EP3227334A1 - Anticorps anti-trka à propriétés inhibitrices améliorées et dérivés desdits anticorps destinés à être utilisés pour traiter les douleurs osseuses - Google Patents

Anticorps anti-trka à propriétés inhibitrices améliorées et dérivés desdits anticorps destinés à être utilisés pour traiter les douleurs osseuses

Info

Publication number
EP3227334A1
EP3227334A1 EP15807645.5A EP15807645A EP3227334A1 EP 3227334 A1 EP3227334 A1 EP 3227334A1 EP 15807645 A EP15807645 A EP 15807645A EP 3227334 A1 EP3227334 A1 EP 3227334A1
Authority
EP
European Patent Office
Prior art keywords
antibody
trka
antibodies
pain
fragment
Prior art date
Legal status (The legal status 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 status listed.)
Withdrawn
Application number
EP15807645.5A
Other languages
German (de)
English (en)
Inventor
Adrian Walmsley
Sachin Kumar DUBEY
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Ichnos Sciences SA
Original Assignee
Glenmark Pharmaceuticals SA
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
Priority claimed from US14/562,297 external-priority patent/US20150183885A1/en
Application filed by Glenmark Pharmaceuticals SA filed Critical Glenmark Pharmaceuticals SA
Publication of EP3227334A1 publication Critical patent/EP3227334A1/fr
Withdrawn legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2863Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against receptors for growth factors, growth regulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/08Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/04Antineoplastic agents specific for metastasis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/24Immunoglobulins specific features characterized by taxonomic origin containing regions, domains or residues from different species, e.g. chimeric, humanized or veneered
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/52Constant or Fc region; Isotype
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/52Constant or Fc region; Isotype
    • C07K2317/524CH2 domain
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/55Fab or Fab'
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • C07K2317/567Framework region [FR]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/71Decreased effector function due to an Fc-modification
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/73Inducing cell death, e.g. apoptosis, necrosis or inhibition of cell proliferation
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/76Antagonist effect on antigen, e.g. neutralization or inhibition of binding
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/90Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
    • C07K2317/92Affinity (KD), association rate (Ka), dissociation rate (Kd) or EC50 value
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/90Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
    • C07K2317/94Stability, e.g. half-life, pH, temperature or enzyme-resistance

Definitions

  • the present invention relates generally to antibodies directed against TrkA receptor and their uses, including humanized anti-TrkA antibodies and methods of treatment with anti- TrkA antibodies.
  • the present invention relates to humanized anti-TrkA antibodies with enhanced inhibitory properties for use in methods of treating neuroma and/or bone associated pain.
  • the present invention relates to humanized anti-TrkA antibodies with enhanced inhibitory properties comprising a heavy chain variable region, a light chain variable region, a human light chain constant region and a variant human IgG4 heavy chain constant region which exhibit altered exchange properties.
  • Neurotrophins are a family of peptide growth factors (Barde YA (1994) J. Neurobiol. 25(11): 1329-33) structurally related to the first member of the family NGF (Levi-Montalcini R (1987) EMBO J. 6(5): 1145-54). Neurotrophins modulate neuronal differentiation and survival, as well as synaptic transmission, both of the peripheral neurons and of the central nervous system. Furthermore NGF acts on various non neuronal tissues and cells, such as immune cells.
  • TrkA acts through two membrane receptors present in the target cells, the low affinity p75 receptor, and the 140 kDa high affinity transmembrane glycoprotein, TrkA (Kaplan DR et al, (1991) Science 252(5005):554-8; Klein R et al, (1991) Cell 65(1): 189-97) having tyrosine kinase activity. TrkA is expressed in neural-crest neurons, in sympathetic neurons as well as in cholinergic neurons of the basal fore-brain and corpus striatum, where it represents the crucial mediator of NGF activities (Holtzman DM et al, (1992) Neuron 9(3):465-78; Verge VM et al, (1992) J. Neurosci. 12(10):4011-22). TrkA is also expressed in some non neuronal tissues and cells, including B lymphocytes (Torcia M et al, (1996) Cell 85(3):345-56).
  • Nerve growth factor was identified originally as a survival factor for sensory and sympathetic neurons in the developing nervous system (Gorin PD & Johnson EM (1979) PNAS USA, 76(10):5382-6). In adults, NGF is not required for survival but it has a crucial role in the generation of pain and hyperalgesia in several acute and chronic pain states. The expression of NGF is high in injured and inflamed tissues, and activation of trkA on nociceptive neurons triggers and potentiates pain signalling by multiple mechanisms.
  • Inflammation-related pain can be significantly reduced by neutralizing NGF bioactivity in animal models (Woolf CJ et al, (1994) Neuroscience 62(2):327-31; McMahon SB et al, (1995) Nat. Med. l(8):774-80; Koltzenburg M et al, (1999) Eur. J. Neurosci. 11(5): 1698- 704), implying that an enhanced level of this neurotrophin is necessary to generate the full hyperalgesic response.
  • the inhibition of other neurotrophins does not result in antagonizing the induced hyperalgesia, suggesting that this effect is specific to NGF (McMahon SB et al, (1995) Nat. Med.
  • NGF inhibition results in analgesia in different neuropathy-related pain protocols (Koltzenburg M et al, (1999) Eur. J. Neurosci. 11(5): 1698-704; Ro LS et al, (1999) Pain 79(2-3):265-74; Theodosiou M et al, (1999) Pain, 81(3):245-55; Christensen MD & Hulsebosch CE (1997) Exp. Neurol. 147(2):463-75).
  • TrkA instead of NGF may represent a better therapeutic choice as this receptor does not interfere with NGF functions mediated by the p75 receptor, the latter having broad function in neuronal development.
  • the inventors have evaluated the efficacy and safety of using humanized anti-TrkA antibodies in methods of treating deep somatic pain and in particular bone associated pain, such as pain resulting from damage to bones from an external trauma or as a consequence of bone affecting pathology such as cancer which may lead to bone damage, inappropriate inervation and/or other effects leading to bone associated pain.
  • the inventors have also evaluated the efficacy and safety of using humanized anti-TrkA antibodies in methods of treating pain resulting from neuromas that have formed inappropriately as a consequence of a trauma or another pathology.
  • the present disclosure relates generally to humanized anti-TrkA antibodies, methods for their preparation and use.
  • the present disclosure provides a humanized anti-TrkA antibody or fragment thereof comprising:
  • a heavy chain variable domain comprising a sequence selected from the group consisting of SEQ ID NOs: 1-5, and
  • a light chain variable domain comprising a sequence selected from the group consisting of SEQ ID NOs: 6-13,
  • CDR2 of the heavy chain variable domain comprises at least one amino acid substitution and/or wherein the non-CDR region of the heavy chain variable domain comprises an amino acid substitution at an amino acid position selected from the group consisting of 37, 42 and 89, wherein the amino acid position of each group member is indicated utilizing the numbering system set forth in Kabat.
  • the present disclosure provides an isolated nucleic acid encoding a humanized anti-TrkA antibody or fragment thereof, a vector comprising the isolated nucleic acid and a host cell comprising the isolated nucleic acid or the vector. Also provided by the present disclosure is a method of producing a humanized anti-TrkA antibody or fragment thereof.
  • the present disclosure provides a composition comprising a humanized anti-TrkA antibody or fragment thereof and an immunoconjugate comprising a humanized anti-TrkA antibody or fragment thereof linked to a therapeutic agent.
  • the present invention relates to a humanized anti-TrkA antibody or fragment thereof that bind to human TrkA, wherein said anti-TrkA antibody or fragment thereof comprising heavy chain variable domain CDRs 1 , 2 and 3 and light chain variable domain CDRs 1, 2 and 3, and the heavy chain variable domain comprises a sequence selected from the group consisting of SEQ ID NOs: 1-5, and the light chain variable domain comprises a sequence selected from the group consisting of SEQ ID NOs: 6-13, wherein the non-CDR region of the heavy chain variable domain comprise an amino acid substitution at an amino acid position selected from the group consisting of 37, 42, and 89, wherein the amino acid position of each group member is indicated utilizing the numbering system set forth in Kabat for treating bone associated pain.
  • the present disclosure provides a humanized anti-TrkA antibody or fragment thereof, a composition or an immunoconjugate for use in medicine, for use in the treatment of pain, for use in the treatment of chronic pain, for use in the treatment of acute pain, for use in the treatment of pain associated with one or more of the following: pancreatitis, kidney stones, endometriosis, IBD, Crohn's disease, post surgical adhesions, gall bladder stones, headaches, dysmenorrhea, musculoskeletal pain, sprains, visceral pain, ovarian cysts, prostatitis, cystitis, interstitial cystitis, post-operative pain, migraine, trigeminal neuralgia, pain from burns and/or wounds, pain associated with trauma, neuropathic pain, pain associated with musculoskeletal diseases, rheumatoid arthritis, osteoarthritis, ankylosing spondilitis, periarticular pathologies, oncological pain, pain from bone metasta
  • the present disclosure relates to methods for treating deep somatic pain and in particular bone associated pain comprising at least the step of administering an effective amount of a humanized anti-TrkA antibody or fragment thereof according to the present invention to an individual in need.
  • bone associated pain is meant to refer to any sensation of pain experienced as a consequence of an injury, deterioration or any pathology associated with one or more bones in an individual.
  • the pain may occur as a feeling of numbness, a tingling sensation, a crawling sensation, a sensation of heat or cold, or a feeling of tightness, a dull pain such as be a feeling of pressure or tightness, a heavy feeling, or a tingling sensation, a sharp pain such as a stabbing, shooting, tearing, or piercing pain, the pain may be constant, throbbing or intermittant at regular or irregular intervals of time or occur only when the area is touched.
  • the bone associated pain may be a consequence of a bone fracture, chip, break or any other injury or inflammation. This may be due to an accident or as a consequence of an overuse or repetitive movement injury or result from a weakened bone due to a hormone deficiency, infection, bone cancer, a metastatic malignancy, leukaemia, myeloma or another cancer affecting blood or bone associated cells, or due to an interruption in the blood supply to the bone.
  • the bone associated pain may be a consequence of a bone affecting pathology.
  • a bone affecting pathology refers to any pathology which directly or indirectly affects one or more bones in an individual, leading to a sensation of pain. Examples of pathologies include cancer, infection and immune system dysfunction.
  • the bone associated pain may be associated with or result from bone cancer and in particular secondary bone cancer and/or metastatic cancer cells.
  • the bone associated pain may be a consequence of one or more neuroma, in particular resulting from bone cancer or any other pathology.
  • the method involves the administration of an effective amount of a humanized anti-TrkA antibody or fragment thereof according to the present invention, so as to minimise or reduce abberant nerve sprouting and/or neuroma formation or size.
  • an effective amount of the humanized anti-TrkA antibody or fragment thereof is intended to mean an amount sufficient to lead to the alleviation or cessation of one or more pain sensations in an individual in need thereof, when administered to the individual by a clinician or other qualified medical professional.
  • the present disclosure relates to a humanized anti-TrkA antibody or fragment thereof according to the present invention for use in the treatment of deep somatic pain and in particular bone associated pain or neuroma related pain.
  • the bone associated pain may be a consequence of a bone fracture, chip, break or any other injury or inflammation. This may be due to an accident or as a consequence of an overuse or repetitive movement injury or result from a weakened bone due to a hormone deficiency, infection, bone cancer, a metastatic malignancy, leukaemia, myeloma or another cancer affecting blood or bone associated cells, or due to an interruption in the blood supply to the bone.
  • the bone associated pain may be a consequence of a bone affecting pathology.
  • a bone affecting pathology refers to any pathology which directly or indirectly affects one or more bones in an individual, leading to a sensation of pain.
  • pathologies include cancer, infection and immune system dysfunction.
  • bone associated pain may be associated with or result from bone cancer and in particular secondary bone cancer and/or metastatic cancer cells.
  • the bone associated pain may be a consequence of one or more neuroma, in particular resulting from bone cancer or any other pathology.
  • the invention relates to a humanized anti-TrkA antibody or fragment thereof according to the present invention for use in the treatment to minimise or reduce aberrant nerve sprouting and/or neuroma formation or size.
  • the present disclosure provides methods for treating inflammatory pain, osteo arthritic pain and neuropathic pain.
  • administration of a humanized anti-TrkA antibody resulted in a significant reversal of acute inflammatory hyperalgesia at a dose of O.Olmg/kg.
  • administration of a humanized anti-TrkA antibody resulted in a significant and sustained reversal of chronic inflammatory hyperalgesia at a dose of O.Olmg/kg.
  • administering resultsed in a significant and sustained reversal of chronic osteoarthritic hyperalgesia at a dose of O.Olmg/kg.
  • administration of a humanized anti-TrkA antibody resulted in a significant reversal of neuropathic pain at a dose of 1.Omg/kg.
  • the present disclosure provides an article of manufacture comprising a humanized anti-TrkA antibody or fragment thereof, a composition or an immunoconjugate. In a further aspect, the present disclosure provides a kit comprising a humanized anti-TrkA antibody or fragment thereof, a composition or an immunoconjugate.
  • Particular problems also relate to the generation of stable and efficatious pharmaceutical formulations comprising antibodies result from the fact that proteins are larger and more complex than traditional organic and inorganic drugs.
  • a formulation must preserve intact the conformational integrity of at least a core sequence of the protein's amino acids while at the same time protecting the protein's multple functional groups from degradation.
  • Degradation pathways for proteins can involve chemical instability (i.e. any process which involves modification of the protein by bond formation or cleavage resulting in a new chemical entity) or physical instability (i.e. changes in the higher order structure of the protein).
  • Chemical instability can result from deamidation, racemization, hydrolysis, oxidation, beta elimination or disulfide exchange.
  • Physical instability can result from denaturation, aggregation, precipitation or adsorption
  • a stable aqueous pharmaceutical formulation comprising a therapeutically effective amount of an anti-TrkA antibody, a buffer, a surfactant and a tonicifying agent, wherein the pH of the formulation is from about 5 to about 7.
  • the anti-TrkA antibody comprises: a) a heavy chain variable domain comprising a sequence selected from the group consisting of
  • a light chain variable domain comprising a sequence selected from the group consisting of SEQ ID NOs: 6-13,
  • CDR2 of the heavy chain variable domain comprises at least one amino acid substitution and/or wherein the non-CDR region of the heavy chain variable domain comprises an amino acid substitution at an amino acid position selected from the group consisting of 37, 42 and 89, wherein the amino acid position of each group member is indicated utilizing the numbering system set forth in Kabat (Kabat EA et al., (1991) Sequences of proteins of immunological interest. 5th Edition - US Department of Health and Human Services, NIH publication n° 91-3242).
  • the anti-TrkA antibody is present at a concentration of at least 0.1 mg/ml.
  • the anti-TrkA antibody is present at a concentration of at least 0.2 mg/ml to 175 mg/ml.
  • the anti-TrkA antibody is present at a concentration of at least 1 mg/ml.
  • the anti-TrkA antibody is present at a concentration of at least 10 mg/ml.
  • the anti-TrkA antibody is present at a concentration of at least 100 mg/ml.
  • the anti-TrkA antibody is present at a concentration of at least 150 mg/ml.
  • the buffer is selected from the group comprising: citrate, acetate, histidine, phosphate, tris (tris(hydroxymethyl)aminomethane).
  • the stabilising/tonicifying agents is selected from the group comprising: sodium acetate, sodium bicarbonate, sodium carbonate, sodium chloride, potassium acetate, potassium bicarbonate, potassium carbonate, potassium chloride, sucrose, polyols, sugars, amino acids such as histidine, arginine, and glycine, methionine, proline, lysine, glutamic acid, amines and trehalose.
  • the surfactant is selected from the group comprising: tween 20/40/80, polysorbate 20/80, poloxamer, sodium lauryl sulphate.
  • the pH of the formulation is 5.75.
  • the pH of the formulation is 6.0.
  • the formulation may further comprise one or more excipients and bulking agents.
  • a low concentration aqueous formulation of an anti-TrkA antibody adjusted to pH 6.0 comprising: lOmg/ml of an anti-TrkA antibody;
  • a low concentration aqueous formulation of an anti-TrkA antibody adjusted to pH 6.0 comprising: at least 1 Omg/ml of an anti-TrkA antibody;
  • a high concentration aqueous formulation of an anti-TrkA antibody adjusted to pH 5.75 or 6.0 comprising:
  • a high concentration aqueous formulation of an anti-TrkA antibody adjusted to pH 5.75 or 6.0 comprising:
  • the formulation is administered to a person in need at a dose of at least O. lmg per kg of body weight; in particular at least 0.3mg/kg, lmg/kg, 3mg/kg, lOmg/kg or 30mg/kg.
  • the formulation is administered to a patient in need once.
  • the formulation is administered to a patient in need at an interval of at least 1 hour between dosing and preferably 4 hours/6 hours/12 hours/24 hours between dosing.
  • the formulation is administered to a patient in need on demand.
  • the formulation is stable at 5°C for at least 1 year and preferably at least 2 years.
  • the formulation is stable at 25°C for at least 3 months, preferably 6 months and most preferably 1 year.
  • the formulation is stable at 40°C for at least 1 month and preferably 3 months.
  • stability is measured with one or more method selected from the group comprising: determining changes in the clarity, degree of coloration, degree of opalescence and particulate contamination (visible particles) of the formulation, light absorption measurement of wavelength 280nm to determine the concentration of protein present in the formulation; by SDS-page gel visualisation to determine changes in the weight and or breakdown of the antibody; by ELISA to determine any change in the binding properties of the antibody; by HPLC-CEX to determine changes in the positive/negative antibody species make up in the formulation; by HPLC-IEF so as to determine changes in the IsoElectroFocusing profile of the antibody by capillary electrophoresis present in the formulation, by HPLC-SEC analysis to determine changes in the antibody in the formulation.
  • the formulation comprises an amount of an anti-TrkA antibody so that the formulation can be administered to a person in need at a dose of at least O. lmg per kg of body weight; in particular at least 0.3mg/kg, lmg/kg, 3mg/kg, lOmg/kg or 30mg/kg.
  • FIG. 1 Surface Plasmon resonance measurements of anti-TrkA antibodies. Data are expressed as number of response (abbreviated RU; Y axis) vs. time (X axis).
  • FIG. 1A MNAC13 antibody.
  • FIG. IB BXhVH5VLl antibody.
  • FIG. 1C GBR VH5(V37A)VL1 antibody.
  • FIG. ID GBR VH5(K3Q,V37A)VL1 antibody.
  • FIG. 2 Thermostability measurements of anti-TrkA antibodies using differential scanning calorimetry. Data are expressed as excess molar heat capacity (abbreviated Cp [kcal/mol/°C]; Y axis) vs. temperature (X axis).
  • Cp excess molar heat capacity
  • FIG. 2A MNAC13 antibody
  • FIG. 2B BXhVH5VLl antibody
  • FIG. 2C GBR VH5(V37A)VL1 antibody (FAB fragment Tm is Tml at 73.6°C).
  • FIG. 2D GBR VH5(K3Q,V37A)VL1 antibody (FAB fragment Tm is Tml at 73°C).
  • FIG. 2E overlay of FIG. 2C and FIG. 2D.
  • FIG. 2F overlay of FIG. 2B and FIG. 2D.
  • FIG. 2G overlay of FIG. 2A and FIG. 2D.
  • FIG. 3 Functional bioactivity of anti-TrkA antibodies. Effect of humanized anti-TrkA antibodies on the NGF-induced TF-1 cell proliferation; data are expressed as % of proliferative response (Y axis) vs. antibody concentration ⁇ g/ml; X axis).
  • FIG. 3A GBR VH5(V37A)VL1, GBR VH5(K3Q,V37A)VL1 vs. BXhVH5VLl .
  • FIG. 3B GBR VH5(V37A)VL1, GBR VH5(K3Q,V37A)VL1 vs. MNAC13.
  • FIG. 3C GBR VH5(K3Q,V37A)VL1 vs. GBR VH5(K3Q,V37A)VL1 IGHG4 S228P.
  • FIG. 3D BXhVH5VLl, BXhVH5VL3, GBR VH5(V37A)VL1 vs. GBR VH5(V37A)VL3.
  • FIG. 3E BXhVH5VLl, BXhVFBVLl, GBR VH5(V37A)VL1 vs. GBR VH3(V37A)VL1.
  • FIG. 4 Humanized anti-TrkA antibody reverses acute inflammatory paw pain.
  • Acute inflammatory hyperalgesia of the paw was induced by intraplantar injection of CFA into one of the hind limb paws of AMBl mice and measured as the % ratio between weight bearing on the ipsilateral (injected) and contralateral (non- injected) paw (% ipsi/contra, mean ⁇ s.e.m.). Weight bearing readings were taken 23hrs post-CFA injection (0 hr) before treatment initiation at 24hr post-CFA with a single i.p.
  • mice were treated with lOmg/kg indomethacin p.o. (vertically- hatched bars).
  • FIG. 5 Humanized anti-TrkA antibody reverses chronic inflammatory joint pain.
  • Chronic inflammatory hyperalgesia of the joint was induced by intra-articular injection of CFA into one of the hind limb knee joints of AMBl mice and measured as the % ratio between weight bearing on the ipsilateral (injected) and contralateral (non- injected) limb (% ipsi/contra, mean ⁇ s.e.m.). Weight bearing readings were taken immediately prior to CFA injection (naive) and on day 3, 7 and 10 post-CFA before treatment initiation on day 13 post-CFA with a single i.p.
  • mice were treated with 60mg/kg celecoxib twice daily (open diamond) from day 13 onwards and weight bearing was measured at 1 and 8 hrs post-dosing on day 13 post-CFA and then at 1 hr post-dosing on days 14-17 post-CFA.
  • FIG. 6 Humanized anti-TrkA antibody reverses chronic osteoarthritic pain.
  • Chronic osteo arthritic hyperalgesia was induced by intra-articular injection of MIA into one of the hind limb knee joints of AMBl mice and measured as the % ratio between weight bearing on the ipsilateral (injected) and contralateral (non- injected) limb (% ipsi/contra, mean ⁇ s.e.m.).
  • Baseline (BL) weight bearing readings were taken immediately prior to MIA injection and on day 3, 7 and 10 days post-MIA before treatment initiation on day 14 post-MIA with a single i.p.
  • FIG. 7 Humanized anti-TrkA antibody reverses neuropathic pain.
  • Chronic neuropathic hyperalgesia and allodynia were induced by chronic constriction injury of the sciatic nerve of AMBl mice and measured as the threshold force required for paw withdrawal (g) and the latency time for paw withdrawal from a cold plate (sec), respectively. Readings were taken prior to surgery and 7 days after surgery on the day before treatment initiation (Oh). Animals were then treated with a single i.p. injection of either 1000 ⁇ g/kg isotype control antibody (closed triangle) or 10 (closed circle), 100 (closed square) and 1000 ⁇ g/kg (closed diamond) anti-TrkA antibody.
  • Pregabalin (closed inverted triangle) at 30 mg/kg/lOml, p.o. or saline (open circle) were administered once daily over the seven day post-dose period. Post-dose readouts were recorded at 4h, 24h and then every other day until the 7th day post-dose. Post- dose readouts were recorded lh after pregabalin or saline dosing on all days.
  • FIG. 1 Pregabalin (closed inverted triangle) at 30 mg/kg/lOml, p.o. or saline (open circle) were administered once daily over the seven day post-dose period. Post-dose readouts were recorded at 4h, 24h and then every other day until the 7th day post-dose. Post- dose readouts were recorded lh after pregabalin or saline dosing on all days.
  • GBR VH5 K3Q,V37A
  • VL1 IGHG4 S228P label GBR
  • tanezumab squares
  • PBS circles.
  • Statistical analysis was performed as a one-way ANOVA with a Dunnett's post hoc test comparison to PBS treatment: *p ⁇ 0.05, **p ⁇ 0.01, ***p ⁇ 0.001.
  • FIG. 10 Frequency plot of the diameters of all SCG neuronal cell bodies from adult AMB1 mice treated with GBR VH5 (K3Q,V37A) VL1 IGHG4 S228P (label GBR) (1904 neurons, circles), PBS (1547 neurons, squares) or tanezumab (1420 neurons, diamonds). Binning was performed with Graphpad Prism using standard parameters.
  • FIG. 11 Representative H&E-stained sections of the SCG from PBS-treated adult animals, the smallest of the SCG of tanezumab-treated animals and the largest of the SCG of GBR VH5 (K3Q,V37A) VL1 IGHG4 S228P (label GBR) - treated animals.
  • the neuronal cell body is marked with a black arrow.
  • FIG. 12 Neuropathic pain was induced by chronic constriction injury of the sciatic nerve of AMB1 mice.
  • Mechanical hyperalgesia A was measured as the threshold force required for paw withdrawal (g) and cold allodynia (B) was measured as the latency time for paw withdrawal from a cold plate (sec). Readings were taken prior to surgery (-7d) and 7 days after surgery on the day before treatment initiation (Oh). Animals were then treated with a single i.p.
  • FIG. 13 Chronic inflammatory joint pain was induced by multiple intra-articular injections of CFA into the knee joint of AMB1 mice.
  • DO the day of first CFA injection; D3, D10, D17, D24: 3, 10, 17 and 24 days after the first CFA injection.
  • Arrows indicate intra-articular injections (PBS or CFA) on day 0, 7, 14 and 21.
  • 4h, 8h, 24h, 48h, 76h, 96h and 120h 4, 8, 24, 48, 76, 96 and 120 hours after dosing on D24.
  • * P ⁇ 0.05, compared to CFA + Vehicle group, one-way ANOVA.
  • FIG. 15 Representative histological sections of fractured femurs stained with safranin-O/fast green and assessment of the callus composition.
  • A-C PBS -treatment
  • D-F anti-NGF antibody
  • G-I anti-TrkA antibody.
  • Scale bar 500 ⁇ .
  • FIG 17 Stability data for low concentration (lOmg/ml) formulation at 5°C.
  • FIG 18 Stability data for low concentration (lOmg/ml) formulation at 25°C.
  • FIG 19 Stability data for low concentration (lOmg/ml) formulation at 40°C.
  • FIG 20 Stability data for high concentration (lOOmg/ml) formulation A (pH 5.75) at 5°C.
  • FIG 21 Stability data for high concentration (lOOmg/ml) formulation A (pH 5.75) at 25°C.
  • FIG 22 Stability data for high concentration (lOOmg/ml) formulation A (pH 5.75) at 40°C.
  • FIG 23 Stability data for high concentration (lOOmg/ml) formulation B (pH 6) at 5°C.
  • FIG 24 Stability data for high concentration (lOOmg/ml) formulation B (pH 6) at 25°C.
  • FIG 25 Stability data for high concentration (lOOmg/ml) formulation B (pH 6) at 40°C.
  • FIG 26 Stability data for high concentration (150mg/ml) formulation A (pH 5.75) at 5°C.
  • FIG 27 Stability data for high concentration (150mg/ml) formulation A (pH 5.75) at 25°C.
  • FIG 28 Stability data for high concentration (150mg/ml) formulation A (pH 5.75) at 40°C.
  • FIG 29 Stability data for high concentration (150mg/ml) formulation B (pH 6) at 5°C.
  • FIG 30 Stability data for high concentration (150mg/ml) formulation B (pH 6) at 25°C.
  • FIG 31 Stability data for high concentration (150mg/ml) formulation B (pH 6) at 40°C. Detailed description of the invention
  • the present disclosure relates to humanized anti-TrkA antibodies or fragment thereof, methods for their preparation and use.
  • TrkA human TrkA
  • TrkA receptor human TrkA receptor
  • human TrkA receptor human TrkA receptor
  • TrkA is also known as high affinity nerve growth factor receptor or neurotrophic tyrosine kinase receptor type lor TRKl -transforming tyrosine kinase protein or Tropomyosin-related kinase A or Tyrosine kinase receptor or Tyrosine kinase receptor A or Trk-A or gpl40trk or pl40-TrkA or MTC or TRK.
  • TrkA is a receptor tyrosine kinase involved in the development and the maturation of the central and peripheral nervous systems through regulation of proliferation, differentiation and survival of sympathetic and nervous neurons.
  • TrkA is the high affinity receptor for NGF which is its primary ligand; it can also bind and be activated by NTF3/neurotrophin-3.
  • isoform TrkA-I is found in most non-neuronal tissues (UniProt/Swiss-Prot accession number P04629-2), while isoform TrkA-II is primarily expressed in neuronal cells (UniProt/Swiss-Prot accession number P04629-1), and isoform TrkA-III is specifically expressed by pluripotent neural stem and neural crest progenitors (UniProt/Swiss-Prot accession number P04629-4).
  • TrkA-II A fourth isoform which differs from isoform TrkA-II at residues 1-71 and lacks residues 393 to 398 is known as isoform 3 (UniProt/Swiss-Prot accession number P04629-3).
  • TrkA-II isoform is the major known isoform of TrkA.
  • Isoform TrkA-I has enhanced responsiveness to NTF3 neurotrophin whereas isoform TrkA-III is constitutively active and does not bind NGF.
  • the TrkA isoform as used herein is the TrkA-II isoform with SEQ ID NO: 72 and an extracellular region thereof comprising the sequence of SEQ ID NO: 25.
  • anti-TrkA antibody or fragment thereof or “humanised anti-TrkA antibody or fragment thereof" as used herein includes antibodies or a fragment thereof that bind to human TrkA e.g. human TrkA in isolated form, specifically antibodies or fragment thereof that bind to the TrkA-II isoform (SEQ ID NO: 72), more specifically antibodies or fragment thereof that binds to a monovalent form of the human TrkA extracellular region (SEQ ID NO: 25) of the TrkA-II isoform, with an affinity (KD) of 500 nM or less, preferably 350 nM or less, more preferably 150 nM or less, even more preferably 100 nM or less, most preferred 50 nM or less, in particular 30 nM or less.
  • the humanized anti-TrkA antibody or fragment thereof is capable of inhibiting the functional activation of TrkA and/or is capable of blocking or reducing one or more biological activities that would otherwise be induced by the binding of NGF to TrkA.
  • an "anti-NGF antibody” refers to an antibody which is able to bind to NGF, preferably human NGF. Usually the anti-NGF antibody is capable of inhibiting the functional activation of TrkA and/or is capable of blocking or reducing one or more biological activities of TrkA.
  • the binding affinity of an anti-NGF antibody to NGF can be 500 nM or less, preferably 100 nM or less.
  • the anti-NGF antibody should exhibit any one or more of the following characteristics: (a) bind to NGF and inhibit NGF biological activity and/or downstream pathways mediated by NGF signaling function; (b) block or decrease NGF receptor activation (including TrkA receptor dimerization and/or autophosphorylation); (c) increase clearance of NGF; (d) inhibit (reduce) NGF synthesis, production or release.
  • Anti-NGF antibodies are known in the art, see, e.g., PCT Publication Nos. WO 01/78698, WO 01/64247, US Patent Nos. 5,844,092, 5,877,016 and 6,153,189; Hongo et al, (2000) Hybridoma, 19:215-227; GenBank Accession Nos. U39608, U39609, L17078 or L17077.
  • humanized anti-TrkA antibody or fragment thereof capable of inhibiting the functional activation of TrkA refers to humanized anti-TrkA antibodies that exhibit any one or more of the following characteristics: (a) bind to TrkA and inhibit TrkA biological activities and/or downstream pathways mediated by the binding of NGF or NTF3/neurotrophin-3 signaling function; (b) prevent, ameliorate, or treat any aspect of pain; (c) block or decrease TrkA activation, or dimerization and/or autophosphorylation; (d) increase TrkA clearance; (e) inhibit or reduce TrkA synthesis and/or cell surface expression.
  • humanized anti-TrkA antibody or fragment thereof capable of blocking or reducing one or more biological activities of TrkA refers to humanized anti-TrkA antibodies which directly or indirectly reduce, inhibit, neutralize, or abolish TrkA biological activities.
  • TrkA biological activities of TrkA
  • TrkA biological activities refers without limitation to any one or more of the following: the ability to bind NGF or other neurotrophins; the ability to homo-dimerize, or hetero-dimerize and/or autophosphorylate; the ability to activate an NGF induced signalling pathway; the ability to promote cell differentiation, proliferation, survival, growth, migration and other changes in cell physiology, including (in the case of neurons, including peripheral and central neuron) change in neuronal morphology, synaptogenesis, synaptic function, neurotransmitter and/or neuropeptide release and regeneration following damage; and the ability to mediate pain and cancer pain associated with bone metastasis.
  • IC50 as used herein describes the half maximal inhibitory concentration (IC50) which is a measure of the effectiveness of a compound in inhibiting biological function, e.g. inhibition of humanized anti-TrkA antibodies on the proliferation of NGF-induced TF-1 cells.
  • antibody as referred to herein includes full-length antibodies and any antigen binding fragment or single chains thereof. Antibodies and specifically naturally occurring antibodies are glycoproteins which exist as one or more copies of a Y-shaped unit, composed of four polypeptide chains. Each "Y" shape contains two identical copies of a heavy (H) chain, and two identical copies of a light (L) chain, named as such by their relative molecular weights.
  • a Fab fragment consists of the entire light chain and part of the heavy chain.
  • the heavy chain contains one variable domain (heavy chain variable domain or VH) and either three or four constant domains (CHI, CH2, CH3 and CH4, depending on the antibody class or isotype).
  • the region between the CHI and CH2 domains is called the hinge region and permits flexibility between the two Fab arms of the Y-shaped antibody molecule, allowing them to open and close to accommodate binding to two antigenic determinants separated by a fixed distance.
  • the heavy chains of IgA, IgD and IgG each have four domains, i.e. one variable domain (VH) and three constant domains (CHl-3).
  • IgE and IgM have one variable and four constant domains (CHI -4) on the heavy chain.
  • the constant regions of the antibodies may mediate the binding to host tissues or factors, including various cells of the immune system (e.g., effector cells) and the first component (C lq) of the complement system classical pathway.
  • Each light chain is usually linked to a heavy chain by one covalent disulfide bond.
  • Each light chain contains one variable domain (light chain variable domain or VL) and one light chain constant domain.
  • the light chain constant domain is a kappa light chain constant domain designated herein as IGKC or is a lambda light chain constant domain designated herein as IGLC.
  • IGKC is used herein equivalently to CK or CK and has the same meaning.
  • IGLC is used herein equivalently to or CL and has the same meaning.
  • the term "an IGLC domain” as used herein refers to all lambda light chain constant domains e.g.
  • VH and VL regions can be further subdivided into regions of hypervariability, termed complementarity determining regions (CDR), interspersed with regions that are more conserved, termed framework regions (FR or FW or "non-CDR regions").
  • CDR complementarity determining regions
  • FR framework regions
  • Each VH and VL is composed of three CDRs and four FRs, arranged from amino- terminus to carboxy-terminus in the following order: FR1, CDR1, FR2, CDR2, FR3, CDR3, FR4.
  • the variable regions of the heavy and light chains contain a binding domain that interacts with an antigen.
  • Antibodies are grouped into classes, also referred to as isotypes, as determined genetically by the constant region.
  • Human constant light chains are classified as kappa (CK) and lambda (CX) light chains.
  • Heavy chains are classified as mu ( ⁇ ), delta ( ⁇ ), gamma ( ⁇ ), alpha (a), or epsilon ( ⁇ ), and define the antibody's isotype as IgM, IgD, IgG, IgA, and IgE, respectively.
  • isotype as used herein is meant any of the classes and/or subclasses of immunoglobulins defined by the chemical and antigenic characteristics of their constant regions.
  • the known human immunoglobulin isotypes are IgGl (IGHG1), IgG2 (IGHG2), IgG3 (IGHG3), IgG4 (IGHG4), IgAl (IGHA1), IgA2 (IGHA2), IgM (IGHM), IgD (IGHD), and IgE (IGHE).
  • the so-called human immunoglobulin pseudo-gamma IGHGP gene represents an additional human immunoglobulin heavy constant region gene which has been sequenced but does not encode a protein due to an altered switch region (Bensmana M et ah, Nucleic Acids Res. 16(7):3108).
  • the human immunoglobulin pseudo-gamma IGHGP gene has open reading frames for all heavy constant domains (CH1-CH3) and hinge. All open reading frames for its heavy constant domains encode protein domains which align well with all human immunoglobulin constant domains with the predicted structural features.
  • This additional pseudo-gamma isotype is referred herein as IgGP or IGHGP.
  • Other pseudo immunoglobulin genes have been reported such as the human immunoglobulin heavy constant domain epsilon PI and P2 pseudo-genes (IGHEP1 and IGHEP2).
  • the IgG class is the most commonly used for therapeutic purposes. In humans this class comprises subclasses IgGl, IgG2, IgG3 and IgG4. In mice this class comprises subclasses IgGl, IgG2a, IgG2b, IgG2c and IgG3.
  • chimeric antibody or "chimeric anti-TrkA antibody” as used herein includes antibodies in which the variable region sequences are derived from one species and the constant region sequences are derived from another species, such as an antibody in which the variable region sequences are derived from a mouse antibody and the constant region sequences are derived from a human antibody.
  • humanized antibody or "humanised anti-TrkA antibody” as used herein includes antibodies in which CDR sequences derived from the germline of another mammalian species, such as a mouse, have been grafted onto human framework sequences. Additional framework region modifications may be made within the human framework sequences as well as within the CDR sequences derived from the germline of another mammalian species.
  • Fab or "Fab region” as used herein includes the polypeptides that comprise the VH, CHI, VL and CL immunoglobulin domains. Fab may refer to this region in isolation or this region in the context of a full length antibody or antibody fragment.
  • Fc or "Fc region” as used herein includes the polypeptide comprising the constant region of an antibody excluding the first constant region immunoglobulin domain.
  • Fc refers to the last two constant region immunoglobulin domains of IgA, IgD and IgG, and the last three constant region immunoglobulin domains of IgE and IgM, and the flexible hinge N- terminal to these domains.
  • IgA and IgM Fc may include the J chain.
  • Fc comprises immunoglobulin domains Cy2 and Cy3 and the hinge between Cyl and Cy2.
  • the human IgG heavy chain Fc region is usually defined to comprise residues C226 or P230 to its carboxyl-terminus, wherein the numbering is according to the EU numbering system (Edelman GM et al., (1969) PNAS USA 63(1): 78-85).
  • the Fc region is herein defined to comprise residue P232 to its carboxyl-terminus, wherein the numbering is according to the EU numbering system.
  • Fc may refer to this region in isolation or this region in the context of an Fc polypeptide, for example an antibody.
  • the term "hinge” or “hinge region” or “antibody hinge region” herein includes the flexible polypeptide comprising the amino acids between the first and second constant domains of an antibody.
  • the "hinge region” as referred to herein is a sequence region of 6-62 amino acids in length, only present in IgA, IgD and IgG, which encompasses the cysteine residues that bridge the two heavy chains. Structurally, the IgG CHI domain ends at EU position 220, and the IgG CH2 domain begins at residue EU position 237.
  • the antibody hinge is herein defined to include positions 221 (D221 in IgGl) to 231 (A231 in IgGl), wherein the numbering is according to the EU numbering system.
  • parent antibody refers to an unmodified antibody that is subsequently modified to generate a variant.
  • Said parent antibody may be a naturally occurring antibody or a variant or engineered version of a naturally occurring antibody.
  • Parent antibody may refer to the antibody itself, compositions that comprise the parent antibody or the amino acid sequence that encodes it.
  • parental murine antibody or “corresponding parental murine antibody” as used herein is meant an antibody or immunoglobulin that binds human TrkA and is modified to generate a variant, specifically the murine antibody MNAC13 as disclosed in WO00/73344.
  • variant antibody or “antibody variant” as used herein includes an antibody sequence that differs from that of a parent antibody sequence by virtue of at least one amino acid modification compared to the parent.
  • the variant antibody sequence herein will preferably possess at least about 80%, most preferably at least about 90%>, more preferably at least about 95% amino acid sequence identity with a parent antibody sequence.
  • Antibody variant may refer to the antibody itself, compositions comprising the antibody variant or the amino acid sequence that encodes it.
  • amino acid modification herein includes an amino acid substitution, insertion, and/or deletion in a polypeptide sequence.
  • amino acid substitution or “substitution” herein is meant the replacement of an amino acid at a particular position in a parent polypeptide sequence with another amino acid.
  • substitution R94K refers to a variant polypeptide, in this case a heavy chain variable framework region variant, in which the arginine at position 94 is replaced with a lysine.
  • 94K indicates the substitution of position 94 with a lysine.
  • multiple substitutions are typically separated by a slash.
  • R94K/L78V refers to a double variant comprising the substitutions R94K and L78V.
  • amino acid insertion or “insertion” as used herein is meant the addition of an amino acid at a particular position in a parent polypeptide sequence.
  • insert -94 designates an insertion at position 94.
  • amino acid deletion or “deletion” as used herein is meant the removal of an amino acid at a particular position in a parent polypeptide sequence.
  • R94- designates the deletion of arginine at position 94.
  • conservative modifications or “conservative sequence modifications” is intended to refer to amino acid modifications that do not significantly affect or alter the binding characteristics of the antibody containing the amino acid sequence. Such conservative modifications include amino acid substitutions, insertions and deletions. Modifications can be introduced into an antibody of the invention by standard techniques known in the art, such as site-directed mutagenesis and PCR-mediated mutagenesis. Conservative amino acid substitutions are ones in which the amino acid residue is replaced with an amino acid residue having a similar side chain. Families of amino acid residues having similar side chains have been defined in the art.
  • amino acids with basic side chains e.g., lysine, arginine, histidine
  • acidic side chains e.g., aspartic acid, glutamic acid
  • uncharged polar side chains e.g., glycine, asparagine, glutamine, serine, threonine, tyrosine, cysteine, tryptophan
  • non-polar side chains e.g., alanine, valine, leucine, isoleucine, proline, phenylalanine, methionine
  • beta-branched side chains e.g., threonine, valine, isoleucine
  • aromatic side chains e.g., tyrosine, phenylalanine, tryptophan, histidine
  • one or more amino acid residues within the CDR regions or within the framework regions of an antibody of the invention can be replaced with other amino acid residues from the same side chain family and the altered antibody (variant antibody) can be tested for retained function.
  • all human immunoglobulin heavy chain constant domains numbering is according to the "EU numbering system” (Edelman GM et al., ibid.).
  • IGKC human kappa immunoglobulin light chain constant domain
  • IGLCl human lambda immunoglobulin light chain constant domains
  • IGLC2 human lambda immunoglobulin light chain constant domains
  • IGLC3 human lambda immunoglobulin light chain constant domains
  • numbering is according to the "Kabat numbering system" (Kabat EA et al., (1991) Sequences of proteins of immunological interest. 5th Edition - US Department of Health and Human Services, NIH publication n° 91-3242) as described by Dariavach P et al, (1987) PNAS USA 84(24):9074-8 and Frangione B et al, (1985) PNAS USA 82(10):3415-9.
  • the term "variable domain” refers to the domains that mediates antigen-binding and defines specificity of a particular antibody for a particular antigen.
  • the antigen-binding site consists of two variable domains that define specificity: one located in the heavy chain, referred herein as heavy chain variable domain (VH) and the other located in the light chain, referred herein as light chain variable domain (VL).
  • VH heavy chain variable domain
  • VL light chain variable domain
  • specificity may exclusively reside in only one of the two domains as in single-domain antibodies from heavy-chain antibodies found in camelids.
  • the V regions are usually about 110 amino acids long, and consist of relatively invariant stretches of amino acid sequence called framework regions (FRs or "non-CDR regions) of 15-30 amino acids separated by shorter regions of extreme variability called "hypervariable regions" that are 7-17 amino acids long.
  • FRs framework regions
  • variable domains of native heavy and light chains comprise four FRs, largely adopting a beta-sheet configuration, connected by three hypervariable regions, which form loops.
  • the hypervariable regions in each chain are held together in close proximity by FRs and, with the hypervariable regions from the other chain, contribute to the formation of the antigen binding site of antibodies (see Kabat EA et al, ibid.).
  • the term "hypervariable region” as used herein refers to the amino acid residues of an antibody which are responsible for antigen binding.
  • the hypervariable region generally comprises amino acid residues from a "complementary determining region" or "CDR", the latter being of highest sequence variability and/or involved in antigen recognition. For all variable domains numbering is according to Kabat (Kabat EA et al, ibid.).
  • CDR definitions are in use and are encompassed herein.
  • the Kabat definition is based on sequence variability and is the most commonly used (Kabat EA et al., ibid.). Chothia refers instead to the location of the structural loops (Chothia & Lesk J. (1987) Mol. Biol. 196:901-917).
  • the AbM definition is a compromise between the Kabat and the Chothia definitions and is used by Oxford Molecular's AbM antibody modelling software (Martin ACR et al, (1989) PNAS USA 86:9268-9272; Martin ACR et al, (1991) Methods Enzymol.
  • IMGT ® the international ImMunoGeneTics information system ® (http://www.imgt.org) is based on the IMGT numbering for all immunoglobulin and T cell receptor V-REGIONs of all species (IMGT ® , the international ImMunoGeneTics information system ® ; Lefranc MP et al, (1999) Nucleic Acids Res. 27(1):209-12; Ruiz M et al, (2000) Nucleic Acids Res. 28(1):219-21; Lefranc MP (2001) Nucleic Acids Res. 29(l):207-9; Lefranc MP (2003) Nucleic Acids Res.
  • CDRs Complementarity Determining Regions
  • the "non- CDR regions" of the variable domain are known as framework regions (FR).
  • the “non-CDR regions” of the VL region as used herein comprise the amino acid sequences: 1-23 (FR 1), 35- 49 (FR2), 57-88 (FR3), and 99-107 (FR4).
  • the “non-CDR regions” of the VH region as used herein comprise the amino acid sequences: 1-25 (FR1), 36-49 (FR2), 66-94 (FR3), and 1 OS- I (FR4).
  • full length antibody as used herein includes the structure that constitutes the natural biological form of an antibody, including variable and constant regions.
  • the full length antibody of the IgG class is a tetramer and consists of two identical pairs of two immunoglobulin chains, each pair having one light and one heavy chain, each light chain comprising immunoglobulin domains VL and CL and each heavy chain comprising immunoglobulin domains VH, CHI (Cyl), CH2 (Cy2), and CH3 (Cy3).
  • IgG antibodies may consist of only two heavy chains, each heavy chain comprising a variable domain attached to the Fc region.
  • Antibody fragments as used herein refer to antigen-binding fragments include, but are not limited to, (i) the Fab fragment consisting of VL, VH, CL and CHI domains, including Fab' and Fab'-SH, (ii) the Fd fragment consisting of the VH and CHI domains, (iii) the Fv fragment consisting of the VL and VH domains of a single antibody; (iv) the dAb fragment (Ward et al.
  • effector function includes a biochemical event that results from the interaction of an antibody Fc region with an Fc receptor or ligand. Effector functions include FcyR-mediated effector functions such as ADCC (antibody dependent cell-mediated cytotoxicity) and ADCP (antibody dependent cell-mediated phagocytosis), and complement- mediated effector functions such as CDC (complement dependent cytotoxicity).
  • FcyR-mediated effector functions such as ADCC (antibody dependent cell-mediated cytotoxicity) and ADCP (antibody dependent cell-mediated phagocytosis), and complement- mediated effector functions such as CDC (complement dependent cytotoxicity).
  • An effector function of an antibody may be altered by altering, i.e. enhancing or reducing, preferably enhancing, the affinity of the antibody for an effector molecule such as an Fc receptor or a complement component.
  • Binding affinity will generally be varied by modifying the effector molecule binding site and in this case it is appropriate to locate the site of interest and modify at least part of the site in a suitable way. It is also envisaged that an alteration in the binding site on the antibody for the effector molecule need not alter significantly the overall binding affinity but may alter the geometry of the interaction rendering the effector mechanism ineffective as in non-productive binding. It is further envisaged that an effector function may also be altered by modifying a site not directly involved in effector molecule binding, but otherwise involved in performance of the effector function. By altering an effector function of an antibody it may be possible to control various aspects of the immune response, eg enhancing or suppressing various reactions of the immune system, with possible beneficial effects in diagnosis and therapy.
  • the term “subject” includes any human or non-human animal.
  • the term “non-human animal” includes all vertebrates, e.g., mammals and non-mammals, such as primates, sheep, dogs, cats, horses, cows, chickens, amphibians, reptiles, etc.
  • the subject is human.
  • the present invention provides a humanized anti-TrkA antibody or fragment thereof comprising:
  • a heavy chain variable domain comprising a sequence selected from the group consisting of SEQ ID NOs: 1-5, and
  • a light chain variable domain comprising a sequence selected from the group consisting of SEQ ID NOs: 6-13,
  • CDR2 of the heavy chain variable domain comprises at least one amino acid substitution and/or wherein the non-CDR region of the heavy chain variable domain comprises an amino acid substitution at an amino acid position selected from the group consisting of 37, 42 and 89, wherein the amino acid position of each group member is indicated utilizing the numbering system set forth in Kabat.
  • the CDR2 of the heavy chain variable domain comprises at least one conservative amino acid substitution.
  • the non-CDR region of the heavy chain variable domain comprises a conservative amino acid substitution at an amino acid position selected from the group consisting of 37, 42 and 89.
  • the CDR2 of the heavy chain variable domain comprises the sequence of SEQ ID NO: 15.
  • the heavy chain variable domain comprises a sequence selected from the group consisting of SEQ ID NOs: 1, 3 and 5
  • the light chain variable domain comprises a sequence selected from the group consisting of SEQ ID NOs: 6 and 8.
  • the humanized anti-TrkA antibody or fragment thereof comprises a combination of a heavy chain variable domain and a light chain variable domain comprising the sequences selected from the group consisting of SEQ ID NO: 1 and SEQ ID NO: 6, SEQ ID NO: 3 and SEQ ID NO: 6, SEQ ID NO: 3 and SEQ ID NO: 8, SEQ ID NO: 5 and SEQ ID NO: 6, and SEQ ID NO: 5 and SEQ ID NO: 8; preferably the sequences of SEQ ID NO: 5 and SEQ ID NO: 6 or the sequences of SEQ ID NO: 5 and SEQ ID NO: 8; more preferably the sequences of SEQ ID NO: 5 and SEQ ID NO: 6.
  • the heavy chain variable domain of the humanized anti-TrkA antibody or fragment thereof provided by the present disclosure does not comprise the sequence of SEQ ID NO: 71.
  • the heavy chain variable domain of the humanized anti-TrkA antibody or fragment thereof provided by the present disclosure lacks a serine at position 87, wherein the amino acid position of each group member is indicated utilizing the numbering system set forth in Kabat.
  • the heavy chain variable domain of the humanized anti-TrkA antibody or fragment thereof provided by the present disclosure comprises a threonine at position 87, wherein the amino acid position of each group member is indicated utilizing the numbering system set forth in Kabat.
  • the amino acid substitution of the CDR2 of the heavy chain variable domain comprises an amino acid substitution at an amino acid position selected from the group consisting of 50, 60 and 62, preferably selected from the group consisting of 60 and 62, wherein the amino acid position of each group member is indicated utilizing the numbering system set forth in Kabat.
  • the amino acid substitution of the CDR2 of the heavy chain variable domain does not comprise an amino acid substitution selected from the group consisting of Y50A, P60A and T62S, wherein the amino acid position of each group member is indicated utilizing the numbering system set forth in Kabat.
  • the amino acid substitution of the humanized anti-TrkA antibody or fragment in the non-CDR region of the heavy chain variable domain is A49S
  • the amino acid substitution of the humanized anti-TrkA antibody or fragment in the CDR2 of the heavy chain variable domain is not Y50A.
  • the amino acid substitution of the humanized anti-TrkA antibody or fragment in the non-CDR region of the heavy chain variable domain is not A49S and/or the amino acid substitution of the humanized anti-TrkA antibody or fragment in the CDR2 of the heavy chain variable domain is not Y50A.
  • the amino acid substitution of the non-CDR region of the heavy chain variable domain of the antibody or the fragment thereof comprises an amino acid substitution selected from the group consisting of V37A, G42E and V89L, preferably V37A, wherein the amino acid position is indicated utilizing the numbering system set forth in Kabat.
  • the antibody comprises a heavy chain variable domain comprising the sequence of SEQ ID NO: 3, wherein the amino acid substitution of the non-CDR region of the heavy chain variable domain comprises an amino acid substitution selected from the group consisting of V37A, T40A, G42E, R44G, A49S and V89L, preferably selected from the group consisting of V37A, T40A, G42E, R44G and V89L wherein the amino acid position of each group member is indicated utilizing the numbering system set forth in Kabat.
  • the antibody comprises a combination of a heavy chain variable domain and a light chain variable domain comprising the sequences of SEQ ID NO: 3 and SEQ ID NO: 6 or comprising the sequences of SEQ ID NO: 3 and SEQ ID NO: 8, wherein the amino acid substitution of the non-CDR region of the heavy chain variable domain comprises an amino acid substitution selected from the group consisting of V37A, T40A, G42E, R44G, A49S and V89L, preferably selected from the group consisting of V37A, T40A, G42E, R44G and V89L, wherein the amino acid position of each group member is indicated utilizing the numbering system set forth in Kabat.
  • the amino acid substitution of the non-CDR region of the heavy chain variable domain comprises an amino acid substitution selected from the group consisting of K3Q, V37A, G42E, A49S, V89L and R94K, preferably selected from the group consisting of K3Q, V37A, G42E, V89L and R94K, more preferably comprises the amino acid substitutions K3Q and V37A and most preferably comprises the amino acid substitution V37A, wherein the amino acid position of each group member is indicated utilizing the numbering system set forth in Kabat.
  • amino acid substitution of the non-CDR region of the heavy chain variable domain comprises an amino acid substitution selected from the group consisting of V37A and K3Q, V37A and wherein the amino acid position of each group member is indicated utilizing the numbering system set forth in Kabat.
  • the antibody comprises a heavy chain variable domain comprising the sequence of SEQ ID NO: 5, wherein the amino acid substitution of the non-CDR region of the heavy chain variable domain comprises an amino acid substitution selected from the group consisting of K3Q, V37A, G42E, A49S, V89L and R94K, preferably selected from the group consisting of K3Q, V37A, G42E, V89L and R94K, more preferably comprises the amino acid substitutions K3Q and V37A and most preferably comprises the amino acid substitution V37A, wherein the amino acid position of each group member is indicated utilizing the numbering system set forth in Kabat.
  • the antibody comprises a heavy chain variable domain comprising the sequence of SEQ ID NO: 5, wherein the amino acid substitution of the non-CDR region of the heavy chain variable domain comprises an amino acid substitution selected from the group consisting of V37A and K3Q, V37A and wherein the amino acid position of each group member is indicated utilizing the numbering system set forth in Kabat.
  • the present invention provides a humanized anti-TrkA antibody or fragment thereof comprising:
  • a heavy chain variable domain comprising a sequence selected from the group consisting of SEQ ID NOs: 31-49, and
  • a light chain variable domain comprising a sequence selected from the group consisting of SEQ ID NOs: 6-13.
  • the humanized anti-TrkA antibody or fragment thereof comprises: a) a heavy chain variable domain comprising a sequence selected from the group consisting of SEQ ID NOs: 32, 36, 39, 43, 48 and 49, and
  • the humanized anti-TrkA antibody or fragment thereof comprises: a) a heavy chain variable domain comprising a sequence selected from the group consisting of SEQ ID NOs: 32, 36, 48 and 49 and
  • the humanized anti-TrkA antibody or fragment thereof comprises a) a heavy chain variable domain comprising a sequence selected from the group consisting of SEQ ID NOs: 32 and 36, and
  • the humanized anti-TrkA antibody or fragment thereof comprises a) a heavy chain variable domain comprising the sequence of SEQ ID NOs: 36, and b) a light chain variable domain comprising the sequence of SEQ ID NO: 6.
  • the humanized anti-TrkA antibody comprises a combination of a heavy chain variable domain and a light chain variable domain selected from the group comprising the sequences of SEQ ID NO: 32 and SEQ ID NO: 6, SEQ ID NO: 32 and SEQ ID NO: 8, SEQ ID NO: 36 and SEQ ID NO: 6, SEQ ID NO: 48 and SEQ ID NO: 6, SEQ ID NO: 49 and SEQ ID NO: 6, and SEQ ID NO: 49 and SEQ ID NO: 8, preferably selected from the group comprising the sequences of SEQ ID NO: 32 and SEQ ID NO: 6, SEQ ID NO: 32 and SEQ ID NO: 8, SEQ ID NO: 36 and SEQ ID NO: 6, SEQ ID NO: 49 and SEQ ID NO: 6, and SEQ ID NO: 49 and SEQ ID NO: 8, most preferably selected from the combination of sequences of SEQ ID NO: 36 and SEQ ID NO: 6.
  • the humanized anti-TrkA antibody or fragment thereof further comprises heavy and/or light constant regions, preferably heavy and/or light constant regions and a hinge region.
  • the heavy constant regions are of human origin and are e.g. of human IgGl (IGHG1), IgG2 (IGHG2), IgG3 (IGHG3), IgG4 (IGHG4), IgAl (IGHA1), IgA2 (IGHA2), IgM (IGHM), IgD (IGHD), or IgE (IGHE) isotype.
  • the heavy constant regions are of human IGHG1 isotype or are of human IGHG4 isotype.
  • the light constant regions are of human origin and are human kappa (CK) or human lambda (CX) light constant domains, preferably a human kappa light constant domain.
  • the humanized anti-TrkA antibody or fragment thereof further comprises heavy and/or light constant regions and a hinge region, wherein the heavy constant region and the hinge region are of human IGHG1 isotype or are of human IGHG4 isotype. In some embodiments the humanized anti-TrkA antibody or fragment thereof further comprises heavy and/or light constant regions and a hinge region, wherein the heavy constant region and the hinge region are of human IGHG4 isotype and wherein the hinge region comprises amino acid substitution S228P, wherein the amino acid position is indicated utilizing the EU numbering system.
  • the present invention provides a humanized anti-TrkA antibody or fragment thereof comprising:
  • a heavy chain comprising a sequence selected from the group consisting of SEQ ID NOs: 50 to 70, and
  • a light chain comprising a sequence selected from the group consisting of SEQ ID NOs: 29 and 30.
  • the humanized anti-TrkA antibody or fragment thereof comprises a) a heavy chain comprising a sequence selected from the group consisting of SEQ ID NOs: 51, 52, 56, 57, 60, 64, 69 and 70, and
  • a light chain comprising a sequence selected from the group consisting of SEQ ID NOs: 29 and 30, preferably SEQ ID NO: 29.
  • the humanized anti-TrkA antibody or fragment thereof comprises a) a heavy chain comprising a sequence selected from the group consisting of SEQ ID NOs: 51 , 52, 56, 57, 69 and 70, and
  • a light chain comprising a sequence selected from the group consisting of SEQ ID NOs: 29 and 30, preferably SEQ ID NO: 29.
  • the humanized anti-TrkA antibody or fragment thereof comprises a) a heavy chain comprising a sequence selected from the group consisting of SEQ ID NOs: 51, 52, 56, 57, and 70, and
  • a light chain comprising a sequence selected from the group consisting of SEQ ID NOs: 29 and 30, preferably SEQ ID NO: 29.
  • the humanized anti-TrkA antibody or fragment thereof comprises a) a heavy chain comprising a sequence selected from the group consisting of SEQ ID NOs: 51, 52, 56, and 57, and
  • a light chain comprising a sequence selected from the group consisting of SEQ ID NOs: 29 and 30, preferably SEQ ID NO: 29.
  • the humanized anti-TrkA antibody or fragment thereof comprises a) a heavy chain comprising the sequence of SEQ ID NO: 57, and b) a light chain comprising the sequence of SEQ ID NO: 29.
  • the humanized anti-TrkA antibody or fragment thereof is a full length antibody.
  • the humanized anti-TrkA antibody or fragment thereof is an antibody fragment selected from the group consisting of Fab, Fab', Fab'-SH, Fd, Fv, dAb, F(ab')2, scFv, bispecific single chain Fv dimers, diabodies, triabodies and scFv genetically fused to the same or a different antibody; preferably a scFv, or a Fab; more preferably a scFv dimer or a diabody or a F(ab') 2 .
  • the humanized anti-TrkA antibody or fragment thereof comprises a variant Fc region which comprises at least one amino acid modification relative to the Fc region of the parent antibody, whereas the antibody comprising the variant Fc region exhibits altered effector function compared to the parent antibody.
  • Amino acid modification within the Fc region typically alter one or more functional properties of the antibody, such as serum half-life, complement fixation, effector function related to Fc receptor or ligand binding, and/or antigen-dependent cellular cytotoxicity. Modifications within the Fc region as outlined below are according to the EU numbering of residues in the Fc region.
  • the hinge region of CHI is modified such that the number of cysteine residues in the hinge region is altered, e.g., increased or decreased. This approach is described further in U.S. Patent No. 5,677,425 by Bodmer et al.
  • the number of cysteine residues in the hinge region of CHI is altered to, for example, facilitate assembly of the light and heavy chains or to increase or decrease the stability of the antibody.
  • the Fc hinge region of an antibody is mutated to decrease the biological half life of the antibody. More specifically, one or more amino acid mutations are introduced into the CH2-CH3 domain interface region of the Fc-hinge fragment such that the antibody has impaired Staphylococcyl protein A (SpA) binding relative to native Fc-hinge domain SpA binding.
  • SpA Staphylococcyl protein A
  • the antibody is modified to increase its biological half life. Various approaches are possible.
  • the antibody can be altered within the CHI or CL region to contain a salvage receptor binding epitope taken from two loops of a CH2 domain of an Fc region of an IgG, as described in U.S. Patent Nos. 5,869,046 and 6,121,022 by Presta et al.
  • an Fc region is altered by replacing at least one amino acid residue with a different amino acid residue to alter the effector function(s) of the antibody.
  • one or more amino acids selected from amino acid residues 234, 235, 236, 237, 297, 318, 320 and 322 can be replaced with a different amino acid residue such that the antibody has an altered affinity for an effector ligand but retains the antigen- binding ability of the parent antibody.
  • the effector ligand to which affinity is altered can be, for example, an Fc receptor or the CI component of complement. This approach is described in further detail in U.S. Patent Nos. 5,624,821 and 5,648,260, both by Winter et al.
  • one or more amino acids selected from amino acid residues 329, 331 and 322 can be replaced with a different amino acid residue such that the antibody has altered Clq binding and/or reduced or abolished complement dependent cytotoxicity (CDC).
  • CDC complement dependent cytotoxicity
  • This approach is described in further detail in U.S. Patent Nos. 6,194,551 by Idusogie et al.
  • one or more amino acid residues within amino acid positions 231 to 238 in the N- terminal region of the CH2 domain are altered to thereby alter the ability of the antibody to fix complement. This approach is described further in PCT Publication WO 94/29351 by Bodmer et al.
  • the Fc region is modified to increase the ability of the antibody to mediate antibody dependent cellular cytotoxicity (ADCC) and/or to increase the affinity of the antibody for an Fey receptor by modifying one or more amino acids at the following positions: 238, 239, 248, 249, 252, 254, 255, 256, 258, 265, 267, 268, 269, 270, 272, 276, 278, 280, 283, 285, 286, 289, 290, 292, 293, 294, 295, 296, 298, 301, 303, 305, 307, 309, 312, 315, 320, 322, 324, 326, 327, 329, 330, 331, 333, 334, 335, 337, 338, 340, 360, 373, 376, 378, 382, 388, 389, 398, 414, 416, 419, 430, 434, 435, 437, 438 or 439.
  • ADCC antibody dependent cellular cytotoxicity
  • an antibody of the invention may be chemically modified (e.g., one or more chemical moieties can be attached to the antibody) or be modified to alter its glycosylation.
  • the humanized anti-TrkA antibody or fragment thereof is capable of inhibiting the functional activation of TrkA.
  • the humanized anti-TrkA antibody is capable of blocking or reducing one or more biological activities of TrkA.
  • the humanized anti-TrkA antibody or fragment thereof binds to human TrkA with an affinity (K D ) of 500 nM or less, preferably 350nM or less, more preferably 150 nM or less, even more preferably 100 nM or less, most preferably 50 nM or less, in particular 30 nM or less e.g. measured by Surface Plasmon Resonance (SPR) using a BIAcore 2000 instrument (GE Healthcare Europe GmbH, Glattbrugg, Switzerland) or equivalent instrument known in the art by capturing the antibody on the instrument sensor chip with a recombinant monovalent human TrkA extracellular domain (SEQ ID NO: 25) used as analyte.
  • K D affinity
  • TrkA receptor domain a human TrkA receptor domain, like the human TrkA extracellular domain which is not artificially dimerized or multimerized as it would be e.g. if the domain would be amino - terminally fused to an immunoglobulin Fc portion, or which is not naturally dimerized as it would be e.g. if the domain would be associated with its natural ligand NGF.
  • Standard assays to evaluate the binding ability of the antibodies toward e.g. human TrkA are known in the art, including for example, ELISAs, BIAcore, Western blots, RIAs and flow cytometry analysis. Suitable assays are described in detail in the Examples.
  • the binding kinetics (e.g. binding affinity like K D ) of the antibodies also can be assessed by standard assays known in the art, such as by Scatchard or Biacore ® system analysis.
  • the relative binding affinity IQ can be assessed by standard competition assays known in the art.
  • Engineered anti-TrkA antibodies can be assayed for their ability to inhibit the functional activation of TrkA in TF-1 cell proliferation assays.
  • the half maximal inhibitory concentration (IC50) which is a measure of the effectiveness of a compound in inhibiting biological function can be used to select preferred antibodies.
  • the humanized anti-TrkA antibody or fragment thereof has at least an equivalent or lower IC50 in a TF-1 cell proliferation assay than the corresponding parental murine antibody e.g. measured by the ability of the antibody to block cell surface TrkA/beta- NGF mediated cell proliferation using the factor dependent human erythro leukemic cell line TF-1 (Kitamura T et al, (1989) J. Cellular Physiology 140(2):323-34).
  • the humanized anti-TrkA antibody or fragment thereof has an IC50 in a TF-1 cell proliferation assay of 1 ⁇ g/ml or less, more preferably of 0.75 ⁇ g/ml, even more preferably of 0.5 ⁇ g/ml or less, most preferably of 0.3 ⁇ g/ml or less, in particular of 0.1 ⁇ g/ml or less.
  • the humanized anti-TrkA antibody or fragment thereof has a FAB fragment thermostability temperature greater than 65°C, preferably greater than 70°C.
  • FAB fragment thermostability temperature greater than 65°C, preferably greater than 70°C.
  • differential scanning calorimetry measurements are used, whereas a mid-point melting temperature of the FAB fragment in context of a full- length IgG is identified.
  • the humanized antibody of the present invention has a FAB fragment thermostability temperature equivalent to the FAB fragment thermostability temperature of the parental murine antibody while having equivalent affinity as measured by SPR and improved potency as measured by TF-1 cell proliferation assay.
  • the present disclosure also provides a humanized anti-TrkA antibody which has a FAB fragment thermostability temperature which is equivalent to the FAB fragment thermostability temperature of the parental murine antibody with an equivalent affinity for human TrkA and improved inhibitory properties.
  • “Equivalent to the FAB fragment thermostability temperature of the parental murine antibody” as used herein in this context means that the humanized anti-TrkA antibody or fragment thereof has a FAB fragment thermostability temperature which is within a range of ⁇ 20%, preferably within a range of ⁇ 15%, more preferably within a range of ⁇ 10%, even more preferably within a range of ⁇ 5% of the FAB fragment thermostability temperature of the parental murine antibody.
  • the humanized antibody of the present invention has a FAB fragment thermostability temperature not more than 15% lower than the FAB fragment thermostability temperature of the parental murine antibody.
  • “Equivalent affinity for human TrkA” as used herein in this context means that the humanized anti-TrkA antibody or fragment thereof has an affinity which is within a range of ⁇ 20%, preferably within a range of ⁇ 15%, more preferably within a range of ⁇ 10% of the parental murine antibody.
  • the humanized antibody of the present invention has a K D which is at least 5%, preferably at least 10% lower than the K D of the parental murine antibody.
  • the present invention also provides a humanized anti-TrkA antibody or fragment thereof which can be used to treat pain.
  • mice suffering from acute inflammatory hyperalgesia induced by intraplantar injection of Complete Freunds adjuvant (CFA) into the hind paw (see Example 2).
  • CFA Complete Freunds adjuvant
  • Administration of the humanized anti-TrkA antibody at a dose of O.Olmg/kg or above produced a significant reversal of hyperalgesia, which was similar to that observed with the NSAID indomethacin.
  • the effect of a humanized anti-TrkA antibody was tested in mice suffering from chronic inflammatory hyperalgesia induced by intra-articular injection of CFA into the knee joint (see Example 3).
  • Administration of the humanized anti-TrkA antibody at a single dose of 0.01 mg/kg or above produced a significant reversal of hyperalgesia, which was comparable to that observed with multiple dosing of the COX-2 selective NSAID celecoxib.
  • the effect of a humanized anti-TrkA antibody was tested in mice suffering from chronic osteo arthritic hyperalgesia induced by intra-articular injection of monosodium iodoacetate (MIA) into the knee joint (see Example 4).
  • MIA monosodium iodoacetate
  • mice suffering from neuropathic pain induced by chronic constriction of the sciatic nerve (CCI model; see Example 5).
  • Administration of the humanized anti-TrkA antibody at a single dose of 0.01 mg/kg or above produced a significant reversal of mechanical hyperalgesia and cold allodynia, which at the highest dose tested, 1 mg/kg, was comparable to that observed with multiple dosing of pregabalin.
  • a preferred embodiment of the present invention provides a humanized anti-TrkA antibody for the treatment of a patient suffering from acute inflammatory pain, chronic inflammatory pain, osteo arthritic pain and/or neuropathic pain.
  • the present disclosure also provides isolated nucleic acids encoding the anti-TrkA antibodies and fragments thereof, vectors, and host cells comprising the nucleic acid or the vector.
  • the nucleic acids may be present in whole cells, in a cell lysate, or in a partially purified or substantially pure form.
  • a nucleic acid is "isolated” or “rendered substantially pure” when purified away from other cellular components or other contaminants, e.g., other cellular nucleic acids or proteins, by standard techniques, including alkaline/SDS treatment, CsCl banding, column chromatography, agarose gel electrophoresis and others well known in the art, see e.g. Ausubel F et ah, ed.
  • a nucleic acid of the invention can be, for example, DNA or RNA and may or may not contain intronic sequences.
  • the nucleic acid is a cDNA molecule.
  • Nucleic acids of the invention can be obtained using standard molecular biology techniques e.g. cDNAs encoding the light and heavy chains of the antibody or encoding VH and VL segments can be obtained by standard PCR amplification or cDNA cloning techniques.
  • cDNAs encoding the light and heavy chains of the antibody or encoding VH and VL segments can be obtained by standard PCR amplification or cDNA cloning techniques.
  • an immunoglobulin gene library e.g., using phage display techniques
  • one or more nucleic acids encoding the antibody can be recovered from the library.
  • transfection may be either transient or stable.
  • the isolated nucleic acids encoding the anti-TrkA antibodies and fragments thereof comprise a nucleic acid sequence selected from the group consisting of SEQ ID NOs 73-116, usually nucleic acids molecules encoding the light chain variable region or the light chain selected from the group consisting of SEQ ID NOs: 113, 114, 115 and 116 and/or the heavy chain variable region or the heavy chain selected from the group consisting of SEQ ID NOs: 73-112.
  • Preferred nucleic acids molecules of the invention are those encoding the light chain variable region selected from the group consisting of SEQ ID NOs: 113 and 114 and/or the heavy chain variable region selected from the group consisting of SEQ ID NOs: 74, 78, 81, 85, 90 and 91. More preferred are nucleic acids molecules encoding the heavy chain variable region selected from the group consisting of SEQ ID NOs: 74 and 78 and/or encoding the light chain variable region selected from the group consisting of SEQ ID NOs: 113 and 114. Most preferred are nucleic acids molecules encoding the heavy chain variable region comprising the nucleic acid sequence of SEQ ID NO: 74 or 78 and/or encoding the light chain variable region comprising the nucleic acid sequence of SEQ ID NO: 113.
  • nucleic acids molecules of the invention are those encoding the light chain selected from the group consisting of SEQ ID NO: 115 and 116 and/or the heavy chain selected from the group consisting of SEQ ID NO: 93, 94, 98, 99, 102, 106, 111 and 112. More preferred are nucleic acids molecules encoding the heavy chain comprising the nucleic acid sequence of SEQ ID NO: 93, 94, 98, and 99 and/or encoding the light chain selected from the group consisting of of SEQ ID NO: 115 and 116.
  • nucleic acids molecules encoding the heavy chain comprising the nucleic acid sequence of SEQ ID NO: 93, 94, 98, and 99 and/or encoding the light chain comprising the nucleic acid sequence of SEQ ID NO: 115.
  • VH and VL segments are obtained, these DNA fragments can be further manipulated by standard recombinant DNA techniques, for example to convert the variable region genes to full-length antibody chain genes, or to fragment genes corresponding to the fragments described above like e.g. Fab fragment genes or to a scFv gene.
  • a VL- or VH-encoding DNA fragment is operatively linked to another DNA fragment encoding another protein, such as an antibody constant region or a flexible linker.
  • the term "operatively linked”, as used in this context, is intended to mean that the two DNA fragments are joined such that the amino acid sequences encoded by the two DNA fragments remain in-frame.
  • the isolated DNA encoding the VH region can be converted to a full-length heavy chain gene by operatively linking the VH-encoding DNA to another DNA molecule encoding heavy chain constant regions (CHI, CH2 and CH3).
  • CHI, CH2 and CH3 DNA molecule encoding heavy chain constant regions
  • the sequences of human heavy chain constant region genes are known in the art (see e.g., Kabat, EA et al, ibid.) and DNA fragments encompassing these regions can be obtained by standard PCR amplification.
  • the heavy chain constant region can be an IgGl, IgG2, IgG3, IgG4, IgA, IgE, IgM or IgD constant region, but most preferably is an IgG4 constant region, preferably a human IGHG4 constant region wherein the hinge region comprises amino acid substitution S228P.
  • the VH encoding DNA can be operatively linked to another DNA molecule encoding only the heavy chain CHI constant region.
  • the isolated DNA encoding the VL region can be converted to a full-length light chain gene (as well as a Fab light chain gene) by operatively linking the VL-encoding DNA to another DNA molecule encoding the light chain constant region, CL.
  • the sequences of human light chain constant region genes are known in the art (see e.g., Kabat EA et al, ibid.) and DNA fragments encompassing these regions can be obtained by standard PCR amplification.
  • the light chain constant region can be a kappa or lambda constant region, preferably a kappa constant region.
  • the VH- and VL-encoding DNA fragments are operatively linked to another fragment encoding a flexible linker, e.g., encoding the amino acid sequence (Gly4 -Ser)3, such that the VH and VL sequences can be expressed as a contiguous single- chain protein, with the VL and VH regions joined by the flexible linker (see e.g., Bird et al, ibid.; Huston et al, ibid.; McCafferty et al, (1990) Nature 348: 552-554).
  • a flexible linker e.g., encoding the amino acid sequence (Gly4 -Ser)3
  • these fragments were derived via proteolytic digestion of intact antibodies (see e.g. Morimoto et al, (1992) J. Biochem. Biophysical Methods, 24: 107-117 and Brennan et al, (1985) Science, 229:81).
  • these fragments can now be produced directly by recombinant host cells.
  • the antibody fragments can be isolated from the antibody phage libraries discussed above.
  • Fab'-SH fragments can be directly recovered from E. coli and chemically coupled to form F(ab') 2 fragments (Carter et al, (1992) Bio/Technology, 10: 163-167).
  • F(ab')2 fragments can be isolated directly from recombinant host cell culture.
  • the antibody of choice is a single-chain Fv fragment (scFv), see e.g. W093/16185; US Patent Nos. 5,571,894 and 5,587,458.
  • the antibody fragment may also be a "linear antibody", e.g., as described in US Patent No. 5,641,870, for example.
  • the nucleic acids that encode the antibodies of the present invention may be incorporated into a vector, preferably an expression vector in order to express the protein.
  • a variety of expression vectors may be utilized for protein expression.
  • Expression vectors may comprise self-replicating extra- chromosomal vectors or vectors which integrate into a host genome.
  • Expression vectors are constructed to be compatible with the host cell type.
  • vectors, preferably expression vectors, which find use in the present invention include but are not limited to those which enable protein expression in mammalian cells, bacteria, insect cells, yeast, and in in vitro systems.
  • a variety of expression vectors are available, commercially or otherwise, that may find use in the present invention for expressing antibodies.
  • Expression vectors typically comprise a protein operably linked with control or regulatory sequences, selectable markers, any fusion partners, and/or additional elements.
  • operably linked herein is meant that the nucleic acid is placed into a functional relationship with another nucleic acid sequence.
  • regulatory sequence is intended to include promoters, enhancers and other expression control elements (e.g., polyadenylation signals) that control the transcription or translation of the antibody chain genes. Such regulatory sequences are described, for example, in Goeddel (Gene Expression Technology, Methods in Enzymology 185, Academic Press, San Diego, CA (1990)).
  • these expression vectors include transcriptional and translational regulatory nucleic acid operably linked to the nucleic acid encoding the antibody, and are typically appropriate to the host cell used to express the protein.
  • the transcriptional and translational regulatory sequences may include promoter sequences, ribosomal binding sites, transcriptional start and stop sequences, translational start and stop sequences, and enhancer or activator sequences.
  • expression vectors typically contain a selection gene or marker to allow the selection of transformed host cells containing the expression vector. Selection genes are well known in the art and will vary with the host cell used.
  • the selectable marker gene confers resistance to drugs, such as G418, hygromycin or methotrexate, on a host cell into which the vector has been introduced.
  • Preferred selectable marker genes include the dihydro folate reductase (DHFR) gene (for use in dhfr- host cells with methotrexate selection/am lification) and the neo gene (for G418 selection).
  • DHFR dihydro folate reductase
  • Suitable host cells for cloning or expressing the DNA in the vectors herein are prokaryote, yeast, or higher eukaryote cells.
  • Suitable prokaryotes for this purpose include eubacteria, including gram-negative or gram-positive organisms, for example, Enterobacteriaceae such as Escherichia, e.g., E. coli, Enterobacter, Klebsiella, Proteus, Salmonella, e.g., Salmonella typhimurium, Serratia, e.g., Serratia marcescans and Shigella, as well as Bacilli such as B. subtilis and B. licheniformis, Pseudomonas such as P.
  • Enterobacteriaceae such as Escherichia, e.g., E. coli, Enterobacter, Klebsiella, Proteus
  • Salmonella e.g., Salmonella typhimurium
  • Serratia
  • E. coli cloning hosts include E. coli 294 (ATCC 31,446), E. coli B, E. coli XI 776 (ATCC 31,537), and E. coli W3110 (ATCC 27,325).
  • eukaryotic microbes such as filamentous fungi or yeast are suitable cloning or expression hosts. Saccharomyces cerevisiae or common baker's yeast is the most commonly used among lower eukaryotic host microorganisms.
  • Host cells for expressing the recombinant antibodies of the invention are preferably mammalian host cells which include Chinese Hamster Ovary (CHO cells) (including dhfr " CHO cells, described in Urlaub & Chasin (1980) PNAS USA 77:4216-4220, used with a DHFR selectable marker, e.g., as described in Kaufman & Sharp (1982) J. Mol. Biol.
  • NSO myeloma cells 159:601-621
  • COS cells a preferred expression system
  • SP2 cells a preferred expression system
  • HEK293-EBNA1 cells ATCC ® catalogue number: CRL- 10852
  • another preferred expression system is the GS gene expression system disclosed in WO87/04462, WO89/01036 and EP0338841. Construction and Production of Antibodies
  • Antibodies generated against the TrkA polypeptide may be obtained by immunisation of an animal i.e. by administering the polypeptides to an animal, preferably a non-human animal, using well-known and routine protocols, see for example Handbook of Experimental Immunology, Weir DM (ed.), Vol 4, Blackwell Scientific Publishers, Oxford, England, (1986). Many warm-blooded animals, such as rabbits, mice, rats, sheep, cows, camels or pigs may be immunized. However, mice, rabbits, pigs and rats in particular mice are generally most suitable. Antibodies can be produced as well by recombinant DNA techniques known to the skilled person. In additional antibodies can be produced by enzymatic or chemical cleavage of naturally occurring antibodies.
  • Humanized antibodies of the present invention may be constructed by transferring one or more CDRs or portions thereof from VH and/or VL regions from a non-human animal (e.g., mouse) to one or more framework regions from human VH and/or VL regions.
  • the humanized antibodies of the present invention are constructed by transferring one or more CDRs or portions thereof from VH and/or VL regions from murine MNAC13 antibody as disclosed in WO00/73344 to one or more framework regions from human VH and/or VL regions.
  • human framework residues thus present in the VH and/or VL regions may be replaced by corresponding non- human (e.g., mouse) residues when needed or desired for decreasing immunogenicity of the antibody and/or maintaining binding affinity.
  • non-human amino acid residues present in the CDRs may be replaced with human residues.
  • Chimeric or humanized antibodies of the present invention can be prepared based on the sequence of a non-human monoclonal antibody prepared as described above.
  • DNA encoding the heavy and light chain immunoglobulins can be obtained from the non- human hybridoma of interest and engineered to contain non-murine (e.g., human) immunoglobulin sequences using standard molecular biology techniques.
  • murine variable regions can be linked to human constant regions using methods known in the art (see e.g., US Patent No. 4,816,567 to Cabilly et ai).
  • murine CDR regions can be inserted into a human framework using methods known in the art (see e.g., US Patent No. 5,225,539 to Winter and US Patent Nos. 5,530,101; 5,585,089; 5,693,762 and 6,180,370 to Queen et al).
  • Humanized antibodies of the present invention may be constructed wherein the human acceptor molecule for the heavy chain variable region is selected based on homology considerations between potential acceptor molecule variable regions and the heavy chain variable region of the murine antibody. Germline candidate human acceptor molecules are preferred to reduce potential immunogenicity. Germline databases are made up of antibody sequences that read through the end of the heavy chain FW3 region and partially into the CDR3 sequence. For selection of a FW4 region, databases of mature antibody sequences which have been derived from the selected germline molecule can be searched or antibody sequences which have been derived from the selected germline molecule from a human donor can be used.
  • Human acceptor molecules are preferably selected from the same heavy chain class as the murine donor molecule, and of the same canonical structural class of the variable region of the murine donor molecule. Secondary considerations for selection of the human acceptor molecule for the heavy chain variable region include homology in CDR length between the murine donor molecule and the human acceptor molecule. Human acceptor antibody molecules are preferably selected by homology search to the V-BASE database, although other databases such as the Kabat and the public NCBI databases may be used as well. Humanized antibodies of the present invention may be constructed wherein the human acceptor molecule for the light chain variable region is selected based on homology considerations between potential acceptor molecule variable regions and with the light chain variable region of the murine antibody.
  • Germline candidate human acceptor molecules are preferred to reduce potential immunogenicity.
  • Germline databases are made up of antibody sequences that read through the end of the heavy chain FW3 region and partially into the CDR3 sequence. For selection of a FW4 region, databases of mature antibody sequences which have been derived from the selected germline molecule can be searched or antibody sequences which have been derived from the selected germline molecule from a human donor can be used.
  • Human acceptor molecules are preferably selected from the same light chain class as the murine donor molecule and of the same canonical structural class of the variable region of the murine donor molecule. Secondary considerations for selection of the human acceptor molecule for the light chain variable region include homology in CDR length between the murine donor molecule and the human acceptor molecule.
  • Human acceptor antibody molecules are preferably selected by homology searches to the V-BASE database, and other databases such as the Kabat and the public NCBI databases may be used as well.
  • the antibody is produced as recombinant antibody by e.g. introducing genes into mammalian host cells, the antibodies are produced by culturing the host cells for a period of time sufficient to allow for expression of the antibody in the host cells or, more preferably, for secretion of the antibody into the culture medium in which the host cells are grown.
  • Host cells useful for producing antibodies that bind to human TrkA may be cultured in a variety of media.
  • Antibodies may be operably linked to a fusion partner to enable targeting of the expressed protein, purification, screening, display and the like. Fusion partners may be linked to the antibody sequence via a linker sequences.
  • the linker sequence will generally comprise a small number of amino acids, typically less than ten, although longer linkers may also be used. Typically, linker sequences are selected to be flexible and resistant to degradation. As will be appreciated by those skilled in the art, any of a wide variety of sequences may be used as linkers.
  • a common linker sequence comprises the amino acid sequence GGGGS.
  • a fusion partner may be a targeting or signal sequence that directs antibody and any associated fusion partners to a desired cellular location or to the extracellular media.
  • fusion partner may also be a sequence that encodes a peptide or protein that enables purification and/or screening.
  • fusion partners include but are not limited to polyhistidine tags (His-tags) (for example H6 and H10 or other tags for use with Immobilized Metal Affinity Chromatography (IMAC) systems (e.g.
  • tags which are targeted by antibodies (for example c-myc tags, flag- tags and the like).
  • antibodies for example c-myc tags, flag- tags and the like.
  • Screening for antibodies can be performed using assays to measure binding to human TrkA and/or assays to measure the ability to block the binding of TrkA to its ligand NGF.
  • An example of a binding assay is an ELISA.
  • SPR Surface Plasmon Resonance
  • An example of a blocking assay is a flow cytometry based assay measuring the blocking of NGF binding to TrkA.
  • As an assay for evaluating the functional activity of anti-TrkA antibodies e.g.
  • TF-1 cell proliferation assay can be used, wherein the ability of antibodies to block cell surface TrkA/beta-NGF mediated cell proliferation is assayed using the factor dependent human erythro leukemic cell line TF-1 (Kitamura T et al., (1989) J. Cellular Physiology 140(2):323- 34).
  • Antibodies of the present invention may be isolated or purified in a variety of ways known to those skilled in the art.
  • Standard purification methods include chromatographic techniques, including ion exchange, hydrophobic interaction, affinity, sizing or gel filtration and reversed- phase, carried out at atmospheric pressure or at high pressure using systems such as FPLC and HPLC.
  • Purification methods also include electrophoretic, immunological, precipitation, dialysis and chromatofocusing techniques. Ultrafiltration and diafiltration techniques, in conjunction with protein concentration, are also useful.
  • TrkA antibodies selected host cells can be grown in e.g. spinner- flasks for monoclonal antibody purification.
  • a preferred antibody of the present invention is thus an isolated and/or purified antibody that binds to human TrkA.
  • the present invention provides anTrkA antibody or a fragment thereof that binds to human TrkA, linked to a therapeutic agent, such as a cytotoxin, a drug (e.g., an immunosuppressant) or a radiotoxin.
  • a therapeutic agent such as a cytotoxin, a drug (e.g., an immunosuppressant) or a radiotoxin.
  • conjugates are referred to herein as “immunoconjugates”.
  • Immunoconjugates that include one or more cytotoxins are referred to as "immunotoxins.”
  • a cytotoxin or cytotoxic agent includes any agent that is detrimental to (e.g., kills) cells.
  • Examples include taxol, cytochalasin B, gramicidin D, ethidium bromide, emetine, mitomycin, etoposide, tenoposide, vincristine, vinblastine, colchicin, doxorubicin, daunorubicin, dihydroxy anthracin dione, mitoxantrone, mithramycin, actinomycin D, 1- dehydrotestosterone, glucocorticoids, procaine, tetracaine, lidocaine, propranolol, and puromycin and analogs or homologs thereof.
  • Therapeutic agents also include, for example, antimetabolites (e.g., methotrexate, 6- mercaptopurine, 6-thioguanine, cytarabine, 5- fluorouracil decarbazine), alkylating agents (e.g., mechlorethamine, thioepa chlorambucil, melphalan, carmustine (BSNU) and lomustine (CCNU), cyclothosphamide, busulfan, dibromomannitol, streptozotocin, mitomycin C, and cis-dichlorodiamine platinum (II) (DDP) cisplatin), anthracyclines (e.g., daunorubicin (formerly daunomycin) and doxorubicin), antibiotics (e.g., dactinomycin (formerly actinomycin), bleomycin, mithramycin, and anthramycin (AMC)) and anti-mitotic agents (e.g.,
  • An example of a calicheamicin antibody conjugate is commercially available (Mylotarg(R); American Home Products).
  • Cytotoxins can be linked to antibodies of the invention using linker technology available in the art. Examples of linker types that have been used to conjugate a cytotoxin to an antibody include, but are not limited to, hydrazones, thioethers, esters, disulfides and peptide-containing linkers.
  • a linker can be chosen that is, for example, susceptible to cleavage by low pH within the lysosomal compartment or susceptible to cleavage by proteases, such as proteases preferentially expressed in tumor tissue such as cathepsins (e.g., cathepsins B, C, D).
  • proteases such as proteases preferentially expressed in tumor tissue such as cathepsins (e.g., cathepsins B, C, D).
  • Antibodies of the present invention also can be linked to a radioactive isotope to generate cytotoxic radiopharmaceuticals, also referred to as radioimmunoconjugates.
  • radioactive isotopes that can be conjugated to antibodies for use diagnostically or therapeutically include, but are not limited to, iodine-131, indium-I l l, yttrium-90 and lutetium-177.
  • radioimmunoconjugates Methods for preparing radioimmunconjugates are established in the art. Examples of radioimmunoconjugates are commercially available, including Zevalin ® (EDEC Pharmaceuticals) and Bexxar ® (Corixa Pharmaceuticals) and similar methods can be used to prepare radioimmunoconjugates using the antibodies of the invention.
  • the antibody immunoconjugates of the invention can be used to modify a given biological response, and the drug moiety is not to be construed as limited to classical chemical therapeutic agents.
  • the drug moiety may be a protein or polypeptide possessing a desired biological activity.
  • Such proteins may include, for example, an enzymatically active toxin, or active fragment thereof, such as abrin, ricin A, pseudomonas exotoxin, or diphtheria toxin; a protein such as tumor necrosis factor or interferon- ⁇ ; or biological response modifiers such as, for example, lymphokines, interleukin-1 (IL-1), interleukin-2 (IL-2), interleukin-6 (IL-6), granulocyte macrophage colony stimulating factor (GM-CSF), granulocyte colony stimulating factor (G-CSF), or other growth factors.
  • IL-1 interleukin-1
  • IL-2 interleukin-2
  • IL-6 interleukin-6
  • GM-CSF granulocyte macrophage colony stimulating factor
  • G-CSF granulocyte colony stimulating factor
  • the present invention provides an anti-TrkA antibody or a fragment thereof that binds to human TrkA, administered together with a therapeutic agent, such as a cytotoxin, a drug (e.g., an immunosuppressant) or a radiotoxin.
  • a therapeutic agent such as a cytotoxin, a drug (e.g., an immunosuppressant) or a radiotoxin.
  • the present invention provides a composition, e.g., a pharmaceutical composition, comprising the antibody or fragment thereof of the present invention and a pharmaceutically acceptable carrier.
  • a composition may include one or a combination of (e.g., two or more different) antibodies, and/or immunoconjugates of the invention and/or a therapeutic agent, such as a cytotoxin, a drug (e.g., an immunosuppressant) or a radiotoxin as described supra.
  • a pharmaceutical composition of the invention can comprise a combination of antibodies (or immunoconjugates) that bind to different epitopes on the target antigen or that have complementary activities.
  • Pharmaceutical compositions of the invention also can be administered in combination therapy, i.e., combined with other agents as outlined further below.
  • pharmaceutically acceptable carrier includes any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents and the like that are physiologically compatible.
  • the carrier is suitable for intravenous, intramuscular, subcutaneous, parenteral, spinal or epidermal administration (e.g. by injection or infusion).
  • the active compound i.e. antibody or immunoconjugate may be coated in a material to protect the compound from the action of acids and other natural conditions that may inactivate the compound.
  • Pharmaceutically acceptable carriers include sterile aqueous solutions or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersion.
  • the present invention provides a composition comprising the humanized anti-TrkA antibody or fragment thereof of the present invention and another pharmaceutically active agent.
  • the pharmaceutically active agent is one or more of:
  • the present invention provides a composition comprising an immunoconjugate comprising the antibody or fragment thereof that binds to human TrkA linked to a therapeutic agent and a pharmaceutically acceptable carrier.
  • Immunoconjugates and therapeutic agents which can be used are as described supra.
  • a pharmaceutical composition of the invention may also include a pharmaceutically acceptable antioxidant.
  • pharmaceutically acceptable antioxidants include: (1) water soluble antioxidants, such as ascorbic acid, cysteine hydrochloride, sodium bisulfate, sodium metabisulfite, sodium sulfite and the like; (2) oil- soluble antioxidants, such as ascorbyl palmitate, butylated hydroxyanisole (BHA), butylated hydroxytoluene (BHT), lecithin, propyl gallate, alpha-tocopherol and the like; and (3) metal chelating agents, such as citric acid, ethylenediamine tetraacetic-acid (EDTA), sorbitol, tartaric acid, phosphoric acid and the like.
  • water soluble antioxidants such as ascorbic acid, cysteine hydrochloride, sodium bisulfate, sodium metabisulfite, sodium sulfite and the like
  • oil- soluble antioxidants such as ascorbyl palmitate, butylated hydroxy
  • aqueous and non-aqueous carriers examples include water, ethanol, polyols (such as glycerol, propylene glycol, polyethylene glycol and the like) and suitable mixtures thereof, vegetable oils, such as olive oil and injectable organic esters, such as ethyl oleate.
  • polyols such as glycerol, propylene glycol, polyethylene glycol and the like
  • vegetable oils such as olive oil
  • injectable organic esters such as ethyl oleate.
  • Proper fluidity can be maintained, for example, by the use of coating materials, such as lecithin, by the maintenance of the required particle size in the case of dispersions and by the use of surfactants.
  • These compositions may also contain adjuvants such as preservatives, wetting agents, emulsifying agents and dispersing agents.
  • Prevention of presence of microorganisms may be ensured both by sterilization procedures, supra and by the inclusion of various antibacterial and antifungal agents, for example, paraben, chlorobutanol, phenol sorbic acid and the like. It may also be desirable to include isotonic agents, such as sugars, sodium chloride and the like into the compositions. In addition, prolonged absorption of the injectable pharmaceutical form may be brought about by the inclusion of agents which delay absorption such as aluminum monostearate and gelatin.
  • Antibodies of the present invention can be used in medicine to treat various disorders/ conditions, as set out in various categories below.
  • the invention thus provides a method of treatment of the below mentioned conditions which comprises administering to a subject, suitably a mammalian subject, especially a human subject in need thereof, a therapeutically effective amount of an antibody or derivative as described herein such that the condition is thereby treated.
  • the invention also provides use of an antibody or derivative as described herein in the manufacture of a medicament for the treatment of the below mentioned conditions.
  • treatment includes therapeutic treatment of an existing disorder/condition. It also includes prophylactic treatment. It further includes the amelioration of one or more adverse symptoms, even if a patient is not cured of a given disorder/condition. For example, pain may be alleviated or reduced.
  • a preferred medical use is in the treatment of pain.
  • IASP International Association for the Study of Pain
  • pain is generally defined as "An unpleasant sensory and emotional experience associated with actual or potential tissue damage, or described in terms of such damage or both”.
  • the essential element in all forms of pain is the activation of specialized high-threshold receptors and nerve fibers to warn the organism of potential tissue damage. The involvement of inflammatory cells and processes is a common element in many pain states.
  • acute pain means immediate, generally high threshold, pain brought about by injury such as a cut, crush, burn, or by chemical stimulation.
  • chronic pain means pain other than acute pain, both of inflammatory and neuropathic origin. It is understood that chronic pain often is of relatively long duration, for example, months or years and can be continuous or intermittent.
  • Antibodies of the present invention can be used to treat chronic pain or acute pain. The treatment of chronic pain is preferred.
  • the pain may for example be or may be associated with any of the following: inflammatory pain, post-surgical pain, post-operative pain (including dental pain), neuropathic pain, peripheral neuropathy, diabetic neuropathy, diabetic nephropathy, fracture pain, gout joint pain, post-herpetic neuralgia, cancer pain, osteoarthritis or rheumatoid arthritis pain, sciatica, pains associated with sickle cell crises, headaches (e.g., migraines, tension headache, cluster headache), dysmenorrhea, endometriosis, uterine fibroids, musculoskeletal pain, chronic low back pain, fibromyalgia, sprains, visceral pain, ovarian cysts, prostatitis, chronic pelvic pain syndrome, cystitis, interstitial cystitis, painful bladder syndrome and/or bladder pain syndrome, pain associated with chronic abacterial prostatitis, incisional pain, migraine, trigeminal neuralgia, pain from burns and/or wounds, pain
  • the pain may for example be or may be associated with any of the following: pancreatitis, kidney stones, endometriosis, IBD, Crohn's disease, post surgical adhesions, gall bladder stones, headaches, dysmenorrhea, musculoskeletal pain, sprains, visceral pain, ovarian cysts, prostatitis, cystitis, interstitial cystitis, post-operative pain, migraine, trigeminal neuralgia, pain from burns and/or wounds, pain associated with trauma, neuropathic pain, pain associated with musculoskeletal diseases, rheumatoid arthritis, osteoarthritis, ankylosing spondilitis, periarticular pathologies, oncological pain, pain from bone metastases, HIV infection.
  • the nociception hot plate test can be used, as disclosed in WO 00/73344, for example.
  • the experiment can be carried out according to McMahon et al, 1995 (Nature Med. 1 :774-780), using the antibody/derivative as immunoadhesin.
  • the antibody/derivative is infused subcutaneously into hind paw of an adult rat for a period of three weeks or by an osmotic mini-pump.
  • the nociception sensitivity is evaluated at intervals using a hot plate test (Eddy & Leimbach (1953) J. Phar. Exp. Ther.
  • the nociceptive stimulus induces in such a case a response (paw licking and/or jumping) which presumes an integrated coordination higher than simple reflex.
  • the animal is put in a pen having a plate heated to the desired temperature as base, usually 56°C.
  • the latency of any of two responses is measured in control animals (treated with non relevant antibody) and in those treated with the anti-TrkA antibody/derivative.
  • the nociceptive response to formalin can be assessed.
  • This test is disclosed by Porro & Cavazzuti, 1993 (Prog. Neurobiol, 41 :565-607) and was used in WO06/137106. It involves assessing the reduction in pain response by analyzing any subsequent reduction paw licking when a given candidate is administered prior to testing. Saline is typically used as a negative control.
  • An assessment of hyperalgesia can be determined using a weight bearing method.
  • Mice normally distribute their body weight equally between their two limbs. Following a painful stimulus to the limb, for example by the local injection of Complete Freunds adjuvant (CFA) or monosodium iodoacetate (MIA) to the hind paw (intraplantar) or knee joint (intraarticular), the weight is redistributed to reduce that placed on the injected limb and increase that placed on the non- injected limb. Weight bearing is measured using an incapacitance tester with the hind paws placed on separate sensors and the average force exerted by both hind limbs recorded.
  • CFA Complete Freunds adjuvant
  • MIA monosodium iodoacetate
  • Weight bearing can be used to assess acute inflammatory hyperalgesia resulting from intraplantar injection of CFA, chronic inflammatory hyperalgesia resulting from intra-articular injection of CFA or chronic osteoarthritic hyperalgesia resulting from intra-articular injection of MIA, as detailed in Examples 2, 3 and 4, respectively.
  • An assessment of mechanical hyperalgesia can be performed by a number of methods, for example, Von Frey filaments or a Randall-Selitto analgesiometer. Paw withdraw thresholds are measured in response to increasing pressure stimuli applied to the plantar hind paw surface by von Frey filaments or to the dorsal hind paw surface by a wedge-shaped probe of a Randall-Selitto analgesiometer. The latency of hind paw withdraw is measured in control animals and in those treated with the anti-TrkA antibody/derivative. These methods can be used to assess mechanical hyperalgesia resulting from the Chronic Constriction Injury (CCI) animal model as detailed in Example 5.
  • CCI Chronic Constriction Injury
  • the CCI model is also a well known animal model. It involves chronic constriction of the sciatic nerve and is used to induce chronic pain of a neuropathic nature in rodents, such as mice or rats. This model is described by Bennett & Xie, 1998 in Pain 33:87- 107. It was used in WO 06/131592, for example.
  • a major feature of many neuropathic pain states is that normally innocuous cool stimuli begin to produce pain.
  • An increased response to a non- painful stimulus is termed allodynia. The extent of the neuropathic pain state induced can therefore be measured using a cold plate test for allodynia, as detailed in Example 5.
  • the animal is put in a pen having a plate cooled to the desired temperature, which can be in the range of -5°C to 15°C.
  • the latency of hind paw withdraw is measured in control animals and in those treated with the anti-TrkA antibody/derivative. Typically the latency for withdrawal becomes different from the baseline at surface temperatures of 5°C and below.
  • the antibodies can be used in the treatment of cancer, a neuronal disorder, Alzheimer's disease, diabetes mellitus, diabetic nephropathy, a viral disorder, an HIV mediated disorder, leprosy or an inflammatory disorder.
  • the antibodies are also useful in treating other diseases that may be associated with increased levels of NGF including, for example, lupus erythematosus, shingles, postherpetic neuralgia, and hyperalgesia.
  • TrkA Various cancers express TrkA.
  • the interaction of TrkA with NGF may be involved in tumour development (e.g. of prostate and pancreatic cancers). Indeed in certain forms of cancer, an excess of NGF can facilitate the growth and infiltration of nerve fibres. By blocking the action of NGF it is possible to significantly reduce the formation of neuromas.
  • the antibodies can be coupled to a cytotoxic agent and can be used to target cancer cells expressing TrkA. It is not however necessary to couple the antibodies to toxins.
  • ADCC antibody-dependent cell-mediated cytotoxicity
  • Preferred cancers to be treated are prostate cancer, thyroid cancer, lung cancer, prolactinoma, melanoma or bone cancer pain including cancer pain associated with bone metastasis.
  • a preferred cancer to be treated is bone cancer pain including cancer pain associated with bone metastasis.
  • the antibodies can also be used in the treatment of various neuronal disorders which comprise neurodegenerative disorders e.g. the antibodies can be used to reduce the formation of neuromas. They can also be used in the treatment of Alzheimer's disease or in neuroregenerative therapies. Antibodies of the present invention may be useful in such treatments to reduce undesired agonist effects of NGF (see also the "Combination therapy" section below). Furthermore, the antibodies can be used to treat neuropathic pain, as discussed above. This may be associated with a lesion or a dysfunction of the nervous system. NGF has potential use in the treatment of diabetes and leprosy but has undesired agonist properties including an increase in pain sensitivity which could be avoided by using the antibodies of the present invention. A still further application is in the treatment of inflammatory disorders.
  • NGF is released by mast cells, fibroblasts and other cell types in the peripheral sites where inflammatory processes occur.
  • mast cells appear to play a fundamental role. They produce NGF and at the same time express functional TrkA receptors at their surface.
  • the NGF/TrkA system appears to mediate mastocyte activation through an autocrine positive feedback mechanism which allows local amplification of the algogenic inflammatory signal.
  • inflammatory disorders examples include inflammatory forms of the urinary tract and of the pelvic region, osteoarthritis, multiple sclerosis, colitis, inflammatory bowel disease, bladder cystitis, eczema, contact dermititis, arthritis, including chronic arthritis and rheumatoid arthritis, Crohn's disease, psoriasis and asthma.
  • Antibodies of the present invention may be useful in such treatments as mentioned above to reduce undesired agonist effects of NGF.
  • Antibodies or derivatives thereof of the present invention may be used together with one or more other active agents, e.g. pharmaceutically active agents in combination therapy. They may be used for simultaneous, sequential or concerted administration in medicine.
  • the antibody or derivative may be combined with an analgesic agent such as an analgesic opioid or a non-opioid analgesic. It is disclosed in WO06/137106 that small amounts of molecules able to block TrkA biological activity can potentiate the analgesic effects of opioids.
  • Such analgesic opioids include one or more compounds selected from the following: morphine, codeine, dihydrocodeine diacetylmorphine, hydrocodone, hydromorphone, levorphanol, oxymorphone, alfentanil, buprenorphine, butorphanol, fentanyl, sufentanyl, meperidine, methadone, nabumetone , propoxyphene, pentazocine; and their pharmaceutically acceptable derivatives thereof (e.g. pharmaceutically acceptable salts thereof).
  • Suitable non-opioid analgesics include non-steroidal inflammatory drugs (NSAIDs) as well as other analgesics such as acetaminophen.
  • NSAIDs non-steroidal inflammatory drugs
  • Commonly available NSAIDs to treat acute inflammatory pain include asprin, ibuprofen, indomethacin, naproxen and ketoprofen and to treat chronic inflammatory pain include celecoxib and meloxicam.
  • a further combination is that of one or more antibodies of the present invention together with one or more other antibodies.
  • a preferred combination is with one or more other anti-TrkA and/or an anti-NGF antibody.
  • Such combinations may provide increased efficacy in treating one or more of the disorders discussed herein, relative to treatment with a single antibody.
  • combinations of two or more antibodies found to be amongst the most effective in assay procedures used herein may be used.
  • a further combination is that of the antibody of the present invention with NGF.
  • NGF As discussed above, the use of NGF in the treatment of various disorders, including Alzheimer's disease, diabetes mellitus, leprosy, etc., had been proposed, but an increase in pain sensitivity had been noted arising from agonist properties towards peripheral targets. Again, by using an antibody or derivative of the present invention, pain sensitivity can be reduced, thereby making NGF- based therapies more attractive.
  • a further combination is that of the antibody of the present invention with an anti-cancer agent such as e.g. an alkylating agent, an antimetabolite, a topoisomerase II inhibitor, a topoisomerase I inhibitor, an antimitotic drug or a platinum derivative.
  • an anti-cancer agent such as e.g. an alkylating agent, an antimetabolite, a topoisomerase II inhibitor, a topoisomerase I inhibitor, an antimitotic drug or a platinum derivative.
  • the antibodies of the present invention can be administered by any appropriate route.
  • the antibodies can typically be administered for local application by injection (intraperitoneal or intracranial-typically in a cerebral ventricle-or intrapericardiac or intrabursal) of liquid formulations or by ingestion of solid formulations (in the form of pills, tablets, capsules) or of liquid formulations (in the form of emulsions and solutions).
  • compositions for parenteral administration commonly comprise a solution of immunoglobulin dissolved in a compatible, preferably aqueous solution.
  • concentration of the antibody/derivative in these formulations can vary from less than 0.005% to 15-20% w/v. It is selected mainly according to the volumes of the liquid, viscosity, etc, and according to the particular administration mode desired.
  • the antibodies can be prepared for administration in solid form.
  • the antibodies can be combined with different inert or excipient substances, which can include ligands such as microcrystalline cellulose, gelatin or Arabic rubber; recipients such lactose or starch; agents such as alginic acid, Primogel or corn starch; lubricants such as magnesium stearate, colloidal silicon dioxide; sweeteners such as saccharose or saccharin; or flavours, such as mint and methyl salicylate.
  • ligands such as microcrystalline cellulose, gelatin or Arabic rubber
  • recipients such lactose or starch
  • agents such as alginic acid, Primogel or corn starch
  • lubricants such as magnesium stearate, colloidal silicon dioxide
  • sweeteners such as saccharose or saccharin
  • flavours such as mint and methyl salicylate.
  • Other pharmaceutical administration systems include hydrogel, hydroxymethylcellulose, liposomes, microcapsules, microemulsions, microspheres, etc.
  • the anti-TrkA antibodies are suitably administered systemically.
  • Systemic administration can be performed by injection, e.g. continuous intravenous infusion, bolus intravenous infusion, subcutaneous or intramuscular injection.
  • other forms of administration e.g. oral, mucosal, via inhalation, sublingually, etc.
  • delivery of the antibody/ derivative can be performed by local administration (e.g. intra-articular injection or subcutaneous, intramuscular injection) in the vicinity of affected tissues.
  • the anti-TrkA antibody/derivative will suitably be formulated in a pharmaceutical composition appropriate for the intended route of administration.
  • Solutions for injection will suitably contain the antibody/derivative dissolved or dispersed in an aqueous medium (e.g. water for injection) as appropriate containing appropriate buffers and molarity modifiers (e.g. phosphate, salt and/or dextrose).
  • aqueous medium e.g. water for injection
  • appropriate buffers and molarity modifiers e.g. phosphate, salt and/or dextrose.
  • the treatment regime i.e. dose, timing and repetition
  • the interval of dose administration can be subject to modifications depending on the extent and duration of the clinical response, as well as the particular individual and the individual clinical history.
  • the anti-TrkA antibody/derivative has a long duration of action.
  • the clinical effect of the antibody extends following administration may be as long as 21 days as determined from animal studies.
  • anti-TrkA antibodies may manifest clinical benefit for a longer period than that in which its presence can be detected in a relevant biological matrix such as serum or plasma following its administration.
  • the antibody/derivative may be administered to subjects at a frequency of not more than once per week e.g. not more than once per two weeks or once per three weeks or once per four weeks.
  • a suitable daily dose of the anti-TrkA antibody/derivative will typically range from 0.1 mg/kg to 10 mg/kg body weight.
  • a suitable dose of the anti-TrkA antibody for the treatment of acute and chronic inflammatory pain is at least O.Olmg/kg (see Examples 2 and 3).
  • a suitable dose of the anti-TrkA antibody for the treatment of osteoarthritic pain is at least O.Olmg/kg (see Example 4).
  • a suitable dose of the anti-TrkA antibody for the treatment of neuropathic pain is at least 0.01 mg/kg, preferably 0.1 mg/kg and most preferably 1 mg/kg (see Example 5). These doses relate to an in vivo condition in mice only.
  • the present invention relates the use of a humanized anti-TrkA antibody in the treatment of acute inflammatory pain, wherein hyperalgesia is effectively reversed to the same extent as that observed with administration of a NSAID.
  • the present invention also relates to the use of a humanized anti-TrkA antibody in the treatment of chronic inflammatory pain, wherein hyperalgesia is effectively reversed to the same extent as that observed with administration of a NSAID.
  • the present invention also relates to the use of a humanized anti-TrkA antibody in the treatment of osteoarthritic pain, wherein hyperalgesia is effectively reversed to the same extent as that observed with administration of pregabalin and an opiate.
  • the present invention also relates to the use of a humanized anti-TrkA antibody in the treatment of neuropathic pain, wherein hyperalgesia is effectively reversed to the same extent as that observed with administration of pregabalin.
  • administration may be through direct and localized injection into a tumour or a tissue near the tumour site.
  • doses vary from 0.05 mg/kg per day to 500 mg/kg per day, although dosages in the lower region of the range are preferred because they are easier to administer.
  • Dosages can be calibrated for example to guarantee a particular level in the plasma of the antibody/derivative (in the range of about 5-30 mg/ml, preferably between 10-15 mg/ml) and maintain this level for a given period of time until the clinical results are achieved.
  • Effective methods for measuring or assessing the stage of pancreatic or prostatic tumours are based on the measurement of the prostate specific antigen (PSA) in blood, on the measurement of the survival time for pancreas tumours, on the measurement of the slowing or inhibition of diffusion for metastases in the case of both tumour types.
  • PSA prostate specific antigen
  • dosage depends on different factors including the type, stage and volume of the tumour, along with many other variables.
  • typical therapeutic doses may vary from 0.01 mg/ml and 10 mg/ml injections which can be administered with the necessary frequency.
  • humanised antibodies may be eliminated much more slowly and require lower dosages to maintain an effective level in the plasma than non- humanised antibodies. Moreover, with high affinity antibodies, administration may be less frequent and less sizable than with antibodies having lower affinity.
  • each antibody/ derivative can be determined during the treatment by a skilled medical practitioner. If necessary, dosages can be reduced (e.g. to reduce side effects) or increased (to increase therapeutic effects).
  • preparations of antibodies of the invention Prior to administration, preparations of antibodies of the invention can be stored by being frozen or lyophilized. They may then be reconstituted immediately before use in a suitable buffer. Given that lyophilisation and reconstitution can result in a loss in activity, antibody administration levels can be calibrated to compensate for this fact. (For conventional immunoglobulins, IgM antibodies tend to have a greater loss of activity than IgG antibodies.) A shelf life may also be assigned so that antibodies are not used after a certain period of storage.
  • An antibody or derivative thereof of the present invention can be used in the diagnosis or prognosis of any of the diseases/ conditions discussed above in relation to medical uses.
  • TrkA positive tumour markers may be used to facilitate detection of TrkA positive tumour markers, as a precocious marker of the insurgence of Alzheimer's disease, etc.
  • CIPA congenital insensitivity to pain with anhydrosis
  • This is a hereditary, recessive, autosomal syndrome characterised by recurrent episodic fever, anhydrosis, the absence of reaction to nociceptive stimuli, mental retardation and a tendency to self-mutilation. It results from mutations in the TrkA gene.
  • an antibody or derivative of the present invention may be used in the diagnosis or prognosis of a wide range of conditions involving aberrant expression of TrkA (compared to expression of TrkA in a healthy individual or a healthy tissue sample) or an aberrant activity involving TrkA.
  • the present invention therefore includes within its scope a method comprising obtaining a biological sample obtained from a patient and contacting the sample with an antibody or derivative of the present invention. If desired, the antibody/derivative may be immobilised. The method may then include assaying the binding of the antibody / derivative to said sample in a quantitative or qualitative manner. If desired, this may be done with reference and/or to a positive control (indicating a healthy state) or a negative control (indicating the presence/likelihood of a disorder). For diagnostic purposes, the antibodies can be both marked with a detectable marker or can be unmarked. (The term "marker" is used herein to include labels or any other detectable moiety/moiety that can trigger a detectable change.)
  • an article of manufacture comprising the antibody or fragment thereof, the composition or the immunoconjugate of the invention for the treatment of one or more of the above mentioned diseases/conditions.
  • the article of manufacture may comprise a container and a label or package insert on or associated with the container.
  • Suitable containers include, for example, bottles, vials or syringes.
  • the containers may be formed from a variety of materials such as glass or plastic.
  • the container holds a composition that may be effective for treating the condition and may have a sterile access port (e.g., the container may be an intravenous solution bag or a vial having a stopper pierceable by a hypodermic injection needle).
  • At least one active agent in the composition may be the antibody described herein.
  • the label or package insert may indicate that the composition may be used for treating the condition of choice.
  • the article of manufacture may comprise (a) a first container with a composition contained therein, wherein the composition comprises the antibody herein, and (b) a second container with a composition contained therein, wherein the composition comprises a therapeutic agent other than the antibody.
  • the article of manufacture in this embodiment of the disclosure may further comprise a package insert indicating that the first and second compositions can be used in combination.
  • Such therapeutic agent may be any of the adjunct therapies described in the preceding section (e.g., a thrombolytic agent, an anti-platelet agent, a chemotherapeutic agent, an anti-angiogenic agent, an anti-hormonal compound, a cardioprotectant, and/or a regulator of immune function in a mammal, including a cytokine).
  • the article of manufacture may further comprise a second (or third) container comprising a pharmaceutically acceptable buffer, such as bacteriostatic water for injection (BWFI), phosphate-buffered saline, Ringer's solution and dextrose solution. It may further include other materials desirable from a commercial and user standpoint, including other buffers, diluents, filters, needles, and syringes.
  • BWFI bacteriostatic water for injection
  • phosphate-buffered saline such as bacteriostatic water for injection (BWFI), phosphate-buffered saline, Ringer's solution and dextrose solution.
  • BWFI bacteriostatic water for injection
  • phosphate-buffered saline such as phosphate-buffered saline, Ringer's solution and dextrose solution.
  • dextrose solution such as bacteriostatic water for injection (BWFI), phosphate-buffered saline, Ringer's solution
  • kits comprising the antibody, the compositions or the immunoconjugates of the invention and instructions for use.
  • the kit can further contain one or more additional reagents, such as an immunosuppressive reagent, a cytotoxic agent or a radiotoxic agent, or one or more additional antibodies of the invention (e.g., an antibody having a complementary activity which binds to an epitope in the TrkA antigen distinct from the first antibody).
  • additional reagents such as an immunosuppressive reagent, a cytotoxic agent or a radiotoxic agent, or one or more additional antibodies of the invention (e.g., an antibody having a complementary activity which binds to an epitope in the TrkA antigen distinct from the first antibody).
  • the humanized anti-TrkA antibodies described herein represent a novel subgroup of anti-TrkA antibodies, which are highly effective at inhibiting the functional activation of TrkA.
  • the anti-TrkA mouse monoclonal antibody known as MNAC13 disclosed in WO00/73344, and its humanized variants disclosed in WO09/098238 do not achieve the same level of inhibition, which is desired under certain circumstances, e.g. for the treatment of pain and associated cancer pain, where a high level of inhibition can lead to increased efficacy of treatment.
  • Methods to assess antibody mediated inhibition of the functional activation of TrkA are well known in the art and include TF-1 cell proliferation assays, wherein the ability of antibodies to block cell surface TrkA/beta-NGF mediated cell proliferation is assayed using the factor dependent human erythro leukemic cell line TF-1 (Kitamura T et al,. (1989) J. Cellular Physiology 140(2):323-34).
  • Humanized antibodies disclosed in WO09/098238 have demonstrated inhibitory activity in TF-1 proliferation assays with the BXhVH5VLl candidate having the highest degree of inhibition amongst humanized variants of the MNAC13 mouse antibody.
  • humanized antibodies disclosed in WO09/098238 have demonstrated direct binding to the extracellular region of TrkA using Surface Plasmon Resonance (SPR) technique.
  • SPR Surface Plasmon Resonance
  • VH or VL indicates a heavy chain having IGHG1 isotype with a specific heavy chain variable domain denoted by a specific number e.g. BXhVH5, or a light chain having kappa isotype with a specific light chain variable domain denoted by a specific number e.g. BXhVLl, respectively.
  • BXhVH5VLl antibody refers to an antibody having BXhVH5 heavy chain and BXhVLl light chain.
  • Newly engineered humanized variants based on the selected subset of VH and VL variable domains described in WO09/098238 were termed "GBR" antibodies.
  • GBR Newly engineered humanized variants based on the selected subset of VH and VL variable domains described in WO09/098238 were termed "GBR" antibodies.
  • GBR Newly engineered humanized variants based on the selected subset of VH and VL variable domains described in WO09/098238
  • GBR Newly engineered humanized variants based on the selected subset of VH and VL variable domains described in WO09/098238
  • the same nomenclature system is in force with regard to the GBR antibodies.
  • Specific substitutions made within variable heavy chain domains as used herein are further indicated in brackets. All GBR antibodies were of IGHG1 heavy chain is
  • BXhVHlVLl having heavy chain variable domain VH1 (SEQ ID NO: 1) and light chain variable domain VL1 ( SEQ ID NO: 6
  • BXhVH3VLl having heavy chain variable domain VH3 (SEQ ID NO: 3) and light chain variable domain VL1
  • BXhVH3VL3 having heavy chain variable domain VH3 and light chain variable domain VL3
  • BXhVH5VLl having heavy chain variable domain VH5 (SEQ ID NO: 5) and light chain variable domain VL1
  • BXhVH5VL3 having heavy chain variable domain VH5 and light chain variable domain VL1.
  • MNAC13 mouse antibody as used herein has heavy chain variable domain MNAC13 VH with SEQ ID NO: 20, heavy chain with SEQ ID NO 21, and light chain with SEQ ID NO: 22.
  • the human IGHG4 isotype does not carry effector functions such as ADCC or CDC, and as such is a particularly well suited antibody format when effector functions are not required or detrimental to therapy.
  • one drawback of the naturally occurring human IGHG4 isotype is known as the "FAB exchange" phenomenon by which naturally occurring human IGHG4 antibodies are known to exchange heavy chains in vivo (Labrijn AF et al, (2009) Nat. Biotechnol. 27(8):767-71).
  • GBR VH5(V37A)VL1 and GBR VH5(K3Q,V37A)VL1 were both formatted as IGHG4 S228P immunoglobulins for the aforementioned therapeutic purpose and showed equivalent potency to their IGHG1 isotype counterpart, thereby making both GBR VH5(V37A)VL1 IGHG4 S228P and GBR VH5(K3Q,V37A)VL1 IGHG4 S228P antibodies suitable for human therapies wherein only TrkA blockade is required.
  • a 3D model for the VH5-VL1 pair of variable domains was calculated using the structure homo logy-modelling server SWISS-MODEL (Arnold K et al, (2006) Bioinformatics 22(2): 195-201; http://swissmodel.expasy.org) set in automated mode.
  • the retrieved model template was the experimentally solved MNAC13 FAB 3D structure (PDB code 1SEQ, www.pdb.org, Berman HM et al, (2000) Nucleic Acids Res. 28(l):235-42, Covaceuszach S et al, (2005) Proteins 58(3):717-27).
  • engineered antibodies had heavy chain variable domains encompassing substitutions at single or multiple positions in the aforementioned variable domain sequences (VH1, VH3, and VH5 with SEQ ID NO: 1, 3, 5, respectively) selected from the subset of positions described above. More specifically, engineered antibodies consisted of the substituted heavy chain variable domains as described above paired with one of the two previously mentioned light chain variable domains VL1 or VL3 (SEQ ID NO 6 and 8).
  • Engineered heavy chain variable domain coding DNA sequences were created by standard mutagenesis techniques using the vector DNAs for BXhVHl, BXhVH3, and BXhVH5 described in WO 09/098238 as PCR templates.
  • light chain variable domain cDNAs were directly amplified from the vector DNAs for BXhVLl and BXhVL3 described in WO 09/098238.
  • Variable domain cDNAs were further assembled upstream of their respective constant domain cDNA sequences using PCR assembly techniques.
  • the light chain specific vector allowed expression of kappa isotype light chains by ligation of the light chain variable domain cDNA of interest in front of the kappa light chain constant domain cDNA using BamHI and BsiWI restriction enzyme sites; while the heavy chain specific vector was engineered to allow ligation of the heavy chain variable domain cDNA of interest in front of the cDNA sequence encoding the IGHG1 CHI, IGHG1 hinge region, IGHG1 CH2, and IGHG1 CH3 constant domains using BamHI and Sail restriction enzyme sites.
  • secretion was driven by the murine VJ2C leader peptide containing the BamHI site.
  • the BsiWI restriction enzyme site is located in the kappa constant domain; whereas the Sail restriction enzyme site is found in the IGHG1 CHI domain.
  • IGHG4 immunoglobulin formatting having substitution S228P was achieved by replacing the cDNA sequence encoding the IGHG1 CHI , IGHG1 hinge region, IGHG1 CH2, and IGHG1 CH3 constant domains for a cDNA sequence encoding the IGHG4 CHI, IGHG4 hinge region having S228P substitution, IGHG4 CH2, and IGHG4 CH3 constant domains in the heavy chain specific vector described above.
  • Substitution S228P was introduced in a human IGHG4 heavy chain cDNA template by standard PCR mutagenesis techniques. Antibody production
  • antibodies are produced by further culturing the cells for a period of 4 to 5 days to allow for secretion into the culture medium (EX-CELL 293, HEK293-serum-free medium; Sigma, Buchs, Switzerland), supplemented with 0.1% pluronic acid, 4 mM glutamine, and 0.25 ⁇ g/ml geneticin).
  • EX-CELL 293, HEK293-serum-free medium Sigma, Buchs, Switzerland
  • Antibodies were purified from cell-free supernatant using recombinant protein-A streamline media (GE Healthcare Europe GmbH, Glattbrugg, Switzerland), and buffered exchanged into phosphate buffer saline prior to assays.
  • Calorimetric measurements were carried out on a VP-DSC differential scanning microcalorimeter (GE Healthcare Europe GmbH, Glattbrugg, Switzerland). The cell volume was 0.128 ml, the heating rate was 200°C/h, and the excess pressure was kept at 65 p.s.i. All antibodies were used at a concentration of 1 mg/ml in PBS (pH 7.4). The molar heat capacity of each protein was estimated by comparison with duplicate samples containing identical buffer from which the protein had been omitted. The partial molar heat capacities and melting curves were analyzed using standard procedures. Thermograms were baseline-corrected and concentration-normalized before being further analyzed using a Non-Two State model in the Origin software (v7.0, GE Healthcare Europe GmbH, Glattbrugg, Switzerland). Affinity measurements by SPR
  • the fusion protein consisted of the human TrkA extracellular region (SEQ ID NO: 25) fused to an IGHG1 Fc region wherein a protease-specific cleavage sequence was included between the two regions (TEV cleavage protease amino acid sequence: ENLYFQS).
  • TSV cleavage protease amino acid sequence: ENLYFQS TSV cleavage protease amino acid sequence: ENLYFQS.
  • the monovalent form of the human TrkA extracellular region was further purified to homogeneity using standard chromatographic techniques which included a protein-A step to remove Fc fragments.
  • the monovalent human TrkA extracellular region protein was buffer- exchanged into PBS before being diluted in Biacore running buffer for affinity measurements. Sample purity, homogeneity and molecular weight were confirmed by SDS-PAGE and size- exclusion analysis.
  • Antibodies were immobilized via capture of their Fc portion to allow for correct orientation on the sensor chip surface.
  • a monoclonal mouse anti-human IgG (Fc) antibody sensor chip was used to capture all humanized antibodies regardless of their isotypes (Human Antibody Capture Kit, catalogue number BR- 1008-39, GE Healthcare Europe GmbH), and a polyclonal rabbit anti-mouse immunoglobulin sensor chip was used to capture the MNAC13 mouse antibody (Mouse Antibody Capture Kit, catalogue number BR-1008-38, GE Healthcare Europe GmbH).
  • Suspension adapted TF-1 cells (ATCC ® number: CRL-2003) were grown in complete RPMI medium containing 10% FCS, and 5 ng/ml of rhuPNGF (recombinant human beta- NGF, R&D Systems Europe Ltd, Abingdon, UK).
  • rhuPNGF recombinant human beta- NGF, R&D Systems Europe Ltd, Abingdon, UK.
  • TF-1 cells were incubated for 5h in complete RPMI without human beta-NGF. Following this starvation step, cells were centrifuged and seeded in flat-bottomed 96 well plates with various concentrations of each antibody and a fixed concentration of recombinant beta-NGF.
  • TF-1 cells were seeded at a density of 7000 cells/well and the total antibody-cell mixture volume was 200 ⁇ with 5 ng/ml of human beta-NGF (final concentration). This mixture was incubated for 4 days at 37°C with C0 2 supplementation. At day 3, the colorimetric dye Alamar blue (AbD Serotec, Morphosys AbD GmbH, Dusseldorf, Germany) was added to each well without any change to the incubation conditions. At day 4, fluorescence was read on a Bio-Tek SynergyTM 2 spectrophotometer/microplate reader (BioTek Instruments GmbH, Luzern, Switzerland) with a 530-to-560 nm excitation wavelength and a 590 nm emission wavelength. Experiments were performed at least twice and measurements for each antibody concentration were done in triplicates.
  • VH1, VH3, and VH5 3D positions within the different heavy chain variable domains (VH1, VH3, and VH5) were selected for mouse-to -human as well as human-to-mouse mutations; this group consisted of positions: 3, 37, 40, 42, 44, 49, 50, 60, 62, 89, and 94 (Kabat numbering).
  • mice-to-human and human-to-mouse mutations were engineered in the BXhVH5VLl candidate in the context of an IGHG1 isotype format.
  • Production yields and affinities of some of the engineered anti-TrkA antibodies based on these single or combination of substitutions are shown in Table 1 and 2, respectively. Combining most or all mouse-to-human mutations led to poor production yields and complete loss of binding to TrkA.
  • mouse-to- human substitution A49S combined with Y50A induced a complete abrogation of binding, which was not rescued by the other mouse-to -human substitutions and mouse-to-human substitutions K3Q, T40A, R44G, and R94K did not lead to any improvement in affinity either alone or in combination with other mouse-to-human substitutions.
  • V37A substitution was introduced in the other selected candidates originating from WO 09/098238, and the affinities of the engineered anti-TrkA antibodies benchmarked against BXhVH5VLl and MNAC13 antibodies by SPR.
  • Table 3 and 4 show, respectively, the production yields and affinities of the V37A substituted antibodies.
  • engineered antibodies had differences in their production yields, all engineered antibodies exhibited an enhancement in affinity upon introduction of the V37A substitution. Affinities were improved by 8, 20, 30, and 55%, for BXhVFBVLl, BXhVHIVLl, BXhVH3VL3, and BXhVH5VL3, respectively.
  • the human-to-mouse substitution V37A did not only lead to an increase in affinity when introduced in BXhVH5VLl but, and most unexpectedly also led to an increase in affinity amongst all humanized variants.
  • GBR VH5(V37A)VL1 or GBR VH5(K3Q,V37A)VL1 both as IGHG1 or IGHG4 S228P isotype had affinities matching the affinity measured for the MNAC13 mouse antibody (Tables 2 and 4); both being increased by at least two-fold when compared to the BXhVH5VLl antibody (K D is decrease by 2.5 to 2.7 fold, respectively).
  • Table 3 and FIG.2 includes the melting temperatures of the FAB portions measured within the engineered antibodies.
  • Monoclonal antibodies melting profiles are characteristic of their isotypes but the mid-point melting temperature (Tm) of the FAB fragment can be easily identified even in the context of a full-length immunoglobulin (Garber E and Demarest SJ (2007) Biochem. Biophys. Res. Commun. 355(3):751-7).
  • Tm mid-point melting temperature
  • GBR VH5(V37A)VL1 and GBR VH5(K3Q,V37A)VL1 FAB fragments each displayed a single transition at 73.6 and 73°C respectively, both having equivalent thermo-stability with a shape and an amplitude of the FAB transition consistent with a cooperative unfolding which is generally observed for a compactly folded FAB fragment indicating that the engineering process was successful at retaining FAB stability.
  • This is further exemplified, when comparing the FAB Tm of GBR VH5(V37A)VL1 or GBR VH5(K3Q,V37A)VL1 with the MNAC13 FAB Tm (74°C), the differences being within 1°C or less.
  • GBR VH5(V37A)VL1 was the best performer followed by GBR VH5(K3Q, V37A)VL1, GBR VH5(K3Q,V37A)VL1 IGHG4 S228P, and GBR VH3(V37A)VL1 which had identical IC50s.
  • GBR VH3(V37A)VL3 and GBR VH5(V37A)VL3 antibodies had slightly higher IC50s but all tested engineered antibodies performed better than BXhVH5VLl, GBR VH5(V37A)VL1 being ten-fold better.
  • the GBR VH5(V37A)VL1 antibody also exhibited a three-fold potency increase in TF-1 cell proliferation assays when compared to the MNAC13 mouse antibody. All V37A variants had equivalent or better IC50 when compared to the mouse parental antibody.
  • Table 1 production yields from a selection of engineered anti-TrkA antibodies based on BXhVH5VLl; all antibodies are of the IGHGl isotype except as otherwise stated.
  • Table 3 production yields and FAB thermo-stability data from a selection of engineered anti- TrkA antibodies; all antibodies are of the IGHGl isotype.
  • Table 4 affinities of V37A engineered anti-TrkA antibodies; all antibodies are of the IGHGl isotype.
  • Anti-TrkA antibodies kocut(l/Ms) kof f (l/s) K D (nM)
  • Example 2 Humanized anti-TrkA antibody reverses acute inflammatory hyperalgesia of the paw induced by intraplantar injection of CFA.
  • CFA Complete Freunds adjuvant
  • Weight bearing through each hind limb was measured using an incapacitance tester (Linton Instruments, UK). Mice were placed in the incapacitance tester with the hind paws on separate sensors and the average force exerted by both hind limbs was recorded over 2 seconds. Naive AMBl mice were acclimatized to the procedure room in their home cages, with food and water available ad libitum. The AMBl mice were generated by replacement of exon 1 of mouse TrkA with that of its human counterpart and as such they exclusively express the human TrkA protein. Habituation to the incapacitance tester was performed over several days. Baseline weight bearing recordings were taken prior to induction of insult.
  • Inflammatory hyperalgesia was induced by intraplantar injection of CFA (20ul of 1.5mg/ml solution) into the left hind paw. A pre-treatment weight bearing reading was taken to assess hyperalgesia 23 hours post-CFA. Animals were then ranked and randomised according to CFA window in a Latin square design. 24 hours post-CFA, animals were treated with a single i.p. injection of O. lmg/kg isotype control i.p. or 0.0001, 0.001, 0.01 and O. lmg/kg of the antibody GBR VH5(K3Q,V37A)VL1 IGHG4 S228P i.p. (lOml/kg dose volume).
  • the nonselective NSAID indomethacin was used as a positive control at lOmg/kg p.o. (5ml/kg dose volume). Weight bearing readings were taken at 4, 8, 24, 48, 72, 96 and 120 hours post- antibody/drug treatment.
  • Example 3 Humanized anti-TrkA antibody reverses chronic inflammatory hyperalgesia of the knee joint induced by intra-articular injection of CFA
  • Intra-articular injection of CFA into one of the knee joints of the hind limbs of mice induces chronic inflammatory hyperalgesia that can be assessed using the weight bearing method.
  • the weight is re-distributed to lessen that placed on the limb with the injected (ipsilateral) joint and increase that on the limb with the non-injected (contralateral) joint.
  • the development of these signs in this animal model are believed to be clinically relevant as they reflect symptoms displayed by patients presenting chronic inflammatory joint pain associated with underlying conditions such as osteoarthritis and rheumatoid arthritis.
  • Naive AMBl mice were acclimatised to the procedure room in their home cages, with food and water available ad libitum.
  • the AMBl mice were generated by replacement of exon 1 of mouse TrkA with that of its human counterpart and as such they exclusively express the human TrkA protein.
  • Habituation to the incapacitance tester was performed. Baseline weight bearing readings were taken immediately prior to CFA injection. Animals were anaesthetised using isoflurane and oxygen mixed 3: 1 in sterile conditions. The knee area was shaved and cleaned with a dilute hibiscrub solution. The left knee was injected with ⁇ of lOmg/ml CFA. Animals were allowed to recover in a warmed environment before returning to their home cage.
  • weight bearing was measured at 4, 8, 24 and 96 hrs post-dosing.
  • weight bearing was measured at 1 and 8 hrs post-dosing on day 13 post-CFA and then at 1 hr post-dosing on days 14-17 post-CFA.
  • Behavioural assessments were performed blind. Weight bearing (g) readings were taken for both ipsilateral and contralateral hind limbs and expressed as % weight bearing difference (% ipsi/contra). Data were analysed by comparing treatment groups to isotype control group at each time point. Statistical analysis was performed as a repeated measures ANOVA followed by Planned comparison test using InVivoStat, (p ⁇ 0.05 considered significant).
  • Example 4 Humanized anti-TrkA antibody reverses chronic osteoarthritic hyperalgesia of the knee joint induced by intra-articular injection of MIA
  • MIA monosodium iodoacetate
  • Tramadol a weak ⁇ -opioid receptor agonist
  • tramadol does not produce gastrointestinal bleeding or renal problems nor does it affect articular cartilage.
  • tramadol was used alongside pregabalin as a comparator to the antibody GBR VH5(K3Q,V37A)VL1 IGHG4 S228P.
  • Naive AMBl mice were acclimatised to the procedure room in their home cages, with food and water available ad libitum.
  • the AMBl mice were generated by replacement of exon 1 of mouse TrkA with that of its human counterpart and as such they exclusively express the human TrkA protein.
  • Habituation to the incapacitance tester was performed over several days. Base line weight bearing readings measurements were taken. Animals were anaesthetised using isoflurane and oxygen mixed 3: 1 in sterile conditions. The knee area was shaved and cleaned with a dilute hibiscrub solution. MIA, 5 ⁇ 1 of lOOmg/ml (50( ⁇ g) or saline (sham) was injected into the knee joint of the left hind leg.
  • mice were allowed to recover in a warmed environment, before returning to their home cage.
  • animals were assessed at regular intervals using weight bearing for development of knee joint hyperalgesia.
  • weight bearing measurements were taken and animals were ranked and randomised to treatment groups according to their MIA response window followed by treatment with a single i.p. injection of 1 , 10, 100 ⁇ g/kg antibody GBR VH5(K3Q,V37A)VL 1 IGHG4 S228P or 100 ⁇ g/kg isotype control antibody (5ml/kg dose volume).
  • animals were treated on day 14 post-MIA with tramadol at lOmg/kg or pregabalin at 30mg/kg p.o. (5ml/kg dose volume) followed by tramadol at 30mg/kg or pregabalin at lOOmg/kg every other day on days 16-22 post-MIA (5ml/kg dose volume). All animals were assessed using weight bearing at 4, 8, and 24 hours post-dosing on day 14 post-MIA followed by every 24 hours for antibody treated groups and 1 and 24 hours post-dose for tramadol and pregabalin treated groups. Behavioural assessments were performed blind.
  • Weight bearing (g) readings were taken for both ipsilateral and contralateral hind limbs and expressed as % weight bearing difference (% ipsi/contra). Data were analysed by comparing treatment groups to sham group at each time point (day 3-14 post-MIA), for effects of MIA on hyperalgesia. Post-dosing data were analysed by comparing treatment groups to the isotype control group at each time point. Statistical analysis was performed as a repeated measures ANOVA followed by Planned comparison test using InVivoStat, (p ⁇ 0.05 considered significant).
  • the antibody GBR VH5 (K3Q,V37A) VL1 IGHG4 S228P at ⁇ g/kg had no effect on the hyperalgesia.
  • Tramadol (lOmg/kg) and pregabalin (30mg/kg) had no effect at 1 hour post-dosing on day 14 post-MIA, but showed significant reversal of hyperalgesia by 4 hours post-dose (Fig. 6B).
  • the anti-TrkA antibody can be administered at a much lower dose and with a lower dosing frequency than pregabalin and tramadol.
  • Example 5 Humanized anti-TrkA antibody reverses peripheral neuropathic hyperalgesia and allodynia induced by chronic constriction injury of the sciatic nerve
  • Peripheral neuropathic pain is a chronic form of pain arising from injury, dysfunction or disease of the peripheral nervous system. It typically comprises hyperalgesia (heightened response to painful stimuli) as well as allodynia (painful response to non-painful stimuli).
  • Neuropathic pain can be induced in animals by experimental injury of peripheral nerves and this is typically achieved by chronic constriction injury (CCI) of the sciatic nerve via loose ligation (Bennett & Xie (1998) Pain, 33: 87- 107). Since pregabalin is the recommended first- line drug in the treatment of neuropathic pain it is used here as a comparator compound.
  • CCI chronic constriction injury
  • AMB1 mice were generated by replacement of exon 1 of mouse TrkA with that of its human counterpart and as such they exclusively express the human TrkA protein.
  • the left sciatic nerve was exposed at mid-thigh level, aseptically.
  • Three loose ligature (using 4-0 black silk) were place around the sciatic nerve at 1-2 mm before the branching of nerve.
  • the surgical site was treated with betadine solution following skin suture. Animals were allowed to recover for the next seven days.
  • mice were acclimatized to the measuring device (cold plate) and the pre-dose paw withdrawal thresholds were recorded (Ohr). Animals were then regrouped into six groups. The following day, animals were treated with a single i.p. injection of isotype control antibody (1000 ⁇ g/kg/10ml) or different doses of the antibody GBR VH5(K3Q,V37A)VL1 IGHG4 S228P (10, 100 & 1000 Pregabalin (30 mg/kg/lOml, p.o.) or saline were administered once daily over a seven day post-dose period. Post-dose readouts were recorded at 4h, 24h and then every other day until the 7th day post- dose.
  • isotype control antibody 1000 ⁇ g/kg/10ml
  • GBR VH5(K3Q,V37A)VL1 IGHG4 S228P 10, 100 & 1000
  • Post-dose readouts were recorded lh after pregabalin or saline dosing on all days. Post- dose readouts were recorded first for mechanical hyperalgesia and 5 min later for cold allodynia. Mechanical hyperalgesia was measured as the threshold force (g) required to elicit paw withdrawal using a dynamic plantar aesthesiometer (Plantar Von Frey instrument; Ugo Basile Sri, Italy). Cold allodynia was measured as time taken for paw withdrawal from a cold plate in seconds (IITC Life Science Inc., USA).
  • mice Seven days after CCI surgery, mice exhibited marked mechanical hyperalgesia (Fig. 7A) and cold allodynia (Fig. 7B) as seen by a sharp reduction in their paw withdrawal threshold and paw withdrawal latency time, respectively. All doses of the antibody GBR VH5 (K3Q,V37A)VL1 IGHG4 S228P gave a reversal of mechanical hyperalgesia, as seen by an increase in the paw withdrawal threshold (Fig. 7A) and cold allodynia (Fig. 7B), as seen by an increase in the paw withdrawal latency.
  • NGF/TrkA signalling is critically required for the survival of sensory and sympathetic neurons during development (Bibel and Barde, 2000 Genes Dev 14(23):2919-37).
  • blockade of NGF by either active or passive immunization during the embryonic or neonatal periods results in substantial loss of these neurons along with pronounced atrophy of the neuronal cell body and associated neuronal structures such as the sympathetic ganglia (Cattaneo, (2013) Proc Natl Acad Sci U S A. 2013 Mar 26;110(13):4877- 85).
  • Neonatal mice were treated intraperitoneally (i.p.) once per week for 4 weeks from postnatal day 1 with lOOmg/kg GBR VH5 (K3Q,V37A) VL1 IGHG4 S228P, an anti-NGF based on the sequence of tanezumab which is fully cross-reactive to mouse NGF (Cattaneo, 2010 Curr Opin Mol Ther, 12, 94-106) or PBS.
  • mice were anesthetized with a mixture of Ketalar [(Parke- Davis) 75 mg/kg of body weight] and Xylazine [(Streuli) 10 mg/kg of body weight] and then perfused with 0.9% NaCl, followed by freshly prepared 4% paraformaldehyde in 0.1M phosphate buffer, pH 7.3.
  • the right superior cervical ganglion (SCG) in the neck was dissected, protected with foam in plastic embedding cassettes and post-fixed in the same fixative for 24 hours at 4°C.
  • the SCG was dissected with the surrounding arteries to allow for a better identification during the histological procedure. They were then embedded in paraffin (TCA 44-720 Medite automate) and cut with a Leica-microtome (3 ⁇ thin sections; Leica RM2135). Bands of consecutive sections were prepared and each 10th (adult) or 20th (neonate) section was mounted on "Superfrost plus" (Menzel) glass slides and stained with hematoxylin and eosin (H&E).
  • the slices were scanned with a Hamamatsu (Nanozoomer) slide scan system and were magnified with a 40x objective.
  • the pictures (.ndpi) were magnified in the NDP-Viewer2 software with either a lOx or 20x objective and then exported in jpeg format for the volume determination.
  • the volume of a ganglion was determined according to the Cavalieri method (Estimating Volume in Biological Structures; Cold Spring Harb Protoc 2012 Nov 1; 2012(11): 1129-39; doi: 10.1101/pdb.top071787).
  • the ImageJ software with appropriate plugins (“grid” and "cell counter") was used for the volume determination. Grid size was chosen to get approximately 100 points or more per ganglion.
  • volume of Ganglion number of points on ganglion tissue x grid area x thickness x distance between slices.
  • the mean volume of a cell was determined according to the nucleator principle (The nucleator, J Microsc. 1988 Jul; 151 (Pt 1):3-21).
  • the area (A) and the radius (R) of an object correlate with its volume (V), also for irregular objects like cells. Therefore, by measuring the area of a large number of cells, a very good approximation of the mean cell volume in a ganglion can be obtained.
  • the average of all volumes was taken as mean cell volume for the ganglion.
  • the cell number in one ganglion was calculated according to the formula: Total cell volume of ganglion mean cell volume in ganglion.
  • the cell density was calculated according to the formula: Number of Cells in Ganglion/Volume of Ganglion.
  • Example 7 Effects of an anti-TrkA antibody on sympathetic neuron survival during adulthood
  • NGF NGF-independent.
  • sympathetic neuronal survival becomes NGF-independent.
  • NGF continues in adulthood to modulate their morphology.
  • NGF blockade by either active or passive immunization leads to sympathetic neuronal cell body atrophy that overall contributes to an atrophy of the sympathetic ganglia (Levi-Montalcini and Booker, 1960b Proc Natl Acad Sci U S A. Mar;46(3):384-91; Angeletti et al, 1971a . Brain Res. Apr 2;27(2):343-55; Bjerre et al, 1975 Brain Res. Jul 11;92(2):257- 78; Goedert et al, 1978 Brain Res.
  • GBR VH5 (K3Q,V37A) VL1 IGHG4 S228P and tanezumab clearly exert opposing effects on sympathetic neurons in both neonate and adult animals, although the effects of GBR VH5 (K3Q,V37A) VL1 IGHG4 S228P are diminished in the latter.
  • Example 8 Effects of an anti-TrkA antibody on Peripheral neuropathic pain
  • Peripheral neuropathic pain is a chronic form of pain arising from injury/dysfunction/disease of the peripheral nervous system (Bridges et al, (2001) Br J Anaesth. ul;87(l): 12-26.). It typically comprises hyperalgesia (heightened response to painful stimuli) as well as allodynia (painful response to non-painful stimuli). Neuropathic pain can be induced in animals by experimental injury of peripheral nerves and this is typically achieved by chronic constriction injury (CCI) of the sciatic nerve via loose ligation.
  • CCI chronic constriction injury
  • GBR VH5 (K3Q,V37A) VL1 IGHG4 S228P clearly gave a higher reversal of mechanical hyperalgesia and cold allodynia than the anti-NGF antibody based on tanezumab.
  • Example 9 Effects of anti-TrkA antibody on abberant sensory and sympathetic nerve sprouting
  • Persistent knee joint inflammation induced by multiple intra-articular injections of Complete Freunds adjuvant (CFA) over several weeks in mice leads to ectopic sensory and sympathetic neuronal sprouting within the joint along with nociceptive behaviours (Ghilardi et al, 2012 Arthritis Rheum. Jul;64(7):2223-32).
  • CFA Complete Freunds adjuvant
  • Similar ectopic sprouting is also observed in mouse models of bone cancer pain and has been proposed to be involved in the maintenance of pain (Jimenez-Andrade et al, 2011 Pain. Nov;152(l l):2564-74).
  • PBS phosphate buffer solution
  • the mice were acclimatised to the environment for experiments for three days by training them to stand on the pads of an Incapacitance Meter (weight-bearing apparatus, Linton) for 2-3 min. The average values of weight load exerted by the left (ipsilateral) and right (contralateral) hind-paws over a period of 5 sec were noted. Three measurements were taken and the mean calculated for each time point.
  • Incapacitance Meter weight-bearing apparatus, Linton
  • mice received a total of 4 injections of CFA at days 0, 7, 14 and 21.
  • mice with a weight-bearing ratio lower than 70% were randomly divided into vehicle control group, 1 mg/kg GBR VH5 (K3Q,V37A) VL1 IGHG4 S228P group and 1 mg/kg tanezumab group. All vehicle/compounds were dosed i.p. at a single dose of 5 ml/kg.
  • Multiple intra- articular injections of CFA induced sustained hyperalgesia as detected by a shift in weight bearing from the ipsilateral to the contralateral hind paw resulting in a drop in the %ipsi/contra ratio to around 50% (Fig. 13). After a single i.p.
  • NSAID non-steroidal anti- inflammatory drugs
  • NSAID cyclooxygenase
  • PGs prostaglandins
  • PGE-2 has pleiotropic effects on bone, and COX-2 function is essential for bone healing (Blackwell et al.(2010) Trends Endocrinol Metab, 21, 294-301; Simon et al, (2002) J Bone Miner Res, 17, 963-76).
  • NGF nerve growth factor
  • TrkA neurothrophic tyrosine kinase receptor type 1
  • Blockade of this signalling pathway using an anti-NGF monoclonal antibody was shown to be effective for the treatment of chronic low back pain and arthrosis (Katz et al.,(2011) Pain, 152, 2248- 2258:, McKelvey et al, (2013) J Neurochem, 124, 276-89).
  • the binding of NGF to TrkA leads to receptor dimerization and activation by receptor auto-phosphorylation.
  • Ligand- receptor interaction can be abolished by the application of specific antibodies that selectively bind and neutralize NGF or by the application of small molecular weight inhibitors of the kinase activity of Trk receptors (Kumar and Mahal, (2012) J Pain Res, 5, 279-87: Watson et al, (2008) . Biodrugs, 22, 349-359).
  • the neutralizing anti-NGF monoclonal antibody is a human IgG2 based on the sequence of tanezumab and is fully cross-reactive to mouse NGF (Cattaneo, (2010) Curr Opin Mol Ther, 12, 94-106).
  • the neutralizing anti-TrkA antibody is GBR VH5 (K3Q,V37A) VL1 IGHG4 S228P.
  • mice Male AMB1 mice have a knock-in for human TrkA at the mouse TrkA locus and were obtained from Charles River (Charles River Italia, Calco, Italy). The mice were housed in groups of up to four animals with a 14-h light, 10-h dark cycle at 23°C and 55 ⁇ 10% humidity. Standard rodent chow and water were available ad libitum. Surgical procedures were conducted under sterile conditions.
  • mice Prior to surgery, the mice received a single dose of antibiotic (clindamycin-2- dihydrogenphosphate, 45 mgxkg -1 ; Clindamycin, Ratiopharm, Ulm, Germany).
  • antibiotic clindamycin-2- dihydrogenphosphate, 45 mgxkg -1 ; Clindamycin, Ratiopharm, Ulm, Germany.
  • tramadol-hydrochloride 25 mgxL -1 , Grunenthal, Aachen, Germany was provided via the drinking water 1 day before and after surgery.
  • the antibodies were administered intraperitoneally at a concentration of 10 mgxkg -1 bodyweight. The substances were applied on days 1, 6 and 11 after surgery.
  • mice were euthanized 7, 14 or 25 days after surgery, and the operated and contra- lateral femurs were explanted.
  • the activity of the mice after surgery and the vertical ground reaction force (GRF) of the operated limb was assessed, because pain leads to reduced activity and presumably reduced loading of the osteotomised limb.
  • the analyses were performed on days 2, 5, 7, 14 and 20 after surgery.
  • the mice were allowed to move freely through an acrylic glass tunnel containing a force plate in the floor (HE6x6, AMTI, Watertown, MA, USA).
  • the peak vertical GRF during the stance phase of the operated limb was recorded by a blinded observer and averaged from a minimum of four measurements of each mouse per time point.
  • the activity of the mice was recorded overnight using an infrared beam system fitted to specialized cages (ActiMot, TSE Systems GmbH, Bad Homburg, Germany).
  • the postoperative values were related to the preoperative measurement.
  • Flexural rigidity was calculated from the slope (k) of the linear region of the force-deflection curve. The distance between the load vector and the proximal (a) and distal (b) supports was considered when the fracture callus was not located exactly at the middle between the supports (1/2). Flexural rigidity (EI) was calculated according to the formula for asymmetrical bending: Micro-computed tomography ( u CT)
  • Femurs harvested on days 14 and 25 were scanned using a ⁇ CT device (Skyscan 1172, Skyscan, Kontich, Belgium) at a resolution of 8 ⁇ per pixel at a voltage of 50 kV and 200 ⁇ . Within each scan, two phantoms with a defined density of hydroxyapatite (HA) (250 and 750 mgxcm 3 ) were scanned to determine the bone mineral density (BMD).
  • HA hydroxyapatite
  • Femurs were fixed in 4% formalin for 24 h, decalcified in 20% ethylenediaminetetraacetic acid (pH 7.2-7.4) for 10-12 days and embedded in paraffin (Paraplast, Leica Biosystems, Wetzlar, Germany). Longitudinal sections of 6- ⁇ thickness were cut and stained using Safranin-0 and fast green. Evaluation of the tissue composition was performed using light microscopy (Leica DMI6000 B; Software MetaMorph ® , Leica Microsystems, Mannheim, Germany) under 50-fold magnification. At all time points, the whole callus, consisting of the periosteal callus and the osteotomy gap, was analysed for the relative amounts of fibrous tissue, cartilage and bone.
  • mice The activity and vertical GRF of the mice were assessed 1 to 2 days before and 2, 5, 7, 14 and 20 days following surgery. As expected, the activity declined following surgery in all groups.
  • the anti-TrkA-antibody-treated mice displayed significantly higher activity compared to PBS-treated animals (Fig. 14 A), indicating that the antibody-treated mice suffered less pain. There were no significant differences between PBS and anti-NGF-antibody or anti-NGF-antibody and anti-TrkA-antibody treatment. At later time points, no significant inter-group differences were detected (Figure 14A).
  • Fig. 15 A-I Histomorphometric assessment of the callus composition was performed on days 7, 14 and 25. Representative sections are depicted in Fig. 15 A-I. We did not detect any statistically significant differences in the callus composition between the treatment groups at any time point (Fig. 15 J-L). Three-dimensional assessment of the callus on day 14 using ⁇ CT showed no significant differences in callus size, BV, BV/TV or BMD (Table 6). The same was found on day 25 on analysing the osteotomy or the whole callus (Table 7). The flexural rigidity of the calli, determined using a non-destructive three-point bending test, was not significantly influenced by anti-NGF antibody or anti-TrkA antibody compared to PBS administration ( Figure 16).
  • Table 6 ⁇ CT-analysis of the fracture callus on day 14 after osteotomy.
  • BMD 288 ⁇ 14 269 ⁇ 17 264 ⁇ 17 TV, total callus volume; BV, bone volume; BV/TV, bone volume per callus volume; BMD, bone mineral density. Data is presented as the mean ⁇ SEM; n 7-8.
  • Table 7 ⁇ CT-analyses of the osteotomy and whole fracture calli of mice treated with PBS, anti-NGF antibody or anti-TrkA antibody after a healing period of 25 days.
  • NGF expression was demonstrated in proliferating, mature and hypertrophic chondrocytes and in osteoblasts near the ossification front. Although these findings imply a function for NGF/TrkA signalling during fracture healing, the exact role has not been elucidated. It was demonstrated that topical NGF application improved fracture healing in a rib fracture model in rats (Grills et al, (1997) J Orthop Res, 15, 235-42). Although the underlying mechanism has not been extensively investigated, the authors speculated that increased sympathetic innervation stimulated the differentiation of osteoprogenitor cells towards chondrocytes (Grills et al, (1997) J Orthop Res, 15, 235-42).
  • mice's activity and the GRF of the operated limb longitudinally were assessed for the analgesic effects of the antibody treatment.
  • Significantly higher activity was found for the anti-TrkA antibody-treated mice compared to the PBS-treated animals on day 2 following surgery.
  • the inventors have generated and tested a first low concentration liquid formulation of an anti-TrkA antibody adjusted to pH 6.0, comprising:
  • an anti-TrkA antibody comprising a light chain variable sequence comprising: SEQ ID NO: 5 and a heavy chain variable region comprising SEQ ID NO: 7;
  • the inventors have characterised the stability of the anti-TrkA antibody using a number of criteria at 5°C, 25°C and 40°C at time points (in months) TO, Tl, T2, T3, T6, T9, T12, T18, T24, T30.
  • the antibody present in the formulation was characterised at the relevant time point, by determining the clarity, degree of coloration, degree of opalescence and particulate contamination (visible particles) of the formulation or a portion thereof; light absorption measurement of wavelength 280nm to determine the concentration of protein present in the formulation; by SDS-page gel visualisation to determine changes in the weight and or breakdown of the antibody; by ELISA to determine any change in the binding properties of the antibody; by HPLC-CEX to determine changes in the positive/negative antibody species make up in the formulation; by HPLC-IEF so as to determine changes in the IsoElectroFocusing profile of the antibody by capillary electrophoresis present in the formulation, by HPLC-SEC analysis so as to determine changes in the the antibody in the formulation.
  • assessments of the properties of the formulation and the antibody therein were made using standard techniques. In each case comparison at least being made between one or more time points and the TO value and/or a previously established standard for the antibody used as a standard material for quality assurance and quality control purposes.
  • Stability data is provided for 5°C in figure 17, 25°C in figure 18 and 40°C in figure 19.
  • Example 12 Stability data for high concentration aqueous formulation
  • the inventors have generated and tested a liquid formulation of an anti-TrkA antibody adjusted to pH 5.75 or 6.0, comprising:
  • an anti-TrkA antibody comprising a light chain variable sequence comprising: SEQ ID NO: 5 and a heavy chain variable region comprising SEQ ID NO: 7;
  • the inventors have characterised the stability of the anti-TrkA antibody using a number of criteria at 5°C, 25°C and 40°C at time points (in months) TO, Tl, T2, T3, T6, T9, T12, T18, T24, T30.
  • the antibody present in the formulation was characterised at one or more time points by determining changes in the clarity, degree of coloration, degree of opalescence and particulate contamination (visible particles) of the formulation, light absorption measurement of wavelength 280nm to determine the concentration of protein present in the formulation; by SDS-page gel visualisation to determine changes in the weight and or breakdown of the antibody; by ELISA to determine any change in the binding properties of the antibody; by HPLC-CEX to determine changes in the positive/negative antibody species make up in the formulation; by HPLC-IEF so as to determine changes in the IsoElectroFocusing profile of the antibody by capillary electrophoresis present in the formulation, by HPLC-SEC analysis to determine changes in the antibody in the formulation.. In each case comparison between made between each time point of the TO value and a previously established standard for the antibody used as a standard material for quality assurance and quality control purposes.
  • Stability data is provided for the formulation adjusted to pH 5.75, at 5°C in figure 20, 25°C in figure 21 and 40°C in figure 22.
  • Stability data is provided for the formulation adjusted to pH 6, at 5°C in figure 23, 25°C in figure 24 and 40°C in figure 25.
  • Example 13 Stability data for high concentration aqueous formulation
  • the inventors have generated and tested a first low concentration liquid formulation of an anti-TrkA antibody comprising:
  • an anti-TrkA antibody comprising a light chain variable sequence comprising: SEQ ID NO: 5 and a heavy chain variable region comprising SEQ ID NO: 7;
  • the inventors have characterised the stability of the anti-TrkA antibody using a number of criteria at 5°C, 25°C and 40°C at time points (in months) TO, Tl, T2, T3, T6, T9, T12, T18, T24, T30.
  • the antibody present in the formulation was characterised at one or more time points by determining changes in the clarity, degree of coloration, degree of opalescence and particulate contamination (visible particles) of the formulation, light absorption measurement of wavelength 280nm to determine the concentration of protein present in the formulation; by SDS-page gel visualisation to determine changes in the weight and or breakdown of the antibody; by ELISA to determine any change in the binding properties of the antibody; by HPLC-CEX to determine changes in the positive/negative antibody species make up in the formulation; by HPLC-IEF so as to determine changes in the IsoElectroFocusing profile of the antibody by capillary electrophoresis present in the formulation, by HPLC-SEC analysis to determine changes in the antibody in the formulation.
  • Stability data is provided for the formulation adjusted to pH 5.75, at 5°C in figure 26, 25°C in figure 27 and 40°C in figure 28.
  • Stability data is provided for the formulation adjusted to pH 6, at 5°C in figure 29, 25°C in figure 30 and 40°C in figure 31.
  • Example 14 Sub-visible particle, viscocity and syringeability of lOOmg/ml and 150mg/ml formulations
  • Proteins usually aggregate from partially unfolded molecules, which can be part of the native state ensemble of molecules. Even though product formulations are developed to maximize and maintain the fraction of the protein molecules present in the native state, significant amounts of aggregates can form, especially over pharmaceutically-relevant time scales and under stress conditions. The levels and sizes of protein particles present in a given product can be changed by many factors relevant to commercial production of therapeutic proteins. It is known that large protein assemblies with repetitive arrays of antigens, in which the protein molecules have native conformation, are usually the most potent at inducing immune responses.
  • the standard test for sub-visible particulate analysis specifies that particulates >10 ⁇ in size are controlled at or below 6000 particles per ml and particles >25 ⁇ are limited to at or below 600 particles per ml.
  • Sub visible particle analysis was performed using HIAC system (Beckman Coulter Lifesciences) using the protocol provided by the manufacturer. Measurement is based on light obscuration and involves measurement of particles based on the geometry of the particles.
  • Viscoscity analysis was performed using a Haake Reostress 1 (Thermo Scientific) using the protocol provided by the manufacturer.
  • the 150mg/ml formulation A (50mM Histidine, 150mM NaCl, Tween 80 - 0.05 %, pH 5.75, 150 mg/ml) showed exceptional properties having a viscosity close to that of the tested lOOmg/ml formulations.
  • the second tested 150mg/ml formulation (50mM Histidine, 150mM NaCl, Tween 80 - 0.05 %, pH 6, 150 mg/ml) also showed good properties and better that comparable marketed 150mg/ml therapeutic antibody formulations.
  • Syringeability is a key-product performance parameter of any parenteral dosage form. This refers to the ability of an injectable therapeutic to pass easily through a hypodermic needle on transfer from a vial prior to an injection. Syringeability includes such factors as ease of withdrawal, clogging and foaming tendencies, and accuracy of dose measurements. Syringeability can be affected by the needle geometry, i.e. inner diameter, length, shape of the opening, as well as the surface finish of the syringe (4). This is of particular significance for self- injection devices, such as pens and auto -injectors, which are equipped with very thin needles. Indeed, patients can use pen injectors which employ 29-31-G needles.
  • Syringeability analysis was performed using a TA-XT Plus (Stable Micro Systems) using the protocol provided by the manufacturer.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • General Chemical & Material Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Animal Behavior & Ethology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Immunology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Rheumatology (AREA)
  • Biochemistry (AREA)
  • Biophysics (AREA)
  • Genetics & Genomics (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Physical Education & Sports Medicine (AREA)
  • Engineering & Computer Science (AREA)
  • Pain & Pain Management (AREA)
  • Orthopedic Medicine & Surgery (AREA)
  • Oncology (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Preparation Of Compounds By Using Micro-Organisms (AREA)
  • Medicinal Preparation (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Peptides Or Proteins (AREA)

Abstract

La présente invention concerne des anticorps dirigés contre le récepteur TrkA et leurs utilisations, y compris des anticorps anti-TrkA humanisés, et des méthodes de traitement avec des anticorps anti-TrkA.
EP15807645.5A 2014-12-05 2015-12-07 Anticorps anti-trka à propriétés inhibitrices améliorées et dérivés desdits anticorps destinés à être utilisés pour traiter les douleurs osseuses Withdrawn EP3227334A1 (fr)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US14/562,297 US20150183885A1 (en) 2012-06-08 2014-12-05 Anti-trka antibodies with enhanced inhibitory properties and derivatives thereof
EP15165107 2015-04-24
PCT/EP2015/078875 WO2016087677A1 (fr) 2014-12-05 2015-12-07 Anticorps anti-trka à propriétés inhibitrices améliorées et dérivés desdits anticorps destinés à être utilisés pour traiter les douleurs osseuses

Publications (1)

Publication Number Publication Date
EP3227334A1 true EP3227334A1 (fr) 2017-10-11

Family

ID=54838344

Family Applications (1)

Application Number Title Priority Date Filing Date
EP15807645.5A Withdrawn EP3227334A1 (fr) 2014-12-05 2015-12-07 Anticorps anti-trka à propriétés inhibitrices améliorées et dérivés desdits anticorps destinés à être utilisés pour traiter les douleurs osseuses

Country Status (7)

Country Link
EP (1) EP3227334A1 (fr)
JP (1) JP2018505853A (fr)
CN (1) CN107207599A (fr)
CA (1) CA2969940A1 (fr)
HK (1) HK1244012A1 (fr)
MA (1) MA41097A (fr)
WO (1) WO2016087677A1 (fr)

Families Citing this family (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2018094316A1 (fr) 2016-11-21 2018-05-24 Just Biotherapeutics, Inc. Formulations d'aflibercecept et leurs utilisations
EP3713591A1 (fr) * 2017-11-20 2020-09-30 Just-Evotec Biologics, Inc. Formulations d'aflibercept contenant un sel de lysine en tant qu'agent de tonicification et leurs utilisations
AR114110A1 (es) 2018-02-28 2020-07-22 Lilly Co Eli Anticuerpo anti-trka
JP2022533855A (ja) * 2019-05-30 2022-07-26 サンシャイン・レイク・ファーマ・カンパニー・リミテッド 抗TrkA抗体及びその使用
AR122141A1 (es) 2020-05-28 2022-08-17 Lilly Co Eli Inhibidor de trka
WO2023125490A1 (fr) * 2021-12-28 2023-07-06 4B Technologies (Suzhou) Limited Anticorps anti-trka et application de celui-ci

Family Cites Families (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
IT1306704B1 (it) * 1999-05-26 2001-10-02 Sirs Societa Italiana Per La R Anticorpi monoclonali e suoi derivati sintetici o biotecnologici ingrado di agire come molecole antagoniste per il ngf.
US7569364B2 (en) * 2002-12-24 2009-08-04 Pfizer Inc. Anti-NGF antibodies and methods using same
ITRM20030601A1 (it) * 2003-12-24 2005-06-25 Lay Line Genomics Spa Metodo per l'umanizzazione di anticorpi e anticorpi umanizzati con esso ottenuti.
PL3415535T3 (pl) * 2005-05-20 2021-06-14 Ablynx N.V. Ulepszone Nanociała TM do leczenia zaburzeń, w których pośredniczy agregacja
ITRM20050290A1 (it) * 2005-06-07 2006-12-08 Lay Line Genomics Spa Uso di molecole in grado di inibire il legame tra ngf e il suo recettore trka come analgesici ad effetto prolungato.
ITRM20050332A1 (it) * 2005-06-24 2006-12-25 Lay Line Genomics Spa Uso di molecole in grado di bloccare l'attivita' di trka per potenziare gli effetti di analgesici oppiacei sul dolore.
EA021284B1 (ru) * 2008-02-04 2015-05-29 Лей Лайн Дженомикс С.П.А. Антитела и их производные
JP6190723B2 (ja) * 2010-12-01 2017-08-30 アルダーバイオ ホールディングス エルエルシー 抗ngf組成物およびその使用
KR20150023679A (ko) * 2012-06-08 2015-03-05 그렌마크 파머수티칼스 에스. 아. 아미노산 치환을 갖는 인간화된 항-TrkA 항체

Also Published As

Publication number Publication date
CA2969940A1 (fr) 2016-06-09
HK1244012A1 (zh) 2018-07-27
MA41097A (fr) 2017-10-10
CN107207599A (zh) 2017-09-26
WO2016087677A1 (fr) 2016-06-09
JP2018505853A (ja) 2018-03-01

Similar Documents

Publication Publication Date Title
EP2859019B1 (fr) Anticorps humanisés anti-trka avec substitutions d'acides aminés
EP2731677B1 (fr) Anticorps qui se lient à l'ox40 et leurs utilisations
RU2473564C2 (ru) Антитела человека с высокой аффинностью к фактору роста нервов человека
US20200079863A1 (en) Antibodies that bind to tl1a and their uses
WO2016087677A1 (fr) Anticorps anti-trka à propriétés inhibitrices améliorées et dérivés desdits anticorps destinés à être utilisés pour traiter les douleurs osseuses
KR20190140472A (ko) 톨-유사 수용체 작용제를 포함하는 항체 접합체 및 병용 요법
KR102367723B1 (ko) 브라디키닌 b1 수용체 리간드에 대한 항체
BR112017019785B1 (pt) Anticorpo biespecífico, seu uso e composição farmacêutica
EP3099713B1 (fr) Protéine chimérique composee de ngf antagoniste domaine et un tnfa antagoniste domain
KR102570405B1 (ko) Il-7r 알파 서브유닛에 대한 항체 및 이의 용도
US20160319027A1 (en) Anti-trka antibodies with enhanced inhibitory properties and derivatives thereof
US20170362327A1 (en) Anti-trka antibodies with enhanced inhibitory properties and derivatives thereof for use in treating bone associated pain
Blein et al. Humanized anti-TrkA antibodies with amino acid substitutions.
WO2023125490A1 (fr) Anticorps anti-trka et application de celui-ci
OA17182A (en) Humanized anti-TrkA antibodies with amino acid substitutions.
WO2023250415A2 (fr) Anticorps ciblant l'intégrine alpha 11 bêta 1 et leurs utilisations
CA3195380A1 (fr) Composes et methodes de traitement de la douleur
NZ619849B2 (en) Antibodies that bind to ox40 and their uses

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 20170703

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

AX Request for extension of the european patent

Extension state: BA ME

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE APPLICATION IS DEEMED TO BE WITHDRAWN

18D Application deemed to be withdrawn

Effective date: 20180201