WO2013064585A1 - Traitement de troubles fibroprolifératifs musculo-squelettiques - Google Patents

Traitement de troubles fibroprolifératifs musculo-squelettiques Download PDF

Info

Publication number
WO2013064585A1
WO2013064585A1 PCT/EP2012/071642 EP2012071642W WO2013064585A1 WO 2013064585 A1 WO2013064585 A1 WO 2013064585A1 EP 2012071642 W EP2012071642 W EP 2012071642W WO 2013064585 A1 WO2013064585 A1 WO 2013064585A1
Authority
WO
WIPO (PCT)
Prior art keywords
composition
cell
disease
treatment
inhibitor
Prior art date
Application number
PCT/EP2012/071642
Other languages
English (en)
Inventor
Jagdeep Nanchahal
Kim Suzanne Midwood
Original Assignee
Isis Innovation Ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Isis Innovation Ltd filed Critical Isis Innovation Ltd
Publication of WO2013064585A1 publication Critical patent/WO2013064585A1/fr

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K33/00Medicinal preparations containing inorganic active ingredients
    • A61K33/24Heavy metals; Compounds thereof
    • A61K33/244Lanthanides; Compounds thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/185Acids; Anhydrides, halides or salts thereof, e.g. sulfur acids, imidic, hydrazonic or hydroximic acids
    • A61K31/19Carboxylic acids, e.g. valproic acid
    • A61K31/194Carboxylic acids, e.g. valproic acid having two or more carboxyl groups, e.g. succinic, maleic or phthalic acid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/43Enzymes; Proenzymes; Derivatives thereof
    • A61K38/46Hydrolases (3)
    • A61K38/48Hydrolases (3) acting on peptide bonds (3.4)
    • A61K38/4886Metalloendopeptidases (3.4.24), e.g. collagenase
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system

Definitions

  • This invention relates to the treatment of musculoskeletal fibroproliferative disorders such as fibromatosis and, in particular, Dupuytren's disease.
  • musculoskeletal fibroproliferative disorders such as fibromatosis and, in particular, Dupuytren's disease.
  • Dupuytren's disease which is alternatively known as palmar fibromatosis (or in its established disease state Dupuytren's contracture), is a disease associated with the build up of extracellular matrix materials including collagen on the connective tissue of the hand (the palmar fascia) causing it to thicken and shorten with the physical effect of causing the fingers to curl, most commonly the ring finger and little finger.
  • Dupuytren's disease affects approximately 5% of the white Caucasian population. The commonest manifestation is progressive flexion contracture of the digits of the hand, resulting in significantly compromised function. It affects both males and females, but the incidence is higher in males.
  • Dupuytren's disease has traditionally been invasive surgical techniques. Primarily, the treatment has involved surgical excision of the offending tissue. In severe or recurrent disease, the surgical excision may be combined with excision of the overlying palmar skin and resurfacing of the cutaneous defect with full-thickness skin graft. Surgery is typically followed by prolonged rehabilitation, usually lasting 3 months and complications have been reported in up to 20% of cases. Such surgical correction is the mainstay treatment of later stage disease when secondary changes to tendons and joints have developed. A less invasive surgical intervention is needle fasciotomy in which the fibrous bands (contractures) in connective tissue are divided using the bevel of a needle.
  • Enzymatic cleavage of the affected tissue has been the focus of development to reduce invasiveness associated with surgery and improve recovery time. This approach has led to trials of collagenase.
  • a bacterial collagenase Clostridial collagenase, has been granted FDA approval as XiaflexTM to Pfizer and Auxilium.
  • USRE39941, US5589171 and US6086872 describe the use of bacterial collagenase for the enzymatic cleavage of connective tissue in the treatment of Dupuytren's disease.
  • Bacterial collagenases suffer from certain disadvantages: for example lack non-selective cleaving of various collagen materials including collagen type IV associated with blood vessels; and, in the case of XiaflexTM, possible allergic reactions and potential immunogenicity; and administration may cause haemorrhage whilst the prolonged activity of collagenase limits the dose that can be administered locally due to risk of side effects as the drug disperses.
  • WO 2010/102202 describes a novel temperature sensitive recombinant collagenase in which the activity is observed at significantly below body temperature, but which is comparatively inactive at body temperature.
  • Dupuytren's syndrome can be treated by administering such recombinant collagenase at lower temperatures, which it is claimed restricts the duration of activity, increases the possible local dose and reduces collagenase-related side effects.
  • Non-surgical treatments include steroid injections for early disease and to reduce recurrence, application of vitamin E cream applied as topical therapy, ultrasonic therapy and low-dose radiation therapy (for slowing the progression of early stage disease), such as X-rays and electron beam therapy.
  • Dupuytren's disease Most research for treatments of Dupuytren's disease has focused on detecting pre-disposition to Dupuytren's (e.g. US-A-2004/0161761) and on the extracellular matrices produced, which has resulted in the collagenase-based treatments. There has been very little conclusive insight into potential treatments gained from studies into the biochemical pathway of Dupuytren's disease.
  • Dupuytren's disease and other musculoskeletal fibroproliferative disorders particularly fibromatosis and like diseases including and preferably selected from plantar fibromatosis (or Ledderhose's disease), adhesive capsulitis (frozen shoulder) and Peyronie's disease (fibromatosis of the penis).
  • compositions for use in the treatment or prophylaxis of a musculoskeletal fibroproliferative disorder comprising a cell-cell junction (or intercellular junction) inhibitor or modulator.
  • a cell-cell junction (or intercellular junction) inhibitor or modulator for use in the treatment of a musculoskeletal fibroproliferative disorder.
  • a cell- cell junction (or intercellular junction) inhibitor or modulator in the manufacture of a medicament for the treatment of a musculoskeletal fibroproliferative disorder.
  • a method for the treatment of a musculoskeletal fibroproliferative disorder comprising administering to a patient in need thereof an effective amount of a cell- cell junction (or intercellular junction) inhibitor or modulator.
  • the method comprising locally administering to a patient a cell-cell junction (or intercellular junction) inhibitor or modulator.
  • composition comprising a combination of at least two of an adherens junction inhibitor or modulator, a gap junction inhibitor or modulator and a mechanosensitive ion channel inhibitor or modulator.
  • a method for the treatment of a musculoskeletal fibroproliferative disorder comprising administration to a patient in need thereof a combined effective amount of at least two of an adherens junction inhibitor or modulator, a gap junction inhibitor or modulator and a mechanosensitive ion channel inhibitor or modulator.
  • compositions and methods of the present invention enable musculoskeletal fibroproliferative disorders, such as Dupuytren's (and other fibromatosis and like disease characterized by contracture) to be slowed, halted or reversed.
  • musculoskeletal fibroproliferative disorders such as Dupuytren's (and other fibromatosis and like disease characterized by contracture)
  • Figure 1 is a photograph of a clinical presentation of a patient with Dupuytren's disease.
  • Figure 2 shows images of nodules and cord in an intraoperative view
  • Figure 3 is a chart showing a distribution of a-SMA rich cells in tissue excised from different parts of diseased Dupuytren's tissue;
  • Figures 4A-4D are charts illustrating the effect on contraction and a-SMA and COL1 expression in myofibroblasts and non-palmar dermal fibroblasts of gadolinium;
  • Figures 5A-5F are charts illustrating the effect on contraction and a-SMA expression in myofibroblasts and non-palmar dermal fibroblasts of carbenoxolone; Fig 5B showing distribution of gap junctions in myofibroblasts and non-palmar dermal fibroblasts;
  • Figures 6A-6B illustrate the expression of a-SMA and COL1 and expression of OB cadherin and N-cadherin in myofibroblasts and non-palmar dermal fibroblasts;
  • Figure 6C showing the distribution of adherens junctions in myofibroblasts and non-palmar dermal fibroblasts;
  • Figure 7 is a chart illustrating the rate of contraction of Dupuytren's nodular cells untreated and treated with N-cadherin antibody and E-cadherin antibody.
  • the invention provides for an improved treatment of a musculoskeletal fibroproliferative disorder, especially Dupuytren's disease (or other fibromatosis and like disease such as plantar fibromatosis, adhesive capsulitis and Peyronie's disease), which comprises administration to a patient in need thereof, especially a patient showing signs of early disease state, a therapeutic, prophylactic or progression-inhibitive amount of a cell-cell junction inhibitor or modulator. Further, the invention provides, by administration of a cell-cell junction inhibitor or modulator to a patient having or showing signs of developing
  • a primary surgical intervention e.g. a fasciotomy or fasciectomy
  • primary therapeutic treatment e.g. an extracellular matrix degradation, depletion or cleaving agent, such as a matrix metalloproteinase or collagenase.
  • the invention provides, by administration of a cell-cell junction inhibitor or modulator to a patient, prevention of recurrence of disease as an adjunctive therapy to primary surgical intervention or therapeutic treatment of established disease.
  • the term 'cell-cell junction' is used herein, the term 'intercellular junction' may be used in the alternative.
  • Musculoskeletal fibroproliferative disorders are characterized by excessive or uncontrolled production of extracellular matrix in association with a musculoskeletal structure, often associated with contraction in later stage disease.
  • musculoskeletal fibroproliferative disorders include fibromatosis disorders.
  • the terms 'musculoskeletal fibroproliferative disorders' and 'fibromatosis disease' may be used interchangeably herein, where the context allows).
  • the present invention is concerned with the treatment and, in particular, the inhibition of progression and recurrence (e.g. after primary treatment by surgery or therapy) of such diseases.
  • the present invention is concerned with diseases selected from Dupuytren's disease, plantar fibromatosis, adhesive capsulitis and Peyronie's disease, especially Dupuytren's disease.
  • diseases selected from Dupuytren's disease, plantar fibromatosis, adhesive capsulitis and Peyronie's disease, especially Dupuytren's disease.
  • the remainder of this document will discuss compositions and methods for treatment of musculoskeletal fibroproliferative disorders generally, with specific reference to Dupuytren's disease. Where the context allows, it should be understood that the disclosure may be read also with the generality or other specified diseases in place of Dupuytren's disease.
  • cell-cell junction inhibitor or modulator it is meant an agent that interferes, inhibits or modulates cell-cell junctions or mechanosensitive channels.
  • cell-cell junctions play a critical role in the development of Dupuytren's disease and other musculoskeletal fibroproliferative disorders by facilitating the contraction of and between myofibroblast cells whereby extracellular matrix may be contracted and remodeled to form a cord or other adhesion, such as in frozen shoulder. By inhibiting those cell-cell junction interactions in myofibroblast cells, it is believed that contraction may be inhibited.
  • the cell-cell junction inhibitor or modulator is an inihibitor or modulator of one or more of adherens junctions, gap junctions or mechanosensitive ion channels.
  • mechanosensitive channels are not, strictly speaking inter-cellular channels, they are implicated in cell-cell junction activity since it is believed that they are associated with adherens junctions insofar as when the cell feels contraction, mechanosensitive channels open to allow Ca 2+ ion into the cell, which is necessary for effecting cell contraction via a-smooth muscle actin (a-SMA). Tension may be exerted on the cell via the adherens junction by a neighbouring cell.
  • the treatment comprises administering to a patient a combination of inihibitors or modulators of adherens junctions and/or inhibitors or modulators of gap junctions and/or inhibitors or modulators of mechanosensitive ion channels.
  • the administration according to the treatment of the present invention is local administration (e.g. by injection into or adjacent to the affected tissue). Most preferably, e.g. for the treatment or prevention of
  • Dupuytren's disease the delivery is by injection into a, the or each nodule site (where, typically, there is an accumulation of myofibroblasts).
  • Any suitable dose may be delivered that is effective in the treatment or prophylaxis or progression inhibition of the musculoskeletal fibroproliferative disease according to the present invention.
  • the composition comprises a mechanosensitive ion channel inhibitor or modulator.
  • mechanosensitive ion channel inhibitor it is meant an agent that blocks or inhibits mechanosensitive ion channel inhibitor formation, maintenance or function by blocking a component necessary for the formation or function of a mechanosensitive ion channel inhibitor.
  • Any suitable mechanosensitive ion channel inhibitor may be used in accordance with the present invention.
  • the mechanosensitive ion channel inhibitor may be selected from calcium channel blockers, a low affinity selective compound isolate from tarantula venom, GsMTx-4, aminoglycosides, amiloride or gadolinium.
  • the mechanosensitive ion channel inhibitor is gadolinium, optionally administered in a complex such as diethylenetriamine pentaacetic acid
  • the mechanosensitive ion channel inhibitor may be administered in any suitable dose.
  • each administration e.g. injection to each nodule site
  • each site may be injected with 0.05 to 0.5 ml, more preferably 0.1 to 0.3 ml of agent formulation.
  • the composition comprises an adherens junction inhibitor or modulator, preferably an adherens junction antagonist.
  • an adherens junction antagonist it is meant an agent that blocks or inhibits adherens junction formation, maintenance or function by blocking a component necessary for the formation or function of adherens junctions.
  • the adherens junction inhibitor or modulator comprises one or more anti-cadherin or, preferably one or more cadherin antagonist.
  • An anti-cadherin agent is one that inhibits or blocks the formation or function of a cadherin.
  • the anti-cadherin or cadherin antagonist may be effective against any cadherin which may be implicated in myofibroblast contraction, e.g.
  • the anti- cadherin has broad spectrum anti-cadherin activity susch as ⁇ -catenin antibody, a- catenin antibody or vinculin antibody.
  • the anti-cadherin may be broad spectrum or specific cadherin antibodies (e.g. specific to any of the cadherins referred to above), such as an OB- cadherin antibody or an N-cadherin antibody such as those (or producible by the method) described in WO-A-2011/119888 or WO-A-2010/054377, the disclosures of which antibodies and antibody production methods are incorporated herein by reference.
  • specific cadherin antibodies e.g. specific to any of the cadherins referred to above
  • an OB- cadherin antibody or an N-cadherin antibody such as those (or producible by the method) described in WO-A-2011/119888 or WO-A-2010/054377, the disclosures of which antibodies and antibody production methods are incorporated herein by reference.
  • the anti-cadherin or cadherin antagonist is effective against N-cadherin and/or OB-cadherin and more preferably the anti-cadherin is an N-cadherin antagonist or an OB-cadherin antagonist. Still more preferably the anti-cadherin is an N-cadherin antagonist or inhibitor.
  • the anti-cadherin or cadherin antagonist, especially the N-cadherin antagonist is a cadherin antibody, preferably an N-cadherin antibody.
  • N-cadherin antibody is the commercially available monoclonal anti-N-cadherin antibody, clone GC-4, available from Sigma- Aldrich ® (and having the product number 3865 and MDL no: MFC D00164511).
  • the inventors have surprisingly found that N-cadherin antagonism is effective in reducing contraction of myofibroblasts from Dupuytren's patients.
  • Previous studies e.g. Hinz et al, Mol. Biol. Cell, 2004 15, 4310-20
  • the anti-cadherin preferably the anti-N-cadherin may be administered in any suitable dose.
  • a suitable amount may be determined by methods as known in the art, but may comprise, for example, administering an amount of from 1 to 500 ⁇ g, optionally 5 to 250 ⁇ g, 10 to 100 ⁇ g or 50 to 75 ⁇ g.
  • the composition comprises a Gap junction inhibitor or modulator.
  • Gap junction inhibitor it is meant an agent that blocks or inhibits Gap junction formation, maintenance or function by blocking a component necessary for the formation or function of a Gap junction.
  • the Gap junction inhibitor may be selected from one or a combination of a glycerrhetinic acid, a fatty alcohol such as 2-octanol, antimalarials such as mefloquine or primaquine, thapsigargin, halothane and amiloride.
  • Gap junction inhibitors useful in accordance with the present invention include anti-connexin agents such as connexin inhibitors, in particular connexin 43 inhibitors.
  • the anti-connexin is an antibody to a connexin, preferably an antibody to connexin 43.
  • the connexin inhibitors are selected from glycerrhetinic acid and derivatives thereof and more preferably the connexin inhibitor is carbenoxolone (18-a-glycyrrhetinic acid).
  • each administration e.g. injection to each nodule site
  • each administration may comprise a dose in the range of 0.005 to 100 ⁇ , preferably, 0.01 to 50 ⁇ , more preferably, 0.05 to 10 ⁇ and optionally 2 to 8 ⁇ .
  • the agent defined according to any of the above embodiments is an antibody
  • it may be a monoclonal antibody or fragment thereof; a chimeric monoclonal antibody (such as a human-murine chimeric monoclonal antibody); a fully human monoclonal antibody; a recombinant human monoclonal antibody; or a humanized antibody fragment, such as a Fab, F(ab')2 or Fv fragment.
  • the fragment may pegylated or encapsulated (e.g. for stability and/or sustained release).
  • the treatment comprises administering to a patient a combination of inihibitors or modulators of N-cadherin and inhibitors or modulators of mechanosensitive ion channels, such as calcium channel blockers or gadolinium.
  • cell-cell junction inhibitor or modulator in the treatments of the present invention is due to the crucial role played by cell-cell junctions in contractile effect of differentiated myofibroblast cells, which are understood also to be the main culprits in induction of uncontrolled matrix generation in Dupuytren's disease (and other fibromatosis diseases).
  • the inventors have demonstrated that certain cell-cell interactions affect contracture and can be inhibited resulting in reduced rates of contracture in Dupuytren's nodular cells.
  • Dupuytren's disease occurs in people with genetic predisposition and further risk factors to manifestation of Dupuytren's disease include local trauma, poor lifestyle (e.g. smoking and drinking alcohol and poor diet), liver disease and diabetes.
  • Established disease presents as flexion contracture which may typically be presented as contracture of the metacarpophalangeal joints (MCPJ) alone, less frequently contracture of the proximal interphalangeal joints (PIP J) alone, and often both.
  • MCPJ metacarpophalangeal joints
  • PIP J proximal interphalangeal joints
  • myofibroblasts are concomitant with early and active disease and that such cells are implicated in proliferative extra-cellular matrix (ECM) generation or deposition and, in particular, collagen deposition.
  • ECM extra-cellular matrix
  • TGF- ?1 leads to the development of the myofibroblast phenotype.
  • Myofibroblasts are also believed to be responsible for contractile behavior.
  • Myofibroblasts characteristically express a-smooth muscle actin (a-SMA), which is the actin isoform typical of vascular smooth muscle cells.
  • a-SMA is believed to be the protein responsible for the contractility of
  • myofibroblasts and is the most reliable marker for myofibroblasts.
  • the present invention preferably comprises a composition and method for treating, and more preferably inhibiting or halting the progression or recurrence of, musculoskeletal fibroproliferative disorders, such as fibromatosis disease, especially Dupuytren's disease, by administering to a patient a therapeutic, prophylactic or progression-inhibiting amount of a cell-cell junction inhibitor or modulator (preferably an N-cadherin antagonist such as a anti-N- cadherin antibody and/or a mechanosensitive ion channel inhibitor such as gadolinium).
  • a cell-cell junction inhibitor or modulator preferably an N-cadherin antagonist such as a anti-N- cadherin antibody and/or a mechanosensitive ion channel inhibitor such as gadolinium.
  • the administration is local administration (e.g. by injection into or adjacent to the affected tissue).
  • a first main embodiment of the invention comprises a composition and method for treating early disease state musculoskeletal fibroproliferative disorders, especially early disease state Dupuytren's disease, by administering to a patient presenting early state disease, e.g. prior to the presence of palpable cord, an effective amount of a cell-cell junction inhibitor or modulator (preferably an N- cadherin antagonist such as a anti-N-cadherin antibody and/or a mechanosensitive ion channel inhibitor such as gadolinium).
  • a cell-cell junction inhibitor or modulator preferably an N- cadherin antagonist such as a anti-N-cadherin antibody and/or a mechanosensitive ion channel inhibitor such as gadolinium.
  • a composition comprising a cell- cell junction inhibitor or modulator (preferably an N-cadherin antagonist such as a anti-N-cadherin antibody and/or a mechanosensitive ion channel inhibitor such as gadolinium) may be administered to a patient for preventing disease progression (to established disease state) and resultant flexion contracture.
  • a cell- cell junction inhibitor or modulator preferably an N-cadherin antagonist such as a anti-N-cadherin antibody and/or a mechanosensitive ion channel inhibitor such as gadolinium
  • the method comprise local administration (e.g. by injection) directly into the clinical nodule(s).
  • the method further comprises administering to the patient, preferably locally (and more preferably directly to the clinical nodule(s) identified), an extracellular matrix degradation, depletion or cleavage agent, which is preferably a collagen degradation, depletion or cleavage agent and may be, for example a matrix metalloproteinase (MMP) and/or a collagenase (but may be, for example, a MMP or collagenase up-regulating or inducing agent).
  • MMP matrix metalloproteinase
  • collagenase but may be, for example, a MMP or collagenase up-regulating or inducing agent.
  • the matrix metalloproteinase or collagenase may disrupt collagen and extra- cellular matrix local to the clinical nodule(s) thereby enhancing access of administered a cell-cell junction inhibitor or modulator to the proliferative fibrotic foci and thus enhance efficacy of treatment. It is believed that administration of the cell-cell junction inhibitor or modulator in this manner may be considered prophylactic or progression halting or inhibiting treatment.
  • the primary treatment is a cell-cell junction inhibitor or modulator to which the extracellular matrix degradation or cleavage agent is preferably adjunctive.
  • a cell-cell junction inhibitor or modulator preferably an N-cadherin antagonist such as a anti-N- cadherin antibody and/or a mechanosensitive ion channel inhibitor such as gadolinium
  • an extracellular matrix degradation, depletion or cleavage agent e.g. matrix metalloproteinase and/or coUagenase
  • the cell-cell junction inhibitor or modulator and the extracellular matrix degradation, depletion or cleavage agent e.g. coUagenase
  • both a cell-cell junction inhibitor or modulator preferably an N-cadherin antagonist such as a anti-N-cadherin antibody and/or a
  • mechanosensitive ion channel inhibitor such as gadolinium
  • the extracellular matrix degradation, depletion or cleavage agent e.g. coUagenase
  • they may be administered locally, for example by injection.
  • they may be administered
  • a composition comprising both a cell-cell junction inhibitor or modulator and coUagenase (e.g. by injectable solution) or by applying two separate compositions at the same time.
  • the cell-cell junction inhibitor or modulator and the extracellular matrix degradation, depletion or cleavage agent e.g. coUagenase
  • coUagenase e.g. coUagenase
  • the extracellular matrix degradation, depletion or cleavage agent e.g. coUagenase
  • the cell-cell junction inhibitor or modulator preferably an N-cadherin antagonist such as a anti-N-cadherin antibody and/or a mechanosensitive ion channel inhibitor such as gadolinium
  • the cell-cell junction inhibitor or modulator preferably an N-cadherin antagonist such as a anti-N-cadherin antibody and/or a mechanosensitive ion channel inhibitor such as gadolinium
  • a cell-cell junction inhibitor or modulator preferably an N-cadherin antagonist such as an anti-N-cadherin antibody and/or a
  • a mechanosensitive ion channel inhibitor such as gadolinium
  • the extracellular matrix degradation, depletion or cleavage agent are administered simultaneously for the treatment of early disease state musculoskeletal fibroproliferative disorders.
  • a composition is provided for local administration (e.g.
  • injectable solution, sustained release composition or implant for treating early disease state musculoskeletal fibroproliferative disorders, preferably Dupuytren's disease, which composition comprises an effective amount of a cell-cell junction inhibitor or modulator (or configured to release an effective amount of a cell-cell junction inhibitor or modulator if, for example, the composition is a sustained release composition) optionally in combination with an extracellular matrix degradation, depletion or cleavage agent (preferably a matrix metalloproteinase and/or collagenase) preferably in an adjunctive amount and a pharmaceutically acceptable carrier.
  • an extracellular matrix degradation, depletion or cleavage agent preferably a matrix metalloproteinase and/or collagenase
  • the cell-cell junction inhibitor or modulator (preferably an N-cadherin antagonist such as an anti-N- cadherin antibody and/or a mechanosensitive ion channel inhibitor such as gadolinium) is provided in an amount effective to inhibit disease progression without inducing systemic complications.
  • the cell-cell junction inhibitor or modulator is provided in an amount to reduce progression or recurrence by at least 30%, preferably at least 50% and more preferably at least 70%.
  • an effective amount of a cell-cell junction inhibitor or modulator is that which will result in a reduction in clinical nodule size (e.g. at least a 20%), or even at least a 50%>, reduction in size, as measured by degree of protrusion or lateral or longitudinal extent) in up to two weeks post administration.
  • Efficacy of a cell-cell junction inhibitor or modulator treatment preferably is observable by an overall reduction in the progression of disease. Without being bound by theory, it is believed that myofibroblasts require tension to develop and persist and the cell-cell junction inhibitors used in the present invention can reduce local tension thereby reducing the tendency of new myofibroblasts to develop.
  • an extracellular matrix degradation, depletion or cleavage agent e.g. a matrix metalloproteinase and/or coUagenase
  • a cell-cell junction inhibitor or modulator adjunctive amount by which it is meant an amount effective to enhance the efficacy of the a cell-cell junction inhibitor or modulator.
  • the extracellular matrix degradation, depletion or cleavage agent e.g. matrix metalloproteinase or coUagenase
  • the extracellular matrix degradation, depletion or cleavage agent is provided in an amount of up to 1-2 mg.
  • the extracellular matrix degradation, depletion or cleavage agent e.g. matrix metalloproteinase or coUagenase
  • the extracellular matrix degradation, depletion or cleavage agent is administered in an amount significantly below (e.g. 0.01 to 0.5 times) the extracellular matrix degradation, depletion or cleavage agent (e.g. matrix metalloproteinase or coUagenase) dose that would be required to achieve an enzymatic fasciotomy in established disease state fibromatosis.
  • the extracellular matrix degradation, depletion or cleavage agent e.g. matrix metalloproteinase or coUagenase
  • the extracellular matrix degradation, depletion or cleavage agent may assist the a cell-cell junction inhibitor or modulator (preferably an N-cadherin antagonist such as a anti-N- cadherin antibody and/or a mechanosensitive ion channel inhibitor such as gadolinium) in accessing the cell mass, as well as assisting in disaggregating of the extracellular matrix of the clinical nodule.
  • a cell-cell junction inhibitor or modulator preferably an N-cadherin antagonist such as a anti-N- cadherin antibody and/or a mechanosensitive ion channel inhibitor such as gadolinium
  • a second main embodiment of the invention comprises a composition and method for treating established disease state Dupuytren's (or other musculoskeletal fibroproliferative) disease by administering to a patient an effective amount of a cell-cell junction inhibitor or modulator (preferably an N- cadherin antagonist such as a anti-N-cadherin antibody and/or a mechanosensitive ion channel inhibitor such as gadolinium), preferably in combination with, simultaneous to, sequentially with, in association with, concomitantly with, in combined administration with or adjunctive to surgical fasciectomy, a fasciotomy and/or a extracellular matrix degradation, depletion or cleavage agent (e.g.
  • a cell-cell junction inhibitor or modulator preferably an N- cadherin antagonist such as a anti-N-cadherin antibody and/or a mechanosensitive ion channel inhibitor such as gadolinium
  • the method comprises surgical fasciectomy, needle fasciotomy or extracellular matrix degradation, depletion or cleavage agent (e.g. a matrix metalloproteinase or a collagenase) administration, which provides improvement (i.e. enabling greater extension of the affected digits), more preferably correction (i.e. to within 5° of full extension) and most preferably full correction (complete extension) of the established disease.
  • the method comprises administration of extracellular matrix degradation, depletion or cleavage agent (e.g. a matrix metalloproteinase or a collagenase) to sites local to the disease site.
  • a cell-cell junction inhibitor or modulator may be provided for combined treatment by simultaneous, sequential or separate administration, e.g. for combined, concomitant or adjunctive therapy.
  • a cell-cell junction inhibitor or modulator is adjunctive to the extracellular matrix degradation, depletion or cleavage agent (e.g. a matrix metalloproteinase or a collagenase) treatment.
  • a cell-cell junction inhibitor or modulator is a cell-cell junction inhibitor or modulator
  • an N-cadherin antagonist such as a anti-N-cadherin antibody and/or a mechanosensitive ion channel inhibitor such as gadolinium
  • a depletion or cleavage agent e.g. a matrix metalloproteinase or a collagenase
  • depletion or cleavage agent e.g. a matrix metalloproteinase or a collagenase
  • up to 14 days before or after administration of the extracellular matrix degradation, depletion or cleavage agent
  • a matrix metalloproteinase or a collagenase still more preferably at least 30 minutes before or after administration of the extracellular matrix degradation, depletion or cleavage agent (e.g. a matrix metalloproteinase or a collagenase) and more preferably after the extracellular matrix degradation, depletion or cleavage agent (e.g. a matrix metalloproteinase or a collagenase), e.g. in the period 4 hours to 7 days after the extracellular matrix degradation, depletion or cleavage agent (e.g. a matrix metalloproteinase or a collagenase), whereby administration of a cell-cell junction inhibitor or modulator may gain better access to the disease site but be administered at a point when myofibroblasts can be optimally inhibited.
  • depletion or cleavage agent e.g. a matrix metalloproteinase or a collagenase
  • a cell-cell junction inhibitor or modulator is provided in an amount effective to inhibit disease recurrence without inducing systemic complications.
  • an effective amount of a cell-cell junction inhibitor or modulator is that which will prevent a palpable or as measured (e.g. by ultrasound scan) increase in clinical nodule presentation and/or in clinical nodule size (e.g. a 25% increase in size, as measured by degree of protrusion or lateral or longitudinal extent) in up to two to twelve weeks post administration.
  • Efficacy of a cell-cell junction inhibitor or modulator treatment preferably is observable by an overall prevention in the recurrence of disease (e.g. post surgery or enzymatic treatment).
  • re-establishment of established state disease manifested by flexion contracture can be managed or prevented, e.g. flexion contracture maintained to 10° or less, more preferably 5° or less further contraction compared with post- correction treatment extent, within a period after administration of the a cell-cell junction inhibitor or modulator, e.g. up to 6 weeks, preferably up to 6 months.
  • repeat administrations may be provided in order to achieve this (e.g. two to four weekly).
  • the treatment comprises administration local to disease site of an extracellular matrix degradation, depletion or cleavage agent (e.g. a matrix metalloproteinase or a collagenase).
  • the extracellular matrix degradation, depletion or cleavage agent e.g. a matrix metalloproteinase or a collagenase
  • should be administered in an amount sufficient to enable improvement and/or correction of disease-associated contraction e.g. to 5° or less of full extent in the case of Dupuytren's
  • an extracellular matrix degradation, depletion or cleavage agent such as a coUagenase (e.g.
  • Clostridium coUagenase is provided for local administration in an amount of up to 10 mg administered in one or more locations along each contracture, preferably from 0.1 to 5 mg per administration and more preferably from 0.15 to 2 mg and most preferably 0.5 to 1 mg.
  • extracellular matrix degradation, depletion or cleavage agent e.g. a matrix metalloproteinase or a coUagenase
  • a cell-cell junction inhibitor or modulator administered for recurrence inhibition
  • a single combined dose of the extracellular matrix degradation, depletion or cleavage agent (e.g. a matrix metalloproteinase or a coUagenase) and a cell-cell junction inhibitor or modulator e.g. a matrix metalloproteinase or a coUagenase
  • an extracellular matrix degradation, depletion or cleavage agent may be administered (e.g. injected) into diseased cord tissue in an effective amount, whilst a cell-cell junction inhibitor or modulator may be administered (e.g. injected) into clinical nodule(s) and/or cord tissue in a recurrence-inhibitory amount.
  • the extracellular matrix (ECM) degradation, depletion or cleavage agent may be any suitable agent capable of degrading, cleaving or causing or inducing degradation or cleavage of extracellular matrix, including collagen.
  • the ECM degradation or cleavage agent may be an ECM degradation enzyme or an ECM degradation enzyme expression up-regulator (e.g. relaxin).
  • the ECM degradation or cleavage agent is a matrix metalloproteinase or a coUagenase, more preferably a coUagenase, such as a bacterial coUagenase (e.g.
  • co agenase is time or temperature dependent or is
  • photo dynamically activated or deactivated to allow higher local doses to be administered without systemic or long-lasting side-effects.
  • it is a
  • Cathepsin-L or a mutant or recombinant thereof examples include those described in: GB-A-2323530, US 5589171, USRE39941, US6086272 & WO-A-2010/102262 (and for established disease optionally in the amounts described therein, the disclosure of which collagenases and amounts and modes of administration are incorporated herein by reference).
  • early disease state it is meant that indications of disease are present, e.g. histological markers or more particularly clinical nodules in tissue, but in the absence of, for example, palpable cord or significant contracture.
  • early disease state Dupuytren's disease it is meant that indications of Dupuytren's disease are present, e.g. histological markers or more particularly clinical nodules in palmar and/or digital tissue, but in the absence of significant (e.g. at least 5°) flexion contracture (or, for example, palpable cord).
  • established disease state it is meant that clinical nodules are present, palpable cord is present and contracture is evident.
  • Dupuytren's disease it is meant that clinical nodules are present on the palm and digits of the hand and flexion contracture is evident (e.g. at least 5°).
  • clinical nodule it is meant a palmar or digital nodule evident as a palpable subcutaneous lump.
  • histological nodule it is meant a collection of cells (mainly myofibroblast cells with some inflammatory cells such as macrophages and mast cells) which may range from tiny foci of cells to larger collections of cells, but not clinically palpable.
  • the initial clinically palpable nodule(s) is the focus of proliferating fibroblasts in disease progression, but that numerous histological nodules will form at various locations in the palm and/or digits which will ultimately contribute to cord formation, contraction and flexion contracture.
  • 'nodule' may be clinical or histological nodules (or either) as will be apparent from the context.
  • a first embodiment may relate to a composition and method for treating
  • Dupuytren's disease characterized by joint contractures of less than 20° and a second embodiment may relate to a composition and method for treating
  • Dupuytren's disease characterized by joint contractures of at least 20°.
  • the contracture of 20° is identified as a transition phase, since at less than 20° contracture, many patients may choose to stop progression of the disease without wishing to undergo surgery since their mobility and operative use of the hand is still largely adequate, whilst at greater than 20°, many patients will find surgery or other collagen depleting therapy (such enzymatic fasciotomy) essential to restore full function to the hand.
  • cell-cell junctions are an optimal therapeutic target for early Dupuyten's disease (i.e. early disease state).
  • Dupuytren's disease an ideal combination is a matrix metalloproteinase such as collagenase with a cell-cell junction inhibitor reduce the impact of any recurrence, which is typically associated with enzymatic fasciotomy.
  • the cell-cell junction inhibitor or modulator may be provided in a multiple administrations over an extended (or continuous) term in order to prevent or inhibit disease progression or recurrence.
  • intermittent treatment may be provided by, e.g. low-dose fortnightly, monthly or six-monthly administration.
  • continuous treatment may be provided by low-dose releasing sustained or delayed intermittent release implant or patch.
  • repeat doses may be initiated by signs of disease progression in the early disease state and may optionally comprise a combined extracellular matrix degradation or cleavage agent (e.g. a matrix metalloproteinase or a coUagenase) and cell-cell junction inhibitor or modulator treatment (e.g. consistent with the first embodiment described above).
  • a combined extracellular matrix degradation or cleavage agent e.g. a matrix metalloproteinase or a coUagenase
  • cell-cell junction inhibitor or modulator treatment e.g. consistent with the first embodiment described above.
  • the progression of early disease state disease e.g. Dupuytren's disease
  • a cell-cell junction inhibitor or modulator e.g., a cell-cell junction inhibitor or modulator
  • the cell-cell junction inhibitor or modulator may be administered separately or simultaneously in combination with or adjunctively to a coUagenase and/or matrix metalloproteinase.
  • a coUagenase especially a photo- responsive or temperature dependent coUagenase, may be administered for local effect to enhance the cell-cell junction inhibitor or modulator disease progression inhibition effect by enhancing access to treatment sites by cleaving early stage extracellular matrix formation.
  • a temperature dependent coUagenase is one which (typically a recombinant or mutant coUagenase) has coUagenase activity dependent upon temperature and typically is active at below body temperature, e.g. at 25°C and below, thereby allowing extremely high doses of coUagenase to act very locally (e.g. by injecting at the disease site at say 20°C without having any systemic action or other side effects associated with longevity of action).
  • composition and method of the present invention may utilise any suitable means of administration, which is preferably local.
  • the cell-cell junction inhibitor or modulator should be administered locally, e.g. by applying directly into a surgical incision during surgery, by injection (preferably directly into the clinical nodule(s) and/or cord tissue), by release from a sustained and/or delayed release particles or device that may be implanted into or close to the disease site or a sustained and/or delayed release patch formulation, by topical application or any other suitable route.
  • a composition is preferably suitably formulated and typically comprises the required dose of cell-cell junction inhibitor or modulator along with a pharmaceutical acceptable carrier or excipient.
  • Formulations for parenteral administration may typically comprise a sterile aqueous preparation of the active ingredient, which is preferably isotonic with the blood of the recipient.
  • Formulations for intra-articular administration may be in the form of a sterile aqueous preparation of the active ingredient.
  • Formulations suitable for topical administration may include liquid and semi liquid preparations such as liniments, lotions and applications; oil-in- water and water-in- oil emulsions such as creams, ointments and pastes; and solutions and suspensions.
  • a formulation for frequent, e.g. daily, periodic or occasional (preferably daily), topical application to the musculoskeletal fibroproliferative disorder area e.g. the hands, and in particular palms and digits, in the case of Dupuytren's disease
  • the formulation comprising a cell-cell junction inhibitor or modulator suitable for topical administration (e.g. selected from such cell-cell junction inhibitor or modulator defined above) and a suitable excipient.
  • the formulation may be provided as a cream or lotion, a patch or a medicated glove (in which the glove is impregnated for release of the active component from the internal surface).
  • the formulation comprises cell-cell junction inhibitor or modulator in a concentration for administration by topical application of a low dose, such as 0.001 to 0.05, preferably 0.001 to 0.01, of the dose of the selected cell-cell junction inhibitor or modulator that would yield a systemic response.
  • compositions and methods of the present invention may further comprise further active ingredients that may be effective in the treatment or progression-inhibition of musculoskeletal fibroproliferative disorders such as Dupuytren's disease.
  • a cell-cell junction inhibitor or modulator and an agent of the vascular endothelial growth factor family, such as VEGF-A, VEGF-B, VEGF-C or VEGF-D or an agent encoding said VEGF or a functional fragment thereof (such as described in WO-A-2004/082705), which combination is preferably a development retarding combination (or composition) for use in association with surgery, or needle or enzyme fasciotomy.
  • an activator of PPARy such as pioglitazone for reducing myofibroblast populations local to the disease site.
  • a method of treatment and composition comprising the cell-cell junction inhibitor or modulator in one or more compositions and/or formulations as described above for the treatment of keloid and/or hypertrophic scars.
  • Such methods and composition may be administered in a similar manner to that described above.
  • myofibroblast cells Dupuytren's nodular cells
  • HDF cells non- palmar human dermal fibroblast cells
  • Figure 1 illustrates a clinical presentation of Dupuytren's disease in a patient's little finger, with contracture of the metacarpophalangeal and proximal interphalangeal joints. The clinical presentation is also clearly illustrated in Figure 1 of Rehman et al. Arthritis Research & Therapy 2011, 13:238.
  • FIG. 2 shows: A: intraoperative view of Dupuytren's cord, with location of proximal interphalangeal joint (PIP J; 1) marked; B: Low magnification photomicrograph of histological section stained for a- smooth muscle actin. A collection of a-SMA rich cells in a nodule is located in the vicinity of the PIP J; C: High magnification view of nodular area, showing a -SMA positive cells
  • Dupuytren's cords correlation with digital contracture', J Hand Surg Am 34:1785- 1794) has shown that cells from and close to the nodules in Dupuytren's patients have a significantly higher percentage of a-SMA positive cells than non-nodular cells (e.g. cord cells) implying such nodular cells have a higher myofibroblast concentration.
  • a chart of total cell count for each of several different cell types and the percentage of a-SMA positive cells within that population is shown in Figure 3.
  • Gadolinium was used as the mechanosensitive ion channel blocker. It is believed that gadolinium blocks cationic mechanosensitive channels possibly through binding to anionic phospholipids.
  • An isometric force contraction experiment was conducted using the method set out in Methods below.
  • Figure 4 illustrates that blocking mechanosensitive junctions downregulates myofibroblast activity.
  • gadolinium 300 ⁇ inhibited the contractility of myofibroblasts (Figure 4A) in a dose-dependent manner, but not HDF ( Figure 4B) [data shown are from n>3 patients (each in triplicate) and are expressed as mean ⁇ S.E.M. ** P ⁇ 0.01, *** P ⁇ 0.001].
  • Gene expression was assessed by quantitative RT-PCR and data are presented as fold change compared to GAPDH, and normalized to expression in untreated myofibroblasts or HDF respectively. Protein expression was assessed by western blotting using vimentin as a loading control. Gels shown are representative of gels from 3 patients. Data are shown as the mean ⁇ S.E.M from n>3 patients (each assay was performed in triplicate). ** P ⁇ 0.01.
  • the mechanosensitive ion channel inhibitor gadolinium inhibited contraction of collagen gel populated by Dupuytren's nodular cells (myofibroblast cells) whilst having no impact on contraction rates of non- palmar human dermal fibroblast cell populations from the same patients.
  • gadolinium and mechanosensitive junction inhibitors more generally may be effective in reducing the contraction and thus useful in treating or inhibiting the progression of Dupuytren's disease and other musculoskeletal fibroproliferative disorders.
  • Carbenoxolone was used as the gap junction inhibitor.
  • Carbenoxolone is a saponin that inhibits gap junction communication by disassembling gap junction plaques composed of connexin 43.
  • Connexin 43 in the plaques is phosphorylated and, without being bound by theory, carbenoxolone is believed to act by dephosphorylating and not by inhibiting phosphorylation.
  • Figures 5 C and D Carbenoxolone inhibited the contractility of myofibroblasts (Figure 5C), but not HDF ( Figure 5D). Data are shown as the mean ⁇ S.E.M from n>3 patients (each assay was performed in triplicate). ** P ⁇ 0.01, *** P ⁇ 0.001.
  • Figure 10E Carbenoxolone ( ⁇ ) reduced a-SMA and COL1 mRNA and a-SMA protein in myofibroblasts compared to HDF. Carbenoxolone also reduced a-SMA mRNA and protein expression in HDF.
  • carbenoxolone (a gap junction inhibitor) inhibited contraction of collagen gel populated by Dupuytren's myofibroblast cells (nodular cells) whilst having no impact on contraction rates of non-palmar dermal fibroblast cell (HDF) populations from the same patients. Furthermore, the inhibitory effect of the gap junction inhibition by carbenoxolone on Dupuytren's myofibroblast (nodule) cell contraction is dose dependent. It may be concluded that inhibition of contracture in Dupuytren's myofibroblast (nodule) cells may be achieved by a gap junction inhibitor (such as carbenoxolone).
  • a gap junction inhibitor such as carbenoxolone
  • MF myofibroblasts
  • HDF non-palmar human dermal fibroblasts
  • Figure 6 A shows baseline levels of a-SMA and COL1 mRNA, and a-SMA protein, were significantly higher in Dupuytren's myofibroblasts (MF) compared to human dermal fibroblasts (HDF).
  • Figure 6B Baseline levels of OB cadherin mRNA and protein were also higher in MF compared to HDF, whereas N cadherin expression was significantly higher in HDF compared to MF.
  • Gene expression was assessed by quantitative RT-PCR and data are presented as fold change compared to GAPDH, and normalized to expression in HDF. Protein expression was assessed by Western blotting using vimentin as a loading control. Gels shown are representative of gels from 3 patients.
  • the present examples used early passage primary human cells from patients with Dupuytren's disease whereas in earlier publications subcutaneous rat fibroblasts up to passage 7 were compared to myofibroblasts generated by exposure to TGF- ⁇ over 4 days in one case, whilst embryonic rat subcutaneous and lung fibroblasts and myofibroblasts generated by exposure to TGF- ⁇ over 5 days were studied in another.
  • intercellular adherence between myofibroblasts is crucial to their function such that blockade of even the relatively few N cadherin- containing adherens junctions had a profound effect on contractility.
  • N-cadherin inhibitors and antagonists and antibodies
  • this illustrates the surprising reduction in contractility of myofibroblasts by an N-cadherein inihibitor
  • N-cadherin inhibitors and antagonists and antibodies
  • Tissue samples were obtained following informed consent (REC 07/H0706/81).
  • Dupuytren's nodular tissue and matched full-thickness skin were obtained from patients with Dupuytren's disease undergoing dermofasciectomy.
  • HDF Human dermal fibroblasts
  • Dupuytren's myofibroblasts were isolated from a-SMA-rich nodules (Verjee et al (2010) J Cell Physiol 224:681-90). Tissue samples were dissected into small pieces and digested in Dulbecco's modified Eagle's medium (DMEM) (Lonza) with 1% penicillin-streptomycin (PAA) and 5% fetal bovine serum (FBS) (Gibco) with type I collagenase (Worthington Biochemical Corporation) + DNase I (Roche Diagnostics) for up to 2h at 37°C. Cells were cultured in DMEM with 10% FBS and 1% penicillin-streptomycin at 37°C in a humidified incubator with 5% CO 2 . Cells up to passage 2 were used for experiments.
  • DMEM Dulbecco's modified Eagle's medium
  • PAA penicillin-streptomycin
  • FBS fetal bovine serum
  • Gibco fetal bovine serum
  • Cell populated matrices were cultured in DMEM with 10% FBS and 1% penicillin- streptomycin at 37°C in a humidified incubator for 24h with 5% CO 2 and treated with anti-N-cadherin antibody (Sigma), anti-E-cadherin antibody (Invitrogen), IgG isotype control, gadalolinium or carbenoxolone (Sigma). Cell-mediated force generation was abolished by addition of a saturating dose of 2( ⁇ g/ml cytochalasin D (Sigma) to fibroblast populated collagen matrices (Townley et al. (2009) J Hand Surg Eur Vol 34:783-7). Experiments using each patient sample were performed in triplicate.
  • Inventoried TaqMan® Gene expression Assays were used for a-SMA (Hs00426835-gl), COL1 (Hs00164004-ml), N-cadherin (Hs00362037-ml), OB-cadherin
  • Cell lysates were prepared in lysis buffer (25 mM HEPES (pH 7.0), 150 mM NaCl, and 1% Nonidet P-40), containing protease inhibitor cocktail (Roche Biochemicals) and then electrophoresed on 10% SDS polyacrylamide gels (Life Technologies), followed by electrotransfer of proteins onto PVDF transfer membranes (Perkin Elmer Life Sciences). Membranes were blocked in 5% BS A/TBS + 0.05% Tween and incubated overnight at 4°C with primary antibodies against a-SMA primary antibody, anti-N-cadherin antibody (Sigma), connexin and vimentin (Abeam).
  • Cells were cultured in monolayer with DMEM, 10% FBS and 1% penicillin- streptomycin for 24h then fixed for 10 min with 3% paraformaldehyde in PBS and permeabilized with 0.2% Triton X-100 (Sigma). Cells were stained with either a rabbit monoclonal anti- ⁇ catenin or a rabbit polyclonal anti-connexin (Abeam) followed by Alexa Fluor 568-conjugated goat anti-rabbit antibody (Invitrogen), Alexa Fluor 488 Phalloidin (Invitrogen) and DNA with DAPI (Sigma). Secondary antibody alone was used as an immuno labelling control.
  • the rate of FPCL contraction was calculated by measuring the average gradient of the curve between 6 and 24 h. Analysis of single variance was used for comparing all conditions. All statistical analyses were performed using software (GraphPad Software version 5.0c). Significance was achieved if p ⁇ 0.05.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Epidemiology (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Physical Education & Sports Medicine (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Organic Chemistry (AREA)
  • Inorganic Chemistry (AREA)
  • Neurology (AREA)
  • Orthopedic Medicine & Surgery (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Immunology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

La présente invention concerne des compositions comprenant des agents bloquants des canaux ioniques mécano-sensibles, tels que le gadolinium, ou des antagonistes de la N-cadhérine ou des inhibiteurs de la connexine 43, qui permettent de ralentir, d'interrompre ou d'inverser la progression de troubles fibroprolifératifs musculo-squelettiques, tels que la maladie de Dupuytren et autres fibromatoses ou maladies analogues, caractérisés par la contracture.
PCT/EP2012/071642 2011-11-04 2012-11-01 Traitement de troubles fibroprolifératifs musculo-squelettiques WO2013064585A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
GBGB1119089.9A GB201119089D0 (en) 2011-11-04 2011-11-04 Treatment of musculoskeletal fibroproliferative disorders
GB1119089.9 2011-11-04

Publications (1)

Publication Number Publication Date
WO2013064585A1 true WO2013064585A1 (fr) 2013-05-10

Family

ID=45421293

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2012/071642 WO2013064585A1 (fr) 2011-11-04 2012-11-01 Traitement de troubles fibroprolifératifs musculo-squelettiques

Country Status (2)

Country Link
GB (1) GB201119089D0 (fr)
WO (1) WO2013064585A1 (fr)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2013144758A1 (fr) * 2012-03-27 2013-10-03 Novartis Ag Traitement de la fibrose
EP3019017A4 (fr) * 2013-07-11 2017-09-27 180 Therapeutics LP Méthode de traitement de troubles fibro-prolifératifs comprenant la maladie de dupuytren par une ou plusieurs métalloprotéinases matricielles et un antagoniste de tnf
EP3634476A4 (fr) * 2017-06-06 2021-06-02 The Regents of The University of California Anticorps anti-n-cadhérine humanisés et leurs utilisations

Citations (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1991001624A2 (fr) * 1989-07-31 1991-02-21 Massachusetts Institute Of Technology Utilisation d'antagonistes du calcium pour le traitement de cicatrices
US5589171A (en) 1994-08-22 1996-12-31 Advance Biofactures Of Curacao Treatment of Dupuytren's disease with collagenase
GB2323530A (en) 1997-03-27 1998-09-30 Advance Biofactures Curacao Collagenase and Dupuytren's Disease
US6031005A (en) * 1998-08-03 2000-02-29 Easterling; W. Jerry Composition and method for treating Peyronie's disease and related connective tissue disorders
US6086272A (en) 1996-03-26 2000-07-11 Seiko Epson Corporation Printing apparatus and control method therefor
US20040161761A1 (en) 2001-03-02 2004-08-19 Ferguson Mark William James Genetic testing
WO2004082705A1 (fr) 2003-03-18 2004-09-30 Delta Ltd Traitement de la contracture de dupuytren
WO2009085270A2 (fr) * 2007-12-21 2009-07-09 Coda Therapeutics, Inc. Traitement d'états fibreux
WO2010054377A2 (fr) 2008-11-10 2010-05-14 The Regents Of The University Of California Anticorps entièrement humains dirigés contre la n-cadhérine
WO2010102262A1 (fr) 2009-03-06 2010-09-10 Halozyme, Inc. Mutants thermosensibles de métalloprotéase matricielle 1 et leurs utilisations
WO2010102202A2 (fr) 2009-03-06 2010-09-10 Kenneth William Gregg Composition pour commander le comportement de poissons
WO2011119888A2 (fr) 2010-03-24 2011-09-29 The Regents Of The University Of California N-cadhérine: une cible pour diagnostic et traitement du cancer

Patent Citations (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1991001624A2 (fr) * 1989-07-31 1991-02-21 Massachusetts Institute Of Technology Utilisation d'antagonistes du calcium pour le traitement de cicatrices
US5589171A (en) 1994-08-22 1996-12-31 Advance Biofactures Of Curacao Treatment of Dupuytren's disease with collagenase
US6086272A (en) 1996-03-26 2000-07-11 Seiko Epson Corporation Printing apparatus and control method therefor
USRE39941E1 (en) 1997-03-27 2007-12-18 Advance Biofactures Corporation Amelioration of Dupuytren's disease
GB2323530A (en) 1997-03-27 1998-09-30 Advance Biofactures Curacao Collagenase and Dupuytren's Disease
US6086872A (en) 1997-03-27 2000-07-11 Advance Biofactures Of Curacao, Nv Amelioration of dupuytren's disease
US6031005A (en) * 1998-08-03 2000-02-29 Easterling; W. Jerry Composition and method for treating Peyronie's disease and related connective tissue disorders
US20040161761A1 (en) 2001-03-02 2004-08-19 Ferguson Mark William James Genetic testing
WO2004082705A1 (fr) 2003-03-18 2004-09-30 Delta Ltd Traitement de la contracture de dupuytren
WO2009085270A2 (fr) * 2007-12-21 2009-07-09 Coda Therapeutics, Inc. Traitement d'états fibreux
WO2010054377A2 (fr) 2008-11-10 2010-05-14 The Regents Of The University Of California Anticorps entièrement humains dirigés contre la n-cadhérine
WO2010102262A1 (fr) 2009-03-06 2010-09-10 Halozyme, Inc. Mutants thermosensibles de métalloprotéase matricielle 1 et leurs utilisations
WO2010102202A2 (fr) 2009-03-06 2010-09-10 Kenneth William Gregg Composition pour commander le comportement de poissons
WO2011119888A2 (fr) 2010-03-24 2011-09-29 The Regents Of The University Of California N-cadhérine: une cible pour diagnostic et traitement du cancer

Non-Patent Citations (12)

* Cited by examiner, † Cited by third party
Title
BADALAMENTE ET AL., J HAND SURG AM, vol. 32, 2007, pages 767 - 774
HINZ ET AL., MOL BIOL CELL, vol. 15, 2004, pages 4310 - 20
HINZ ET AL., MOL. BIOL. CELL, vol. 15, 2004, pages 4310 - 20
HURST ET AL., N. ENGL J. MED, vol. 361, 2009, pages 968 - 979
L. FOLLONIER ET AL: "Myofibroblast communication is controlled by intercellular mechanical coupling", JOURNAL OF CELL SCIENCE, vol. 121, no. 20, 15 October 2008 (2008-10-15), pages 3305 - 3316, XP055049240, ISSN: 0021-9533, DOI: 10.1242/jcs.024521 *
REHMAN ET AL., ARTHRITIS RESEARCH & THERAPY, vol. 13, 2011, pages 238
ROMBOUTS, J HAND SURG AM, vol. 14, 1989, pages 644 - 652
TOWNLEY ET AL., J HAND SURG EUR, vol. 34, 2009, pages 783 - 7
VEJEE ET AL., J CELL PHYSIOL, vol. 224, 2010, pages 681 - 90
VERJEE ET AL., J CELL PHYSIOL., vol. 224, 2010, pages 681 - 690
VERJEE LS; MIDWOOD K; DAVIDSON D; EASTWOOD M; NANCHAHAL J: "Post-transcriptional regulation of alpha-smooth muscle actin determines the contractile phenotype of Dupuytren's nodular cells", J CELL PHYSIOL., vol. 224, 2010, pages 681 - 690
VERJEE LS; MIDWOOD K; DAVIDSON D; ESSEX D; SANDISON A; NANCHAHAL J: "Myofibroblast distribution in Dupuytren's cords: correlation with digital contracture", J HAND SURG AM, vol. 34, 2009, pages 1785 - 1794

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2013144758A1 (fr) * 2012-03-27 2013-10-03 Novartis Ag Traitement de la fibrose
EP3019017A4 (fr) * 2013-07-11 2017-09-27 180 Therapeutics LP Méthode de traitement de troubles fibro-prolifératifs comprenant la maladie de dupuytren par une ou plusieurs métalloprotéinases matricielles et un antagoniste de tnf
EP3634476A4 (fr) * 2017-06-06 2021-06-02 The Regents of The University of California Anticorps anti-n-cadhérine humanisés et leurs utilisations

Also Published As

Publication number Publication date
GB201119089D0 (en) 2011-12-21

Similar Documents

Publication Publication Date Title
US10669334B2 (en) Treatment for Dupuytren's disease
He et al. Irisin inhibits osteocyte apoptosis by activating the Erk signaling pathway in vitro and attenuates ALCT-induced osteoarthritis in mice
Olaso et al. Impaired dermal wound healing in discoidin domain receptor 2-deficient mice associated with defective extracellular matrix remodeling
JP4668902B2 (ja) 創傷治癒を促進するための薬剤組成物及び方法
Qian et al. Downregulating PI3K/Akt/NF-κB signaling with allicin for ameliorating the progression of osteoarthritis: in vitro and vivo studies
Krieger et al. TSH/IGF1 receptor crosstalk: mechanism and clinical implications
Huang et al. Pharmacological treatment for keloids
US11389512B2 (en) Use of low molecular weight fractions of human serum albumin in treating diseases
Quang et al. Peripheral endothelin B receptor agonist-induced antinociception involves endogenous opioids in mice
JP2016518357A (ja) 骨格筋幹細胞を若返らせる方法および組成物
Xiang et al. Ivabradine abrogates TNF-α-induced degradation of articular cartilage matrix
EP3019017A2 (fr) Méthode de traitement de troubles fibro-prolifératifs comprenant la maladie de dupuytren par une ou plusieurs métalloprotéinases matricielles et un antagoniste de tnf
Zhou et al. Tigogenin inhibits adipocytic differentiation and induces osteoblastic differentiation in mouse bone marrow stromal cells
WO2013064585A1 (fr) Traitement de troubles fibroprolifératifs musculo-squelettiques
Ma et al. Time course of disassociation of bone formation signals with bone mass and bone strength in sclerostin antibody treated ovariectomized rats
Wu et al. The protective effect of sophocarpine in osteoarthritis: an in vitro and in vivo study
Rejnmark et al. New and emerging antiresorptive treatments in osteoporosis
EP3302530B1 (fr) Agents thérapeutiques à base de cdh26 et leur utilisation
KR102026138B1 (ko) Crif1 억제제를 함유하는 켈로이드 및 비후성 반흔 형성 억제 또는 치료용 조성물
Sah et al. Novel isonahocol E3 exhibits anti-inflammatory and anti-angiogenic effects in endothelin-1-stimulated human keratinocytes
WO2009131850A2 (fr) Inhibiteurs de l’expression et de l’activité de pai-1 pour le traitement des troubles oculaires
AU2012308097A1 (en) Treatment of bone diseases
KR20190000841A (ko) Gas6 단백질 또는 이의 수용체 활성화제를 포함하는 섬유증의 예방 또는 치료용 조성물
Yu et al. Irisin mitigates rheumatoid arthritis by suppressing mitochondrial fission via inhibiting YAP-Drp1 signaling pathway
KR20220041847A (ko) 섬유성 병리를 치료하기 위한 화합물 및 방법

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 12783195

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 12783195

Country of ref document: EP

Kind code of ref document: A1