      Nintedanib is one of the two currently used treatments for idiopathic pulmonary fibrosis. Approved for use in the United States in 2014, and in Europe in 2015, Nintedanib is a tyrosine kinase inhibitor with known effects on signaling receptors involved in fibrogenesis, chiefly vascular endothelial growth factor, fibroblast growth factor (FGF), and PDGF.
      • Richeldi L.
      • Costabel U.
      • Selman M.
      • et al.
      Efficacy of a tyrosine kinase inhibitor in idiopathic pulmonary fibrosis.
      Involvement of PDGF and FGF-mediated signaling has been shown in DD fibroblasts in vitro.
      • Krause C.
      • Kloen P.
      • Ten Dijke P.
      Elevated transforming growth factor beta and mitogen-activated protein kinase pathways mediate fibrotic traits of Dupuytren’s disease fibroblasts.
      ,
      • Lappi D.A.
      • Martineau D.
      • Maher P.A.
      • et al.
      Basic fibroblast growth factor in cells derived from Dupuytren’s contracture: synthesis, presence, and implications for treatment of the disease.
      Stimulation of PDGF and FGF signaling pathways can be downstream of TGF-β signaling,
      • Krause C.
      • Kloen P.
      Concurrent inhibition of TGF-beta and mitogen driven signaling cascades in Dupuytren’s disease - non-surgical treatment strategies from a signaling point of view.
      which as discussed earlier, plays a role in DD development. Yet, no studies examining the use of Nintedanib for DD have been performed to date. It could be a potential target in DD. However, since Nintedanib does not directly affect TGF-β-mediated fibrogenesis, it may ultimately have limited clinical utility.

      Pirfenidone

      Pirfenidone (PFD; 5-methyl-1-phenyl-2(1H)-pyridone) is the second most used treatment for idiopathic pulmonary fibrosis. Approved for use in Europe in 2011 and in the United States in 2014, PFD has an inhibitory effect on TGF-β production and TGF-β-mediated fibroblast function and differentiation.
      • Glassberg M.K.
      Overview of idiopathic pulmonary fibrosis, evidence-based guidelines, and recent developments in the treatment landscape.
      ,
      • Noble P.W.
      • Albera C.
      • Bradford W.Z.
      • et al.
      Pirfenidone in patients with idiopathic pulmonary fibrosis (CAPACITY): two randomised trials.
      PFD has been tested in DD fibroblasts in vitro and shown to abrogate TGF-β effects including fibroblast proliferation, myofibroblast development, and matrix formation.
      • Zhou C.
      • Liu F.
      • Gallo P.H.
      • Baratz M.E.
      • Kathju S.
      • Satish L.
      Anti-fibrotic action of pirfenidone in Dupuytren’s disease-derived fibroblasts.
      ,
      • Zhou C.
      • Zeldin Y.
      • Baratz M.E.
      • Kathju S.
      • Satish L.
      Investigating the effects of pirfenidone on TGF-beta1 stimulated non-SMAD signaling pathways in Dupuytren’s disease -derived fibroblasts.
      A PFD formulation that could be delivered locally in DD is currently in development.
      • Panigrahi S.
      • Barry A.
      • Multner S.
      • et al.
      Pirfenidone as a potential antifibrotic injectable for Dupuytren’s disease.

      Tocilizumab and rituximab

      It is worth mentioning two additional therapies currently used in cancer and inflammation, and with proposed effects in fibrosis–tocilizumab and rituximab. Tocilizumab is a monoclonal antibody targeting the IL-6 receptor and preventing the binding of IL-6. Although not currently approved for the treatment of fibrosis, it has been suggested based on the known effect of IL-6 on myofibroblast development.
      • Cardoneanu A.
      • Burlui A.M.
      • Macovei L.A.
      • Bratoiu I.
      • Richter P.
      • Rezus E.
      Targeting systemic sclerosis from pathogenic mechanisms to clinical manifestations: why IL-6?.
      Despite the upregulation of IL-6 in cells from DD tissue, neither the addition of IL-6 nor its blockade has shown significant effects on DD cells in vitro.
      • Verjee L.S.
      • Verhoekx J.S.
      • Chan J.K.
      • et al.
      Unraveling the signaling pathways promoting fibrosis in Dupuytren’s disease reveals TNF as a therapeutic target.
      Rituximab is a monoclonal antibody targeting the B-cell surface protein CD20, leading to downregulation of B-cell differentiation and antibody formation.
      • Smith M.R.
      Rituximab (monoclonal anti-CD20 antibody): mechanisms of action and resistance.
      Its primary clinical applications include B-cell lymphomas, leukemias, and B-cell mediated autoimmune diseases.
      While some have suggested a role for autoantibodies in DD,
      • Brenner P.
      • Sachse C.
      • Reichert B.
      • Berger A.
      [Expression of various monoclonal antibodies in nodules and band stage in Dupuytren’s disease].
      • Menzel E.J.
      • Piza H.
      • Zielinski C.
      • Endler A.T.
      • Steffen C.
      • Millesi H.
      Collagen types and anticollagen-antibodies in Dupuytren’s disease.
      • Pereira R.S.
      • Black C.M.
      • Turner S.M.
      • Spencer J.D.
      Antibodies to collagen types I–VI in Dupuytren’s contracture.
      a definitive link with CD20-positive cells has not been made.

      Conclusion

      Dupuytren disease remains a challenging clinical entity to treat. While historically accepted and proven therapies such as fasciectomy procedures demonstrate good efficacy of treatment, their risk profile has led to a search for minimally invasive techniques. Enzymatic digestion and treatment with collagenase have become a staple in DD treatment for over a decade. Despite the addition and broad acceptance of this pharmacotherapy, there is a need of a primary (or adjuvant) therapy modality that can either stop progression in the 30% to 50% of patients in early stages at risk or prevent disease recurrence following treatment.
      As fibroproliferation underscores the etiology of DD, it is important to take an antifibrogenic approach to find new pharmacotherapies. This review highlights the pathophysiologic basis of the fibrotic response seen in DD, chiefly through TGF-β signaling. The 2 most used pharmacotherapies in DD today, collagenase for enzymatic digestion of diseased cords as well as corticosteroids for the anti-inflammatory effects, are described. For the sake of providing historical background, many of the medications that have been discussed in articles over years for a repurposed use in DD are covered. Lastly, the article presents several medications with current approval for anti-inflammatory or antifibrotic effects that either are being used or may be considered for use in DD.

      Acknowledgments

      All authors contributed to the preparation of the manuscript, including design and figure preparation. This work was supported by the National Institute of Arthritis and Musculoskeletal and Skin Diseases of the National Institutes of Health under Grant 5R01AR056019-13 to senior author (MFB). Figure 1 was created with BioRender.com.

      Supplementary Data

      References

        • Karatzas E.
        • Kakouri A.C.
        • Kolios G.
        • Delis A.
        • Spyrou G.M.
        Fibrotic expression profile analysis reveals repurposed drugs with potential anti-fibrotic mode of action.
        PLOS ONE. 2021; 16e0249687
        • Rosenbloom J.
        • Mendoza F.A.
        • Jimenez S.A.
        Strategies for anti-fibrotic therapies.
        Biochim Biophys Acta. 2013; 1832: 1088-1103
        • Tai Y.
        • Woods E.L.
        • Dally J.
        • et al.
        Myofibroblasts: function, formation, and scope of molecular therapies for skin fibrosis.
        Biomolecules. 2021; 11: 1095
        • Mella J.R.
        • Guo L.
        • Hung V.
        Dupuytren’s contracture: an evidence based review.
        Ann Plast Surg. 2018; 81: S97-S101
        • Raymond A.
        • Parry M.
        • Amirfeyz R.
        Critical angles of deformity in Dupuytren’s contracture of the little and ring fingers.
        Hand Surg. 2015; 20: 290-297
        • Denkler K.A.
        • Vaughn C.J.
        • Dolan E.L.
        • Hansen S.L.
        Evidence-based medicine: options for Dupuytren’s contracture: incise, excise, and dissolve.
        Plast Reconstr Surg. 2017; 139: 240e-255e
        • Shih B.
        • Bayat A.
        Scientific understanding and clinical management of Dupuytren disease.
        Nat Rev Rheumatol. 2010; 6: 715-726
        • Luck J.V.
        Dupuytren’s contracture; a new concept of the pathogenesis correlated with surgical management.
        J Bone Joint Surg Am. 1959; 41-A: 635-664
        • Zhang A.Y.
        • Kargel J.S.
        The basic science of Dupuytren disease.
        Hand Clin. 2018; 34: 301-305
        • Godtfredsen N.S.
        • Lucht H.
        • Prescott E.
        • Sørensen T.I.
        • Grønbaek M.
        A prospective study linked both alcohol and tobacco to Dupuytren’s disease.
        J Clin Epidemiol. 2004; 57: 858-863
        • Burke F.D.
        • Proud G.
        • Lawson I.J.
        • McGeoch K.L.
        • Miles J.N.
        An assessment of the effects of exposure to vibration, smoking, alcohol and diabetes on the prevalence of Dupuytren’s disease in 97,537 miners.
        J Hand Surg Eur Vol. 2007; 32: 400-406
        • Burge P.
        • Hoy G.
        • Regan P.
        • Milne R.
        Smoking, alcohol and the risk of Dupuytren’s contracture.
        J Bone Joint Surg Br. 1997; 79: 206-210
        • Liss G.M.
        • Stock S.R.
        Can Dupuytren’s contracture be work-related?: review of the evidence.
        Am J Ind Med. 1996; 29: 521-532
        • Gudmundsson K.G.
        • Arngrímsson R.
        • Sigfússon N.
        • Björnsson A.
        • Jónsson T.
        Epidemiology of Dupuytren’s disease: clinical, serological, and social assessment. The Reykjavik Study.
        J Clin Epidemiol. 2000; 53: 291-296
        • Noble J.
        • Heathcote J.G.
        • Cohen H.
        Diabetes mellitus in the aetiology of Dupuytren’s disease.
        J Bone Joint Surg Br. 1984; 66: 322-325
        • Dibenedetti D.B.
        • Nguyen D.
        • Zografos L.
        • Ziemiecki R.
        • Zhou X.
        Prevalence, incidence, and treatments of Dupuytren’s disease in the United States: results from a population-based study.
        Hand (N Y). 2011; 6: 149-158
        • Rydberg M.
        • Zimmerman M.
        • Löfgren J.P.
        • et al.
        Metabolic factors and the risk of Dupuytren’s disease: data from 30,000 individuals followed for over 20 years.
        Sci Rep. 2021; 1114669
        • Rabinowitz J.L.
        • Ostermann Jr., L.
        • Bora F.W.
        • Staeffen J.
        Lipid composition and de novo lipid biosynthesis of human palmar fat in Dupuytren’s disease.
        Lipids. 1983; 18: 371-374
        • Bergenudd H.
        • Lindgärde F.
        • Nilsson B.E.
        Prevalence of Dupuytren’s contracture and its correlation with degenerative changes of the hands and feet and with criteria of general health.
        J Hand Surg Br. 1993; 18: 254-257
        • Shih B.
        • Brown J.J.
        • Armstrong D.J.
        • Lindau T.
        • Bayat A.
        Differential gene expression analysis of subcutaneous fat, fascia, and skin overlying a Dupuytren’s disease nodule in comparison to control tissue.
        Hand (N Y). 2009; 4: 294-301
        • Hinz B.
        Formation and function of the myofibroblast during tissue repair.
        J Invest Dermatol. 2007; 127: 526-537
        • Tomasek J.J.
        • Schultz R.J.
        • Haaksma C.J.
        Extracellular matrix-cytoskeletal connections at the surface of the specialized contractile fibroblast (myofibroblast) in Dupuytren disease.
        J Bone Joint Surg Am. 1987; 69: 1400-1407
        • Tomasek J.J.
        • Gabbiani G.
        • Hinz B.
        • Chaponnier C.
        • Brown R.A.
        Myofibroblasts and mechano-regulation of connective tissue remodelling.
        Nat Rev Mol Cell Biol. 2002; 3: 349-363
        • Singer II,
        • Kawka D.W.
        • Kazazis D.M.
        • Clark R.A.
        In vivo co-distribution of fibronectin and actin fibers in granulation tissue: immunofluorescence and electron microscope studies of the fibronexus at the myofibroblast surface.
        J Cell Biol. 1984; 98: 2091-2106
        • Burridge K.
        • Chrzanowska-Wodnicka M.
        Focal adhesions, contractility, and signaling.
        Annu Rev Cell Dev Biol. 1996; 12: 463-518
        • Dugina V.
        • Fontao L.
        • Chaponnier C.
        • Vasiliev J.
        • Gabbiani G.
        Focal adhesion features during myofibroblastic differentiation are controlled by intracellular and extracellular factors.
        J Cell Sci. 2001; 114: 3285-3296
        • Krause C.
        • Kloen P.
        Concurrent inhibition of TGF-beta and mitogen driven signaling cascades in Dupuytren’s disease - non-surgical treatment strategies from a signaling point of view.
        Med Hypotheses. 2012; 78: 385-388
        • Berndt A.
        • Kosmehl H.
        • Mandel U.
        • et al.
        TGF beta and bFGF synthesis and localization in Dupuytren’s disease (nodular palmar fibromatosis) relative to cellular activity, myofibroblast phenotype and oncofetal variants of fibronectin.
        Histochem J. 1995; 27: 1014-1020
        • Baird K.S.
        • Crossan J.F.
        • Ralston S.H.
        Abnormal growth factor and cytokine expression in Dupuytren’s contracture.
        J Clin Pathol. 1993; 46: 425-428
        • Badalamente M.A.
        • Sampson S.P.
        • Hurst L.C.
        • Dowd A.
        • Miyasaka K.
        The role of transforming growth factor beta in Dupuytren’s disease.
        J Hand Surg Am. 1996; 21: 210-215
        • Bisson M.A.
        • McGrouther D.A.
        • Mudera V.
        • Grobbelaar A.O.
        The different characteristics of Dupuytren’s disease fibroblasts derived from either nodule or cord: expression of alpha-smooth muscle actin and the response to stimulation by TGF-beta1.
        J Hand Surg Br. 2003; 28: 351-356
        • Verjee L.S.
        • Verhoekx J.S.
        • Chan J.K.
        • et al.
        Unraveling the signaling pathways promoting fibrosis in Dupuytren’s disease reveals TNF as a therapeutic target.
        Proc Natl Acad Sci U S A. 2013; 110: E928-E937
        • Krause C.
        • Kloen P.
        • Ten Dijke P.
        Elevated transforming growth factor beta and mitogen-activated protein kinase pathways mediate fibrotic traits of Dupuytren’s disease fibroblasts.
        Fibrogenesis Tissue Repair. 2011; 4: 14
        • Tse R.
        • Howard J.
        • Wu Y.
        • Gan B.S.
        Enhanced Dupuytren’s disease fibroblast populated collagen lattice contraction is independent of endogenous active TGF-beta2.
        BMC Musculoskelet Disord. 2004; 5: 41
        • Satish L.
        • LaFramboise W.A.
        • Johnson S.
        • et al.
        Fibroblasts from phenotypically normal palmar fascia exhibit molecular profiles highly similar to fibroblasts from active disease in Dupuytren’s contracture.
        BMC Med Genomics. 2012; 5: 15
        • Rehman S.
        • Xu Y.
        • Dunn W.B.
        • et al.
        Dupuytren’s disease metabolite analyses reveals alterations following initial short-term fibroblast culturing.
        Mol Biosyst. 2012; 8: 2274-2288
        • Verjee L.S.
        • 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: 681-690
        • Gotway M.B.
        • Freemer M.M.
        • King Jr., T.E.
        Challenges in pulmonary fibrosis. 1: Use of high resolution CT scanning of the lung for the evaluation of patients with idiopathic interstitial pneumonias.
        Thorax. 2007; 62: 546-553
        • Martins-Bach A.B.
        • Bachasson D.
        • Araujo E.C.A.
        • et al.
        Non-invasive assessment of skeletal muscle fibrosis in mice using nuclear magnetic resonance imaging and ultrasound shear wave elastography.
        Sci Rep. 2021; 11: 284
        • Murphy A.
        • Lalonde D.H.
        • Eaton C.
        • et al.
        Minimally invasive options in Dupuytren’s contracture: aponeurotomy, enzymes, stretching, and fat grafting.
        Plast Reconstr Surg. 2014; 134: 822e-829e
        • Badalamente M.A.
        • Hurst L.C.
        Development of collagenase treatment for Dupuytren disease.
        Hand Clin. 2018; 34: 345-349
        • Freshwater M.F.
        What were the adverse events for Dupuytren’s patients treated with Xiaflex who had contractures less than 20°?.
        Hand. 2012; 7: 348-349
        • Scherman P.
        • Jenmalm P.
        • Dahlin L.B.
        Three-year recurrence of Dupuytren’s contracture after needle fasciotomy and collagenase injection: a two-centre randomized controlled trial.
        J Hand Surg Eur Vol. 2018; 43: 836-840
        • Skov S.T.
        • Bisgaard T.
        • Søndergaard P.
        • Lange J.
        Injectable collagenase versus percutaneous needle fasciotomy for Dupuytren contracture in proximal interphalangeal joints: A randomized controlled trial.
        J Hand Surg Am. 2017; 42: 321-328.e323
        • Ketchum L.D.
        The rationale for treating the nodule in Dupuytren’s disease.
        Plast Reconstr Surg Glob Open. 2014; 2: e278
        • Zachariae L.
        • Zachariae F.
        Hydrocortisone acetate in the treatment of Dupuytren’s contraction and allied conditions.
        Acta Chir Scand. 1955; 109: 421-431
        • Ketchum L.D.
        • Donahue T.K.
        The injection of nodules of Dupuytren’s disease with triamcinolone acetonide.
        J Hand Surg Am. 2000; 25: 1157-1162
        • Coste F.
        • Weissenbach R.
        [Treatment of Dupuytren’s disease by local injections of hydrocortisone].
        Rev Rhum Mal Osteoartic. 1953; 20: 863-866
        • Meek R.M.
        • McLellan S.
        • Reilly J.
        • Crossan J.F.
        The effect of steroids on Dupuytren’s disease: role of programmed cell death.
        J Hand Surg Br. 2002; 27: 270-273
        • Ketchum L.D.
        • Robinson D.W.
        • Masters F.W.
        The degradation of mature collagen: a laboratory study.
        Plast Reconstr Surg. 1967; 40: 89-91
        • McMillan C.
        • Binhammer P.
        Steroid injection and needle aponeurotomy for Dupuytren contracture: a randomized, controlled study.
        J Hand Surg Am. 2012; 37: 1307-1312
        • Berman B.
        • Maderal A.
        • Raphael B.
        Keloids and hypertrophic scars: pathophysiology, classification, and treatment.
        Dermatol Surg. 2017; 43: S3-S18
        • Davies N.M.
        • McLachlan A.J.
        • Day R.O.
        • Williams K.M.
        Clinical pharmacokinetics and pharmacodynamics of celecoxib: a selective cyclo-oxygenase-2 inhibitor.
        Clin Pharmacokinet. 2000; 38: 225-242
        • Salib C.G.
        • Reina N.
        • Trousdale W.H.
        • et al.
        Inhibition of COX-2 pathway as a potential prophylaxis against arthrofibrogenesis in a rabbit model of joint contracture.
        J Orthop Res. 2019; 37: 2609-2620
        • Chen H.
        • Qian Z.
        • Zhang S.
        • et al.
        Silencing COX-2 blocks PDK1/TRAF4-induced AKT activation to inhibit fibrogenesis during skeletal muscle atrophy.
        Redox Biol. 2021; 38101774
        • Baron S.
        • Tyring S.K.
        • Fleischmann W.R.
        • et al.
        The interferons. Mechanisms of action and clinical applications.
        JAMA. 1991; 266: 1375-1383
        • Pittet B.
        • Rubbia-Brandt L.
        • Desmoulière A.
        • et al.
        Effect of gamma-interferon on the clinical and biologic evolution of hypertrophic scars and Dupuytren’s disease: an open pilot study.
        Plast Reconstr Surg. 1994; 93: 1224-1235
        • Sanders J.L.
        • Dodd C.
        • Ghahary A.
        • Scott P.G.
        • Tredget E.E.
        The effect of interferon-alpha2b on an in vitro model Dupuytren’s contracture.
        J Hand Surg Am. 1999; 24: 578-585
        • Werker P.M.N.
        • Degreef I.
        Alternative and adjunctive treatments for Dupuytren disease.
        Hand Clin. 2018; 34: 367-375
        • Jemec B.
        • Linge C.
        • Grobbelaar A.O.
        • Smith P.J.
        • Sanders R.
        • McGrouther D.A.
        The effect of 5-fluorouracil on Dupuytren fibroblast proliferation and differentiation.
        Chir Main. 2000; 19: 15-22
        • Bulstrode N.W.
        • Bisson M.
        • Jemec B.
        • Pratt A.L.
        • McGrouther D.A.
        • Grobbelaar A.O.
        A prospective randomised clinical trial of the intra-operative use of 5-fluorouracil on the outcome of Dupuytren’s disease.
        J Hand Surg Br. 2004; 29: 18-21
        • Rojkind M.
        • Uribe M.
        • Kershenobich D.
        Colchicine and the treatment of liver cirrhosis.
        Lancet. 1973; 1: 38-39
        • Diegelmann R.F.
        • Peterkofsky B.
        Inhibition of collagen secretion from bone and cultured fibroblasts by microtubular disruptive drugs.
        Proc Natl Acad Sci U S A. 1972; 69: 892-896
        • Akkus E.
        • Carrier S.
        • Rehman J.
        • Breza J.
        • Kadioglu A.
        • Lue T.F.
        Is colchicine effective in Peyronie’s disease? A pilot study.
        Urology. 1994; 44: 291-295
        • Huddleston E.M.
        • Gaffo A.L.
        Emerging strategies for treating gout.
        Curr Opin Pharmacol. 2022; 65102241
        • Guy C.D.
        • Suzuki A.
        • Abdelmalek M.F.
        • Burchette J.L.
        • Diehl A.M.
        • NASH C.R.N.
        Treatment response in the PIVENS trial is associated with decreased Hedgehog pathway activity.
        Hepatology. 2015; 61: 98-107
        • Richards H.J.
        Dupuytren’s contracture treated with vitamin E.
        Br Med J. 1952; 1: 1328
        • Steinberg C.L.
        Tocopherols in treatment of primary fibrositis; including Dupuytren’s contracture, periarthritis of the shoulders, and Peyronie’s disease.
        AMA Arch Surg. 1951; 63: 824-833
        • Meurer S.K.
        • Lahme B.
        • Tihaa L.
        • Weiskirchen R.
        • Gressner A.M.
        N-acetyl-L-cysteine suppresses TGF-beta signaling at distinct molecular steps: the biochemical and biological efficacy of a multifunctional, antifibrotic drug.
        Biochem Pharmacol. 2005; 70: 1026-1034
        • Kopp J.
        • Seyhan H.
        • Müller B.
        • et al.
        N-acetyl-L-cysteine abrogates fibrogenic properties of fibroblasts isolated from Dupuytren’s disease by blunting TGF-beta signalling.
        J Cell Mol Med. 2006; 10: 157-165
        • Timmermans P.B.
        • Wong P.C.
        • Chiu A.T.
        • et al.
        Angiotensin II receptors and angiotensin II receptor antagonists.
        Pharmacol Rev. 1993; 45: 205-251
        • Yu L.
        • Border W.A.
        • Anderson I.
        • McCourt M.
        • Huang Y.
        • Noble N.A.
        Combining TGF-beta inhibition and angiotensin II blockade results in enhanced antifibrotic effect.
        Kidney Int. 2004; 66: 1774-1784
        • Zimman O.A.
        • Toblli J.
        • Stella I.
        • Ferder M.
        • Ferder L.
        • Inserra F.
        The effects of angiotensin-converting-enzyme inhibitors on the fibrous envelope around mammary implants.
        Plast Reconstr Surg. 2007; 120: 2025-2033
        • Stephen C.
        • Touil L.
        • Vaiude P.
        • Singh J.
        • McKirdy S.
        Angiotensin receptors in Dupuytren’s disease: a target for pharmacological treatment?.
        J Plast Surg Hand Surg. 2018; 52: 37-39
        • Lee R.C.
        • Doong H.
        • Jellema A.F.
        The response of burn scars to intralesional verapamil. Report of five cases.
        Arch Surg. 1994; 129: 107-111
        • Rayan G.M.
        • Parizi M.
        • Tomasek J.J.
        Pharmacologic regulation of Dupuytren’s fibroblast contraction in vitro.
        J Hand Surg Am. 1996; 21: 1065-1070
        • Gonzalez-Cadavid N.F.
        • Rajfer J.
        Treatment of Peyronie’s disease with PDE5 inhibitors: an antifibrotic strategy.
        Nat Rev Urol. 2010; 7: 215-221
        • Dunkern T.R.
        • Feurstein D.
        • Rossi G.A.
        • Sabatini F.
        • Hatzelmann A.
        Inhibition of TGF-beta induced lung fibroblast to myofibroblast conversion by phosphodiesterase inhibiting drugs and activators of soluble guanylyl cyclase.
        Eur J Pharmacol. 2007; 572: 12-22
        • Ferrini M.G.
        • Kovanecz I.
        • Nolazco G.
        • Rajfer J.
        • Gonzalez-Cadavid N.F.
        Effects of long-term vardenafil treatment on the development of fibrotic plaques in a rat model of Peyronie’s disease.
        BJU Int. 2006; 97: 625-633
        • Ferrini M.G.
        • Vernet D.
        • Magee T.R.
        • et al.
        Antifibrotic role of inducible nitric oxide synthase.
        Nitric Oxide. 2002; 6: 283-294
        • Vernet D.
        • Ferrini M.G.
        • Valente E.G.
        • et al.
        Effect of nitric oxide on the differentiation of fibroblasts into myofibroblasts in the Peyronie’s fibrotic plaque and in its rat model.
        Nitric Oxide. 2002; 7: 262-276
        • Chau D.
        • Mancoll J.S.
        • Lee S.
        • et al.
        Tamoxifen downregulates TGF-beta production in keloid fibroblasts.
        Ann Plast Surg. 1998; 40: 490-493
        • Kuhn M.A.
        • Wang X.
        • Payne W.G.
        • Ko F.
        • Robson M.C.
        Tamoxifen decreases fibroblast function and downregulates TGF(beta2) in Dupuytren’s affected palmar fascia.
        J Surg Res. 2002; 103: 146-152
        • Degreef I.
        • Tejpar S.
        • Sciot R.
        • De Smet L.
        High-dosage tamoxifen as neoadjuvant treatment in minimally invasive surgery for Dupuytren disease in patients with a strong predisposition toward fibrosis: a randomized controlled trial.
        J Bone Joint Surg Am. 2014; 96: 655-662
        • Lorizio W.
        • Wu A.H.
        • Beattie M.S.
        • et al.
        Clinical and biomarker predictors of side effects from tamoxifen.
        Breast Cancer Res Treat. 2012; 132: 1107-1118
        • Martin-Montalvo A.
        • Mercken E.M.
        • Mitchell S.J.
        • et al.
        Metformin improves healthspan and lifespan in mice.
        Nat Commun. 2013; 4: 2192
        • Park I.H.
        • Um J.Y.
        • Hong S.M.
        • et al.
        Metformin reduces TGF-beta1-induced extracellular matrix production in nasal polyp-derived fibroblasts.
        Otolaryngol Head Neck Surg. 2014; 150: 148-153
        • Baeri A.
        • Levraut M.
        • Diazzi S.
        • et al.
        A role for metformin in the treatment of Dupuytren disease?.
        Biomed Pharmacother. 2022; 150112930
        • Ball C.
        • Izadi D.
        • Verjee L.S.
        • Chan J.
        • Nanchahal J.
        Systematic review of non-surgical treatments for early Dupuytren’s disease.
        BMC Musculoskelet Disord. 2016; 17: 345
        • Husby T.
        • Haugstvedt J.R.
        • Fyllingen G.
        • Skoglund L.A.
        Acute postoperative swelling after hand surgery: an exploratory, double-blind, randomised study with paracetamol, naproxen, and placebo.
        Scand J Plast Reconstr Surg Hand Surg. 2001; 35: 91-98
        • Simon A.R.
        • Rai U.
        • Fanburg B.L.
        • Cochran B.H.
        Activation of the JAK-STAT pathway by reactive oxygen species.
        Am J Physiol. 1998; 275: C1640-C1652
        • Thomson G.R.
        Treatment of Dupuytren’s contracture with vitamin E.
        Br Med J. 1949; 2: 1382
        • Kilic T.
        • Parlakpinar H.
        • Polat A.
        • et al.
        Protective and therapeutic effect of molsidomine on bleomycin-induced lung fibrosis in rats.
        Inflammation. 2014; 37: 1167-1178
        • Nanchahal J.
        • Ball C.
        • Davidson D.
        • et al.
        Anti-tumour necrosis factor therapy for Dupuytren’s disease: A randomised dose response proof of concept phase 2a clinical trial.
        EBioMedicine. 2018; 33: 282-288
        • Nanchahal J.
        • Ball C.
        • Swettenham J.
        • et al.
        Study protocol: A multi-centre, double blind, randomised, placebo-controlled, parallel group, phase II trial (RIDD) to determine the efficacy 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; 2: 37
        • Nanchahal J.
        • Ball C.
        • Rombach I.
        • et al.
        Anti-tumour necrosis factor therapy for early-stage Dupuytren’s disease (RIDD): a phase 2b, randomised, double-blind, placebo-controlled trial.
        Lancet Rheumatol. 2022; 4: E407-E416
        • Richeldi L.
        • Costabel U.
        • Selman M.
        • et al.
        Efficacy of a tyrosine kinase inhibitor in idiopathic pulmonary fibrosis.
        N Engl J Med. 2011; 365: 1079-1087
        • Lappi D.A.
        • Martineau D.
        • Maher P.A.
        • et al.
        Basic fibroblast growth factor in cells derived from Dupuytren’s contracture: synthesis, presence, and implications for treatment of the disease.
        J Hand Surg Am. 1992; 17: 324-332
        • Glassberg M.K.
        Overview of idiopathic pulmonary fibrosis, evidence-based guidelines, and recent developments in the treatment landscape.
        Am J Manag Care. 2019; 25: S195-S203
        • Noble P.W.
        • Albera C.
        • Bradford W.Z.
        • et al.
        Pirfenidone in patients with idiopathic pulmonary fibrosis (CAPACITY): two randomised trials.
        Lancet. 2011; 377: 1760-1769
        • Zhou C.
        • Liu F.
        • Gallo P.H.
        • Baratz M.E.
        • Kathju S.
        • Satish L.
        Anti-fibrotic action of pirfenidone in Dupuytren’s disease-derived fibroblasts.
        BMC Musculoskelet Disord. 2016; 17: 469
        • Zhou C.
        • Zeldin Y.
        • Baratz M.E.
        • Kathju S.
        • Satish L.
        Investigating the effects of pirfenidone on TGF-beta1 stimulated non-SMAD signaling pathways in Dupuytren’s disease -derived fibroblasts.
        BMC Musculoskelet Disord. 2019; 20: 135
        • Panigrahi S.
        • Barry A.
        • Multner S.
        • et al.
        Pirfenidone as a potential antifibrotic injectable for Dupuytren’s disease.
        Pharm Dev Technol. 2022; 27: 242-250
        • Cardoneanu A.
        • Burlui A.M.
        • Macovei L.A.
        • Bratoiu I.
        • Richter P.
        • Rezus E.
        Targeting systemic sclerosis from pathogenic mechanisms to clinical manifestations: why IL-6?.
        Biomedicines. 2022; 10
        • Smith M.R.
        Rituximab (monoclonal anti-CD20 antibody): mechanisms of action and resistance.
        Oncogene. 2003; 22: 7359-7368
        • Brenner P.
        • Sachse C.
        • Reichert B.
        • Berger A.
        [Expression of various monoclonal antibodies in nodules and band stage in Dupuytren’s disease].
        Handchir Mikrochir Plast Chir. 1996; 28: 322-327
        • Menzel E.J.
        • Piza H.
        • Zielinski C.
        • Endler A.T.
        • Steffen C.
        • Millesi H.
        Collagen types and anticollagen-antibodies in Dupuytren’s disease.
        Hand. 1979; 11: 243-248
        • Pereira R.S.
        • Black C.M.
        • Turner S.M.
        • Spencer J.D.
        Antibodies to collagen types I–VI in Dupuytren’s contracture.
        J Hand Surg Br. 1986; 11: 58-60
      1. Rituxan (rituximab) [package insert]. Genentech, Inc., 2021: 1