AU2015224416B2 - Osteoarthritis treatment - Google Patents

Osteoarthritis treatment Download PDF

Info

Publication number
AU2015224416B2
AU2015224416B2 AU2015224416A AU2015224416A AU2015224416B2 AU 2015224416 B2 AU2015224416 B2 AU 2015224416B2 AU 2015224416 A AU2015224416 A AU 2015224416A AU 2015224416 A AU2015224416 A AU 2015224416A AU 2015224416 B2 AU2015224416 B2 AU 2015224416B2
Authority
AU
Australia
Prior art keywords
csf
seq
antibody
aar
ser
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.)
Active
Application number
AU2015224416A
Other versions
AU2015224416A1 (en
Inventor
Andrew David Cook
John Allan Hamilton
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.)
University of Melbourne
Original Assignee
University of Melbourne
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 PCT/AU2009/001672 external-priority patent/WO2010071924A1/en
Application filed by University of Melbourne filed Critical University of Melbourne
Priority to AU2015224416A priority Critical patent/AU2015224416B2/en
Publication of AU2015224416A1 publication Critical patent/AU2015224416A1/en
Application granted granted Critical
Publication of AU2015224416B2 publication Critical patent/AU2015224416B2/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Abstract

H:\aar\Interwoven\NRPortbl\DCC\AAR\8378984_1 .DOC-04.09.2015 - 34 The present invention relates generally to a method for the treatment and/or prophylaxis of osteoarthritis (OA). In accordance with the present invention, an antagonist of GM-CSF can be effective in the treatment of osteoarthritis. An antagonist of GM-CSF includes, but is not limited to, an antibody that is specific for GM-CSF or the GM-CSF receptor. The present invention further provides transgenic animals, such as a GM-CSF knock-out mouse, useful for testing antagonists in certain disease models.

Description

H:\aar\lnterwoven\NRPortbl\DCC\AAR\8378984_1.DOC-04.09.2015 2015224416 09 Sep 2015 - 1 -
Osteoarthritis treatment
This application is a divisional of Australian Application No. 2009329814, the entire contents of which are incorporated herein by reference. 5 [0001] This application claims the benefit of U.S. Provisional Application No. 61/139,679, filed December 22, 2008, and U.S. Provisional Application No. 61/164,486, filed March 30, 2009, which are both incorporated by reference in their entireties.
FIELD OF THE INVENTION 10 [0002] The present invention relates generally to a method for the treatment and/or prophylaxis of osteoarthritis (OA). In accordance with the present invention, an antagonist of GM-CSF can be effective in the treatment of osteoarthritis. An antagonist of GM-CSF includes, but is not limited to, an antibody that is specific for GM-CSF or the GM-CSF 15 receptor. The present invention further provides transgenic animals, such as a GM-CSF knock-out mouse, useful for testing antagonists in certain disease models.
BACKGROUND OF THE INVENTION 20 [0003] Osteoarthritis (OA), also known as degenerative arthritis, is a disease most prevalent in the old and obese. OA is a disease of the articular joints, but, unlike rheumatoid arthritis (RA), the disease is not systemic, usually affecting only one or a few joints. The disease leads to total destruction of the articular cartilage, sclerosis of the underlying bonesT and osteophyte formation, resulting in loss of movement and pain. The 25 ultimate result is often the need for a total joint replacement.
[0004] OA affects about ~21 million people in the US, comprises 25% of all primary care physician visitsT and accounts for 50% of all NSAID (non steroidal anti inflammatory drugs) prescriptions. There is currently no treatment available which slows or halts 30 disease progressionT; today’s drugs merely treat the symptoms. The incidence and severity of the disease increase with age. By the age of 65, 80% of Americans show radiographic evidence of OA though only 60% of them will be symptomatic. 65% of all H:\aar\lnterwoven\NRPortbl\DCC\AAR\8378984_1.DOC-04.09.2015 2015224416 09 Sep 2015 -2- joint disease by the age of 65 are OA. In 2006, there were 735,000 OA-related US hospitalizations.
[0005] Current OA drugs treat the symptoms of OA rather than the disease itself. 5 Commonly used drugs in the treatment of OA include Non-steroidal anti-inflammatory drugs (NSAIDs), such as diacerin, voltaren, mobic and arthrotec (generic names: diclofenac, misoprostol, meloxicam). NSAIDs are mainly oral compounds which act by inhibiting prostaglandin synthesis in the central nervous system (CNS). Other commonly used drugs include non-narcotic analgesics, such as ultram (tramadol), COX-2 inhibitors, 10 such as celebrax and arcoxia (celecoxib, etoricoxib), narcotic analgesiscs, such as duragesic (dextropropoxyphene fentanyl), hyaluraonic acids, such as suparts, hyalgan, orthovisc and synvisc (Hylan G-F20), and corticosteroids, such as predinisolone and methyl predinisolone. Present treatments for OA intend to obviate the need for surgery through tissue engineering, such as chondrocyte transplantation; however, these 15 treatments are-only applicable for the treatment of last stage OA. Other approaches in the treatment of OA that are considered include prolotherapy, in which an irritant, such as dextrose, is injected into the affected joint, thereby causing an acute inflammatory reaction, but also strengthening and hopefully healing the tissues, ligaments, tendons, and cartilage. There is, thus, a high unmet medical need for the treatment of OA. 20 [0006] Some cytokines are known to be involved in osteoarthritis (Blom et al., Current Drug Targets (2008) 8:283). A few cytokines, such as IL-1, a ‘destructive’ cytokine, and the anabolic growth factor transforming growth factor β (TGFd β) are considered as potential drug targets. 25 [0007] Granulocyte macrophage colony-stimulating factor (GM-CSF) is a cytokine that functions as a white blood cell growth factor. GM-CSF stimulates stem cells to produce granulocytes (neutrophils, eosinophils, and basophils) and monocytes. Monocytes exit the circulation and migrate into tissue, whereupon they mature into macrophages. It is, thus, 30 part of the natural immune/inflammatory cascade, by which activation of a small number of macrophages can rapidly lead to an increase in their numbers, a process crucial for fighting infection. The active form of GM-CSF is found extracellularly as a homodimer. In particular, GM-CSF has been identified as an inflammatory mediator in autoimmune disorders, like rheumatoid arthritis (RA), leading to an increased production of pro- 2015224416 09 Sep 2015 H:\aar\lnlerwoven\NRPortbl\DCC\AAR\8378984_1.DOC-04.09-2015 -3- inflammatory cytokines, chemokines and proteases and, thereby, ultimately to articular destruction.
[0008] WO 06/0234412 discloses numerous biomarkers for osteoarthritis, which were 5 identified by protein microarrays. One of the biomarkers identified is GM-CSF, for which a four-fold up-regulation is reported in OA tissue. However, no indication or suggestion is provided that GM-CSF may also be a point for therapeutic intervention, and a mere fourfold up-regulation in OA tissue, as identified with the technology disclosed in WO 06/0234412, also does not suggest the same. In a related vein, Devalaraja et al 10 (US20020141994A1) cursorily mention OA among a long list of potentially suitable indications suitable for treatment with antagonists of colony stimulating factors. The list of indications includes atherosclerosis, sepsis, asthma, autoimmune disease, osteoporosis and rheumatoid arthritis. Besides other colony stimulating factors, such as M-CSF and G-CSF, GM-CSF is one of the colony stimulating factors mentioned in Devalaraja et al. 15 Indeed, Devalaraja et al. include no data or other insights as to why antagonizing GM-CSF would be appropriate to treat a subject suffering from OA. H:\aar\lnlerwoven\NRPortbl\DCC\AAR\8378984_1.DOC-04.09.2015 2015224416 09 Sep 2015 -4-
SUMMARY OF THE INVENTION
[0006] The present invention, for the first time, demonstrates that GM-CSF is a valid target for the treatment of OA. This finding is new, and the prior art does not teach, 5 suggest or provide any rational for such a point of intervention in the treatment of OA. Accordingly, the invention provides, e.g., a method for the treatment of osteoarthritis in a subject, said method comprising the step of administering an effective amount of a GM-CSF antagonist to said subject. 10 [0007] In another aspect, the present invention contemplates a method for the prophylaxis of osteoarthritis in a subject, said method comprising the step of administering an effective amount of GM-CSF antagonist to said subject.
[0008] In another aspect, the present invention is directed to a composition comprising a 15 GM-CSF antagonist capable of antagonizing the ability of GM-CSF from activating, proliferating, inducing growth and/or survival of cells in a subject suffering from osteoarthritis, or being suspected of suffering from osteoarthritis, said composition further comprising one or more pharmaceutically acceptable carriers and/or diluents. 20 [0009] In another aspect, the present invention is directed to a composition comprising a GM-CSF antagonist useful in the treatment of osteoarthritis, said composition further comprising one or more pharmaceutically acceptable carriers and/or diluents.
[0010] In particular aspects of the present invention, the GM-CSF antagonist is an 25 antibody specific for GM-CSF.
[0011] In alternative aspects of the present invention, the GM-CSF antagonist is an antibody specific for the GM-CSF receptor.
30 [0012] In other aspects, the present invention is directed to the use of a GM-CSF antagonist in the preparation of a medicament in the treatment of osteoarthritis.
[0013] In other aspects, the present invention provides GM-CSF antagonists for the treatment of osteoarthritis. 2015224416 09 Sep 2015 H:\aar\lnterwoven\NRPortbl\DCC\AAR\8378984_1.DOC-04.09.2015 - 5- [0014] Throughout this specification, unless the context requires otherwise, the words "comprise", “have” and “include” and their respective variations such as "comprises", "comprising", “has”, “having”, “includes” and “including” will be understood to imply the 5 inclusion of a stated element or integer or group of elements or integers but not the exclusion of any other element or integer or group of elements or integers. H:\aar\lnlerwoven\NRPortbl\DCC\AAR\8378984_1.DOC-04.09.2015 2015224416 09 Sep 2015 - 6-
BRIEF DESCRIPTION OF THE DRAWINGS
[0015] Figure 1 shows quantitative data for joint damage in different regions assessed by histological scoring. The experimental set-up and the scoring system are described in 5 Example 2. “Lat.“ stands for lateral, “Med.” stands for medial. Statistical analysis was performed via Mann-Whitney. The data are statistically significant for Lat. Femur (p=0.02), Lat Tibia (p=0.003), Med, Tibia (p=0.001), and over all regions (Mean; p=0.002).
[0016] Figure 2 shows exemplary histology sections of healthy control knees. 10 Magnification is 100x. No cartilage damage, osteophyte formation, synovitis or deformations can be seen. S = synovial lining, C = cartilage layer.
[0017] Figure 3 shows exemplary histology sections of the left knees of C57/BL6 mice in a model of collagenase-induced OA. Magnification is of the individual sections is indicated 15 in the Figures. The top row of pictures shows that cartilage damage, osteophyte formation and synovitis are evident. O = osteophyte. S = synovial lining. The bottom row of pictures shows that joint deformation is also present.
[0018] Figure 4 shows exemplary histology sections of the left knees of GM-CSF-/- mice 20 in a model of collagenase-induced OA. Magnification is of the individual sections is indicated in the Figures. As can be seen, the abnormalities and/or damages are much less severe compared to the C57/BL6 mice (see Figure 3) and are comparably to the healthy control mice (see Figure 2). O = osteophyte. S = synovial lining. 25 [0019] Figure 5 shows the knee joint histology scoring of the therapeutic treatment with a GM-CSF antibody in a mouse model of OA. Lat.=Lateral. Med.=Medial. Results are expressed as mean ± SEM. As can be seen mice treated with anti-GM-CSF antibody show less disease. 30 [0020] Figure 6 shows the result of an experiment assessing the hind limb weight distribution in an incapacitance meter. Data are significant (unpaired t-test) from day 27 post OA induction onwards, as indicated in the graph. H:\aar\lnterwoven\NRPortbl\DCC\AAR\8378984_1.DOC-04.09.2015 2015224416 09 Sep 2015 - 7-
DETAILED DESCRIPTION OF THE INVENTION
[0021] The present invention demonstrates that GM-CSF is a valid target for the treatment of OA. In this respect, the invention provides, in one aspect, methods of using a 5 GM-CSF antagonist to bring about a prophylactic or therapeutic benefit in the field of OA.
[0022] The present invention provides therapeutic methods comprising the administration of a therapeutically effective amount of a GM-CSF antagonist to a subject in need of such treatment. A "therapeutically effective amount11 or «effective amount”, as used herein, 10 refers to the amount of a GM-CSF antagonist necessary to elicit the desired biological response. In accordance with the subject invention, the therapeutic effective amount is the amount of a GM-CSF antagonist necessary to treat and/or prevent osteoarthritis.
[0023] “GM-CSF antagonists”, as used herein, includes GM-CSF antagonists in its 15 broadest sense; any molecule which inhibits the activity or function of GM-CSF, or which by any other way exerts a therapeutic effect on GM-CSF is included. The term GM-CSF antagonists includes, but is not limited to, antibodies specifically binding to GM-CSF, inhibitory nucleic acids specific for GM-CSF or small organic molecules specific for GM-CSF. Also within the meaning of the term GM-CSF antagonist are antibodies specifically 20 binding to the GM-CSF receptor, inhibitory nucleic acids specific for the GM-CSF receptor or small organic molecules specific for the GM-CSF receptor.
[0024] Inhibitory nucleic acids include, but are not limited to, antisense DNA, triplexforming oligonucleotides, external guide sequences, siRNA and microRNA. Useful 25 inhibitory nucleic acids include those that reduce the expression of RNA encoding GM-CSF by at least 20, 30, 40, 50, 60, 70, 80, 90 or 95 percent compared to controls. Inhibitory nucleic acids and methods of producing them are well known in the art. siRNA design software is available. 30 [0025] Small organic molecules (SMOLs) specific for GM-CSF or the GM-CSF receptor may be identified via natural product screening or screening of chemical libraries. Typically the molecular weight of SMOLs is below 500 Dalton, more typically from 160 to 480 Daltons. Other typical properties of SMOLs are one or more of the following: • The partition coefficient log P is in the range from -0.4 to +5.6 H:\aar\lnterwoven\NRPortbl\DCC\AAR\8378984_1.DOC-04.09.2015 2015224416 09 Sep 2015 - 8- • The molar refractivity is from 40 to 130 • The number of atoms is from 20 to 70 [0026] For reviews see Ghose et al, J Combin Chem: 7:55-68, 1999 and Lipinski et al, 5 Adv Drug Del Rev 23:3-25, 1997.
[0027] Preferably, a GM-CSF antagonist for use in the present invention is an antibody specific for GM-CSF or specific for the GM-CSF receptor. Such an antibody may be of any type, such as a murine, a rat, a chimeric, a humanized or a human antibody. A “human” 10 antibody or functional human antibody fragment is hereby defined as one that is not chimeric (e.g., not “humanized”) and not from (either in whole or in part) a non-human species. A human antibody or functional antibody fragment can be derived from a human or can be a synthetic human antibody. A “synthetic human antibody” is defined herein as an antibody having a sequence derived, in whole or in part, in silico from synthetic 15 sequences that are based on the analysis of known human antibody sequences. In silico design of a human antibody sequence or fragment thereof can be achieved, for example, by analyzing a database of human antibody or antibody fragment sequences and devising a polypeptide sequence utilizing the data obtained therefrom. Another example of a human antibody or functional antibody fragment is one that is encoded by a nucleic acid 20 isolated from a library of antibody sequences of human origin (/.e., such library being based on antibodies taken from a human natural source).
[0028] A “humanized antibody” or functional humanized antibody fragment is defined herein as one that is (i) derived from a non-human source (e.g., a transgenic mouse which 25 bears a heterologous immune system), which antibody is based on a human germline sequence; or (ii) chimeric, wherein the variable domain is derived from a non-human origin and the constant domain is derived from a human origin or (iii) CDR-grafted, wherein the CDRs of the variable domain are from a non-human origin, while one or more frameworks of the variable domain are of human origin and the constant domain (if any) is 30 of human origin.
[0029] The term "chimeric antibody" or functional chimeric antibody fragment is defined herein as an antibody molecule which has constant antibody regions derived from, or corresponding to, sequences found in one species and variable antibody regions derived H:\aar\ln1erwoven\NRPortbl\DCC\AAR\8378984_1.DOC-04.09.2015 2015224416 09 Sep 2015 -9- from another species. Preferably, the constant antibody regions are derived from, or corresponding to, sequences found in humans, e.g. in the human germ line or somatic cells, and the variable antibody regions (e.g. VH , VL , CDR or FR regions) are derived from sequences found in a non-human animal, e.g. a mouse, rat, rabbit or hamster. 5 [0030] As used herein, an antibody “binds specifically to”, “specifically binds to”, is “specific to/for” or “specifically recognizes” an antigen (here, GM-CSF or, alternatively, the GM-CSF receptor) if such antibody is able to discriminate between such antigen and one or more reference antigen(s), since binding specificity is not an absolute, but a relative 10 property. The reference antigen(s) may be one or more closely related antigen(s), which are used as reference points, e.g. IL3, IL5, IL-4, IL13 or M-CSF. In its most general form (and when no defined reference is mentioned), “specific binding” is referring to the ability of the antibody to discriminate between the antigen of interest and an unrelated antigen, as determined, for example, in accordance with one of the following methods. Such 15 methods comprise, but are not limited to Western blots, ELISA-, RIA-,ECL-, IRMA-tests and peptide scans. For example, a standard ELISA assay can be carried out. The scoring may be carried out by standard color development (e.g. secondary antibody with horseradish peroxide and tetramethyl benzidine with hydrogenperoxide). The reaction in certain wells is scored by the optical density, for example, at 450 nm. Typical background 20 (=negative reaction) may be 0.1 OD; typical positive reaction may be 1 OD. This means the difference positive/negative can be more than 10-fold. Typically, determination of binding specificity is performed by using not a single reference antigen, but a set of about three to five unrelated antigens, such as milk powder, BSA, transferrin or the like. Additionally, “specific binding” may relate to the ability of an antibody to discriminate 25 between different parts of its target antigen, e.g. different domains or regions of GM-CSF or the GM-CSF receptor, or between one or more key amino acid residues or stretches of amino acid residues of GM-CSF or the GM-CSF receptor.
[0031] Also, as used herein, an “immunoglobulin” (Ig) hereby is defined as a protein 30 belonging to the class IgG, IgM, IgE, IgA, or IgD (or any subclass thereof), and includes all conventionally known antibodies and functional fragments thereof. A “functional fragment” of an antibody/immunoglobulin hereby is defined as a fragment of an antibody/immunoglobulin (e.g., a variable region of an IgG) that retains the antigenbinding region. An “antigen-binding region” of an antibody typically is found in one or H:\aar\lnlerwoven\NRPortbl\DCC\AAR\8378984_1.DOC-04.09.2015 2015224416 09 Sep 2015 - 10- more hypervariable region(s) of an antibody, i.e., the CDR-1, -2, and/or -3 regions; however, the variable “framework” regions can also play an important role in antigen binding, such as by providing a scaffold for the CDRs. Preferably, the “antigen-binding region” comprises at least amino acid residues 4 to 103 of the variable light (VL) chain 5 and 5 to 109 of the variable heavy (VH) chain, more preferably amino acid residues 3 to 107 of VL and 4 to 111 of VH, and particularly preferred are the complete VL and VH chains (amino acid positions 1 to 109 of VL and 1 to 113 of VH; numbering according to WO 97/08320). A preferred class of immunoglobulins for use in the present invention is IgG. “Functional fragments” of the invention include the domain of a F(ab’)2 fragment, a 10 Fab fragment, scFv or constructs comprising single immunoglobulin variable domains or single domain antibody polypeptides, e.g. single heavy chain variable domains or single light chain variable domains. The F(ab’)2 or Fab may be engineered to minimize or completely remove the intermolecular disulphide interactions that occur between the Cm and CL domains. 15 [0032] An antibody of the invention may be derived from a recombinant antibody library that is based on amino acid sequences that have been designed in silico and encoded by nucleic acids that are synthetically created. In silico design of an antibody sequence is achieved, for example, by analyzing a database of human sequences and devising a 20 polypeptide sequence utilizing the data obtained therefrom. Methods for designing and obtaining in s/V/co-created sequences are described, for example, in Knappik et al, J. Mol. Biol. 296:57, 2000; Krebs et al, J. Immunol. Methods. 254.67, 2001; Rothe et al, J. Mol. Biol. 376:1182, 2008 and U.S. Patent No. 6,300,064 issued to Knappik et al 2000 supra, which hereby are incorporated by reference in their entirety. 25 [0033] Any antibody specific for GM-CSF may be used with the present invention. Exemplary antibodies are disclosed in US 11/914,599, which is incorporated by reference in its entirety. Other exemplary antibodies include antibodies comprising an amino acid sequence of a heavy chain variable region as depicted in SEQ ID NO:1 or an amino acid 30 sequence of a light chain variable region as depicted in SEQ ID NO:2. Yet other exemplary antibodies include antibodies which are derived from antibodies comprising a heavy chain variable region as depicted in SEQ ID NO:1 or an amino acid sequence of a light chain variable region as depicted in SEQ ID NO:2. Yet other exemplary antibodies include antibodies which have the same specificity and/or bind to the same epitope as 2015224416 09 Sep 2015 10 H:\aar\lnterwoven\NRPortbl\DCC\AAR\8378984_1.DOC-04.09.2015 - 11 - antibodies comprising a heavy chain variable region as depicted in SEQ ID NO:1 or an amino acid sequence of a light chain variable region as depicted in SEQ ID NO:2. Yet other exemplary antibodies include antibodies which comprise a heavy chain variable region which is at least 70 %, at least 80 %, at least 90 % or at least 95 % homologous to 5 the sequence depicted in SEQ ID NO:1. Yet other exemplary antibodies include antibodies which comprise a light chain variable region which is at least 70 %, at least 80 %, at least 90 % or at least 95 % homologous to the sequence depicted in SEQ ID NO:2. SEQ ID NO:1: Met Glu Leu lie Met Leu Phe Leu Leu Ser Gly Thr Ala Gly Val His Set Glu Val Gin Leu Gin Gin Ser Gly Pro Glu Leu Val Lys Pro Gly Ala Ser Val Lys lie Ser Cys Lys Ala Ser Gly Tyr Thr Phe Thr Asp Tyr Asn lie His Trp Val Lys Gin Ser His Gly Lys Ser Leu Asp Trp lie Gly Tyr He Ala Pro Tyr Ser Gly Gly Thr Gly Tyr Asn Gin Glu Phe Lys Asn Arg Ala Thr Leu Thr Val Asp Lys Ser Ser Ser Thr Ala Tyr Met Glu Leu Arg Ser Leu Thr Ser Asp Asp Ser Ala Val Tyr Tyr Cys Ala Arg Arg Asp Arg Phe Pro Tyr Tyr Phe Asp Tyr Trp Gly Gin Gly Thr Thr Leu Arg Val Ser Ser Val Ser Gly Ser 2015224416 09 Sep 2015 H:\aar\lnterwoven\NRPortbl\DCC\AAR\8378984_1.DOC-04.09.2015 - 12- SEQ ID NO:2: Met Gly Phe Lys Met Glu Ser Gin lie Gin Val Phe Val Tyr Met Leu Leu Trp Leu Ser Gly Val Asp Gly Asp lie Val Met He Gin Ser Gl n Lys Phe Val Ser Thr Ser Val Gly Asp Arg Val Asn lie Thr Cys Lys Ala Ser Gin Asn Val Gly Ser Asn Val Ala Trp Leu Gin Gin Lys Pro Gly Gin Ser Pro Lys Thr Leu lie Tyr Ser Ala Ser Tyr Arg Ser Gly Arg Val Pro Asp Arg Phe Thr Gly Ser Gly Ser Gly Thr Asp Phe lie Leu Thr He Thr Thr Val Gin Ser Glu Asp Leu Ala Glu Tyr Phe Cys Gin Gin Phe Asn Arg Ser Pro Leu Thr Phe Gly Ser Gly Thr Lys Leu Glu Leu Lys Arg Ala Asp Ala Ala Pro Thr Val Ser He Phe Pro Pro Ser Ser Lys Gly Glu Phe [0034] Alternative exemplary antibodies that can be used in the present invention are 5 antibodies comprising an amino acid sequence of a heavy chain variable region as depicted in SEQ ID NO:3 or an amino acid sequence of a light chain variable region as depicted in SEQ ID NO:4. Other exemplary antibodies include antibodies which are derived from antibodies comprising a heavy chain variable region as depicted in SEQ ID NO:3 or an amino acid sequence of a light chain variable region as depicted in SEQ ID 10 NO:4. Yet other exemplary antibodies include antibodies which have the same specificity and/or bind to the same epitope as antibodies comprising a heavy chain variable region as depicted in SEQ ID NO:3 or an amino acid sequence of a light chain variable region as depicted in SEQ ID NO:4. Yet other exemplary antibodies include antibodies which comprise a heavy chain variable region which is at least 70 %, at least 80 %, at least 90 % 15 or at least 95 % homologous to the sequence depicted in SEQ ID NO:3. Yet other exemplary antibodies include antibodies which comprise a light chain variable region which is at least 70 %, at least 80 %, at least 90 % or at least 95 % homologous to the sequence depicted in SEQ ID NO:4. 20 H:\aar\ln1erwoven\NRPortbl\DCC\AAR\8378984_1.DOC-04.09.2015 2015224416 09 Sep 2015 - 13-
SEQ ID NO. 3: heavy MOR
QVQLVESGGGLVQPGGSLRLSCAASGFTFSSYWMNWVRQAPGKGLEWVSGIENKYAGGA T YY AAS VKGRFTISRDNSKNTL YLQMNSLRAEDT AVYY CARGFGTDF WGQGTL VTV S S
SEQ ID NO. 4: light MOR
DIELTQPPSVSVAPGQTARISCSGDSIGKKYAYWYQQKPGQAPVLVIYKKRPSGIPERFSGSNS GNT ATLTISGTQAEDEAD YY CS AW GDKGMVF GGGTKLTVLGQ
[0035] Alternative exemplary antibodies that can be used in the present invention are antibodies comprising a H-CDR3 sequence selected from:
Ser Gly Leu lie Phe Asp Tyr Trp Leu Asp 15 10 (SEQ ID NO. 5),
Ser Gly Leu lie lie Asp Ala Leu Ser Pro 15 10 (SEQ ID NO:6),
Thr Ser Leu Met Ser lie Tyr Phe Asp Tyr 15 10 10 (SEQ ID NO:7),
Ser Gly Leu Leu Phe Leu Tyr Phe Asp Tyr 15 10 (SEQ ID NO:8),
Ser Gly Leu lie Asn Leu Gly Met His Pro 15 10 (SEQ ID NO:9),
Ser Gly Leu He Phe Asp Ala Leu Arg Asp 15 10 (SEQ ID NO:10),
Ser Gly Leu lie Phe Asp Lys Leu Thr Ser 15 10 (SEQ ID NO: 11),
Ser Gly Leu He Asn Leu His Phe Asp Thr 15 15 10 (SEQ ID NO: 12), H:\aar\lnterwoven\NRPorlbl\DCC\AAR\8378984_1.DOC-04.09.2015 2015224416 09 Sep 2015 - 14-
Ser Thr His Phe Ser Ala Tyr Phe Asp Tyr 15 10 (SEQ ID NO: 13),
Ser Gly Leu lie Met Asp Lys Leu Asp Asn 15 10 (SEQ ID NO: 14),
Ser Gly Leu lie lie Asp Asn Leu Asn Pro 15 10 (SEQ ID NO: 15), and
Ser Gly Leu lie Ala Val Tyr Phe Asp Tyr 15 10 5 (SEQ ID NO: 16).
[0036] Preferably, the antibodies comprising a H-CDR3 sequence selected from any one of SEQ ID NOs. 5-16, additionally comprise the following H-CDR1 sequence:
Asp Tyr Leu Leu His 1 5 (SEQ ID NO: 17), 10 and/or the following H-CDR2 sequence:
Trp Leu Asn Pro Tyr Ser Gly Asp Thr Asn Tyr Ala Gin Lys Phe Gin 15 10 15
Gly (SEQ ID NO: 18), and/or the following L-CDR1 sequence:
Arg Ala Ser Gin Asn lie Arg Asn lie Leu Asn 15 10 15 (SEQ ID NO: 19), and/or the following L-CDR2 sequence:
Ala Ala Ser Asn Leu Gin Ser 1 5 (SEQ ID NO:20), H:\aar\lnlerwoven\NRPortbl\DCC\AAR\8378984_1.DOC-04.09-2015 2015224416 09 Sep 2015 - 15- and/or the following L-CDR3 sequence:
Gin Gin Ser Tyr Ser Met Pro Arc Thr 1 5 (SEQ ID NO:21). 5 [0037] Alternative exemplary antibodies that can be used in the present invention are antibodies comprising the following L-CDR1 sequence:
Arg Ala Ser His Arg Val Ser Ser Asn Tyr Leu Ala 1 5 10 (SEQ ID NO:22), 10 and/or the following L-CDR2 sequence:
Gly Ala Ser Ash Arg Ala Thr 1 5 (SEQ ID NO:23), and/or the following L-CDR3 sequence:
Gin Gin Tyr Ala Ser Ser Pro Val Thr 15 1 5 (SEQ ID NO:24), and/or the following H-CDR1 sequence:
Gly Tyr lie Phe pro Thr Phe Ala Leu His 1 5 10 (SEQ ID NO:25), 20 and/or the following H-CDR2 sequence:
Ser He Asn Thr Ala ser Gly Lys Thr Lys Phe ser Thr Lys Phe Glti 1 5 10 15 (SEQ ID NO:26), 25 and/or the following H-CDR3 sequence:
Asp Arg Phe Gin Asn lie Met Ala Thr lie Leu Asp Val 1 5 10 (SEQ ID NO:27). H:\aar\lnterwoven\NRPortbl\DCC\AAR\8378984_1.DOC-04.09.2015 2015224416 09 Sep 2015 - 16- [0038] Preferably said antibody comprise all the CRDs of SEQ ID NOs. 22-27.
[0039] The GM-CSF receptor is a member of the haematopoietin receptor superfamily. It 5 is heterodimeric, consisting of an alpha and a beta subunit. The alpha subunit is highly specific for GM-CSF whereas the beta subunit is shared with other cytokine receptors, including IL3 and IL5. This is reflected in a broader tissue distribution of the beta receptor subunit. The alpha subunit, GM-CSFR a, is primarily expressed on myeloid cells and non-haematopoetic cells, such as neutrophils, macrophages, eosinophils, dendritic cells, 10 endothelial cells and respiratory epithelial cells. Full length GM-CSFR a is a 400 amino acid type I membrane glycoprotein that belongs to the type I cytokine receptor family, and consists of a 22 amino acid signal peptide (positions 1-22), a 298 amino acid extracellular domain (positions 23-320), a transmembrane domain from positions 321 - 345 and a short 55 amino acid intra-cellular domain. The signal peptide is cleaved to provide the mature 15 form of GM-CSFR a as a 378 amino acid protein. cDNA clones of the human and murine GM-CSFR a are available and, at the protein level, the receptor subunits have 36% identity. GM-CSF is able to bind with relatively low affinity to the a subunit alone (Kd 1-5 nM) but not at all to the β subunit alone. However, the presence of both a and β subunits results in a high affinity ligand-receptor complex (Kd » 100pM). GM-CSF signalling occurs 20 through its initial binding to the GM-CSFR a chain and then cross-linking with a larger subunit the common β chain to generate the high affinity interaction, which phosphorylates the JAK-STAT pathway.
[0040] Any antibody specific for GM-CSF receptor may be used with the present 25 invention. Exemplary antibodies include antibodies comprising an amino acid sequence of a H-CDR3 sequence depicted in any one of SEQ ID No’s.:28-46. Other exemplary antibodies include antibodies which are derived from antibodies comprising an amino acid sequence of a H-CDR3 sequence depicted in any one of SEQ ID No’s.:28-46. Yet other exemplary antibodies include antibodies which have the same specificity and/or bind to 30 the same epitope as antibodies comprising an amino acid sequence of a H-CDR3 sequence depicted in any one of SEQ ID No’s.:28-46. Yet other exemplary antibodies include antibodies which comprise a H-CDR3 sequence which is at least 70 %, at least 80 %, at least 90 % or at least 95 % homologous to the H-CDR3 sequence depicted in any one of SEQ ID No’s.:28-46. H:\aar\lnterwoven\NRPortbl\DCC\AAR\8378984_1.DOC-04.09.2015 2015224416 09 Sep 2015 SEQ ID No:28: Val Gly Ser Phe SEQ ID No:29: Val Gly Ser Phe 5 SEQ ID No:30: Val Gly Ser Phe SEQ ID No:31: Val Gly Ser Phe 10 SEQ ID No:32: Val Gly Ser Phe SEQ ID No:33: Val Gly Ser Phe SEQ ID No:34: Val Gly Ser Phe
Gly lie Ala Tyr Arg Pro 10
Gly Pro Ala Leu Arg Pro 10
Pro Pro Thr Tyr Gly Tyr 10
Gly Tyr Pro Tyr Arg Pro 10
Pro Leu Thr Leu Gly Leu 10
Gly Pro Val Tyr Gly Leu 10
Pro Pro Ala Tyr Arg Pro 10 15 H:\aar\lnlerwoven\NRPortbl\DCC\AAR\8378984_1.DOC-04.09.2015 2015224416 09 Sep 2015 - 18- SEQ ID No:35:
Val Gly Ser Phe Ser Pro Val Thr Tyr Gly Leu 5 10 SEQ ID No:36:
Val Gly Ser Phe Ser Gly Leu Ala Tyr Arg Pro 5 10 5 SEQ ID No:37:
Val Gly Ser Phe Ser Pro lie Thr Tyr Gly Leu 5 10 SEQ ID No:38:
Val Gly Ser Phe Ser Gly Trp Ala Phe Asp Tyr 5 10 SEQ ID No:39:
Val Gly Ser Phe Ser Gly Trp Ala Phe Asp Tyr 5 10 10 SEQ ID No:40:
Leu Gly Ser Val Thr Ala Trp Ala Phe Asp Tyr 5 10 SEQ ID No:41:
Ala Gly Ser lie Pro Gly Trp Ala Phe Asp Tyr 5 10 15 2015224416 09 Sep 2015 5 SEQ ID No:44: H:\aar\lnleiwoven\NRPortbl\DCC\AAR\8378984_1.DOC-04.09.2015 - 19- SEQ ID No:42:
Val Gly Ser Phe Ser Pro Leu Thr Met Gly Leu 5 10 SEQ ID No:43:
Val Gly Ser Phe Ser Pro Leu Thr Met Gly Leu 5 10
Val Gly Ser Phe Ser Gly Pro Ala Leu His Leu 5 10 SEQ ID No:45:
Val Gly Ser Val Ser Arg He Thr Tyr Gly Phe 5 10 SEQ ID No:46:
Val Gly Ser Phe Ser Pro Leu Thr Leu Gly Leu 5 10 10 [0041] In certain aspects, the present invention provides methods for the treatment of osteoarthritis in a subject, said method comprising the step of administering a GM-CSF antagonist to said subject. “Subject”, as used in this context refers to any mammal, 15 including rodents, such as mouse or rat, and primates, such as cynomolgus monkey (Macaca fascicularis), rhesus monkey (Macaca mulatta) or humans (Homo sapienss). Preferably the subject is a primate, most preferably a human.
[0042] In certain aspect, the present invention provides a composition comprising a GM-20 CSF antagonist capable of antagonizing the ability of GM-CSF from activating, proliferating, inducing growth and/or survival of cells in a subject suffering from H:\aar\lnterwoven\NRPortbl\DCC\AARV8378984_1.DOC-04.09.2015 2015224416 09 Sep 2015 -20- osteoarthritis, or being suspected of suffering from osteoarthritis, said composition further comprising one or more pharmaceutically acceptable carriers and/or diluents. Anti-GM-CSF antibodies of the present invention may antagonize any of the roles of GM-CSF in osteoarthritis. 5 [0043] In another aspect, the present invention provides a method for the prophylaxis of osteoarthritis in a subject, said method comprising administering a GM-CSF antagonist to said subject. “Prophylaxis” as used in this context refers to methods which aim to prevent the onset of a disease or which delay the onset of a disease. 10 [0044] In certain aspects, the present invention provides a composition comprising a GM-CSF antagonist useful in the treatment of osteoarthritis, said composition further comprising one or more pharmaceutically acceptable carriers and/or diluents. 15 [0045] In other aspects, the present invention provides the use of a GM-CSF antagonist in the preparation of a medicament in the treatment of osteoarthritis.
[0046] In other aspects, the present invention provides GM-CSF antagonists for the treatment of osteoarthritis. 20 [0047] The compositions of the present invention are preferably pharmaceutical compositions comprising a GM-CSF antagonist and a pharmaceutically acceptable carrier, diluent or excipient, for the treatment of osteoarthritis. Such carriers, diluents and excipients are well known in the art, and the skilled artisan will find a formulation and a 25 route of administration best suited to treat a subject with the GM-CSF antagonists of the present invention.
[0048] In another aspect the present invention provides a genetically engineered mammal having a GM-CSF -/- genotype. In particular aspects said mammal is a mouse. 30 The terms “knock-out” mouse (or mammal), a mouse (or mammal) “disrupted in” a certain gene, and a mouse (or mammal) with a “-/- genotype" are used interchangeably in the present invention and are art recognized. Respective animals are deficient in a respective gene, here GM-CSF, on both alleles of the chromosome. H:\aar\lnterwoven\NRPof1bl\DCC\AAR\8378984_1.DOC-04.09.2015 2015224416 09 Sep 2015 -21 -
Example 1:
Generation of a GM-CSF-/- mouse 5 [0049] The generation of GM-CSF-/- mice is described in Stanley et al (1994). Proc. Natl.
Acad. Sci. USA 91:5592. Briefly, chimeric mice were generated by microinjection of 129/OLA-derived ES cells (H-2b) with a disrupted GM-CSF gene into C57BL/6 (H-2b) host blastocysts. Germline transmitters of the mutated GM-CSF allele were crossed with C57BL/6 mice for 11 generations, giving GM-CSF+/- mice that were interbred to yield the 10 GM-CSF-/-, GM-CSF+/-, and GM-CSF+/+ mice used for the experiments. GM-CSF genotype status was determined by PCR analysis of tail DNA. Animals were fed standard rodent chow and water ad libitum and were housed with same sex littermates in sawdust-lined cages. Mice of both sexes were consigned to experiments at 8 to 15 wk of age 15 Example 2:
Validation of GM-CSF as a target for osteoarthritis [0050] GM-CSF-/- mice were compared to C57/BL6 mice (see e.g. Mills et al, J Immunol 164:6166-6173, 2000) in an experimental model of osteoarthritis. 20 [0051] Method: Mice (n=10 per group) received an intra-articular injection of collagenase in the leftknee on day -2 and day 0 (Blom et al, Arthritis Rheum 56:147-157, 2007). At day 42 the mice were killed, the knee joints collected, fixed, de-calcified, embedded in paraffin and cut at 7pm with a microtome. Slides were then stained with Safranin-O/Fast Green 25 and Haematoxylin and Eosin to demonstrate joint pathology. Pathology investigated includes: cartilage damage, synovitis, osteophyte formation and joint deformation.
[0052] The scoring system used for cartilage pathology was as follows: Grade 30 0 Normal 1 Irregular but intact 1.5 Irregular with rough surface 2 Superficial fibrillation 2.5 Superficial fibrillation with reduced cells in cartilage layer 2015224416 09 Sep 2015 H:\aar\lnlerwoven\NRPortbl\DCC\AAR\8378984_1.DOC-04.09-2015 -22- 5 3 Vertical fissures 3.5 Branching and/or horizontal fissures, tidemark ruptures 4 Cartilage loss not extending to the tide mark 4.5 Cartilage loss extending to the tide mark 5 Cartilage loss beyond the tide mark but not extending to the bone 5.5 Cartilage loss extending to the bone 6 Bone loss/remodeling/deformation 10
Stage 1 <10% area damaged 2 10-25% area damaged 3 25-50% area damaged 4 50-75% area damaged 15 [0053] The grade was multiplied by the stage to give the score.
[0054] This scoring system is based on a recognized method to assess OA histopathology in clinical and experimental OA. See Pritzker et al, Osteoarthritis Cartilage 14\ 13-29, 2006. Grade is defined as OA depth progression into cartilage. Stage is defined 20 as the horizontal extent of cartilage involvement, i.e. how much of the cartilage is affected. Grade is multiplied by the stage to give the score to give an overall score, so as to represent a combined assessment of OA severity and extent. Up to six sections are scored per mouse. 25 [0055] Results: Inspection of these joints showed that the GM-CSF-/- mice show less knee joint pathology than the control mice, indicating the role of GM-CSF in normal osteoarthritis pathology and progression. Pathology observed in the C57/BI6 mice includes severe damage to the cartilage layer, osteophyte formation, joint deformation and synovitis. The GM-CSF-/- mice showed no osteophyte formation or joint deformation and 30 much less cartilage damage and synovitis.
[0056] Quantitatve data on-joint damage in different regions are shown in Figure 1. Representative histology is shown in Figures 2 (healthy control knees), 3 (C57/BL6 left H:\aar\lnterwoven\NRPortbl\DCC\AAR\8378984_1.DOC-04.09.2015 2015224416 09 Sep 2015 -23- knees) and 4 (GM-CSF-/- left knees). GM-CSF gene -deficient mice developed less collagenase-induced OA pathology, compared to C57BL/6 mice.
[0057] In summary, GM-CSF-/- mice showed strongly decreased knee joint pathology 5 compared to C57/BL6 mice in an experimental model of osteoarthritis and validated GM- CSF as a drug target for therapeutic intervention for osteoarthritis.
Example 3:
Therapeutic effectiveness of GM-CSF antagonists in the treatment of OA 10 [0058] In this experiment we used a monoclonal antibody specific for GM-CSF to demonstrate that a GM-CSF antagonist can be effective to treat osteoarthritis.
Collagen-induced OA mouse model: 15 [0059] C57BL/6 mice were given 1 unit of collagenase type VII intra-articularly into the right knee on days 0 and 2 to induce joint instability (see Blom et al. (2004) Osteoarthritis Cartilage.12; 627-35).
Anti-GM-CSF antibody treatment: 20 [0060] 20 mice were randomly divided into 2 groups (10 mice/group).
Group 1 (n = 10): anti-GM-CSF antibody (22E9)
Group 2 (n = 10): lgG2a isotype control antibody.
[0061] Mice were treated intraperitoneally, three times per week for 6 weeks with 250 25 pg/mouse/treatment anti-GM-CSF antibody (22E9) or lgG2a isoptype control antibody.
Treatment started 4 days before the induction of OA (prophylactic), i.e. mice were treated on day -4, day -2, day 0 (the day of the first collagenase injection), then 3 times per week until the end of the experiment at 6 weeks). At weeks 2, 4 and 6, mice were bled. Serum will be checked for antibody content and immunogenicity against 22E9. Both, the control 30 antibody and the anti-GM-CSF antibody were purified to contain less than 10 Endotoxin Units/ml.
[0062] The antibody 22E9 was used as an exemplary anti-GM-CSF antibody. 22E9, which is of lgG2a isotype, is a rat anti-mouse GM-CSF-specific antibody. 22E9 was H:\aar\ln1erwoven\NRPortbl\DCC\AAR\8378984_1.DOC-04.09.2015 2015224416 09 Sep 2015 -24- purchased from AbD Serotec (Martinsried, Germany; Cat.No. 1023501). Alternative suppliers exist, e.g. eBioscience (SanDiego.CA, USA, Cat. No. 14-7331).
Histology: 5 [0063] 6- weeks post final injections, histology was performed on the mice knee joints.
The knee joints were collected, fixed, de-calcified, embedded in paraffin and cut at 7pm with a microtome. Slides were stained with Safranin-O/Fast Green and Haematoxylin and Eosin to demonstrate joint pathology. Pathology investigated included: cartilage damage, synovitis, osteophyte formation and joint deformation. 10 [0064] The same scoring system as in Example 2 was used for cartilage pathology.
Grade was multiplied by the stage to give the score.
[0065] The following scoring system was used for synovitis (Synovial layer scoring 15 system): 0 No changes compared to normal joints 1 Thickening of the synovial lining and some influx of inflammatory cells 2 Thickening of the synovial lining and intermediate influx of inflammatory 20 cells 3 Profound thickening of the synovial lining and maximal observed influx of inflammatory cells
Pain measurements: 25 [0066] An indicator of pain used for OA models is differential distribution of weight measured using an Incapacitance Meter. This instrument measures changes in weight distribution between the operated and contralateral, unoperated hind limb. Mice were allowed to acclimatize to the equipment on three occasions prior to the experiment. Weight placed on each hind limb was measured over a 5 second period. Three separate 30 measurements taken per mouse for each time point then averaged. Measurements were performed 2 times per week throughout the experiment. Results are expressed as collagenase injected limb/control limb x 100. H:\aar\lnterwoven\NRPortbl\DCC\AAR\8378984_1.DOC-04.09.2015 2015224416 09 Sep 2015 -25-
Results: [0067] For all areas analyzed in histology (except the Medial Femur), i.e. the Lateral Femur, the Lateral Tibia, and the Medial Tibia, there was a clear trend towards less disease in mice treated with anti-GM-CSF antibody. Results are depicted in Figure 5. 5 [0068] Assessment of the weight distribution, as a measure of pain associated with the arthritis, showed a significant shift in weight away from the arthritic knee from day 27 onwards in the anti-GM-CSF mAb-treated group compared to the control mAb-treated group. Results are depicted in Figure 6. 10 [0069] Mice treated with a GM-CSF antagonist showed less disease as compared to mice treated with the control antibody. Mice treated with the GM-CSF antagonist also showed significantly less pain in the latter stages of disease compared to mice treated with the control antibody. Mice treated with the isotype control antibody showed significant 15 increased signs of osteoarthritis as compared to the mice which received the GM-CSF-specific antibody. This demonstrates that GM-CSF antagonists are effective in the treatment of OA.
Example 4: 20 Therapeutic effectiveness of a GM-CSF specific antibody comprising SEQ ID NOs. 1 or 2 [0070] Example 3 is repeated, whereby as GM-CSF antagonist, a GM-CSF specific antibody comprising an amino acid sequence of a heavy chain variable region as depicted 25 in SEQ ID NO:1 or comprising an amino acid sequence of a light chain variable region as depicted in SEQ ID NO:2 is used. Another species than mouse may be used, in particular a species to which the antibody used in this experiment is cross reactive. Preferably the animal species used in this experiment is rat. 30 [0071] The animals treated with the isotype control antibody shows significant increased signs of osteoarthritis as compared to the animals which received a GM-CSF specific antibody comprising an amino acid sequence of a heavy chain variable region as depicted in SEQ ID NO:1 or comprising an amino acid sequence of a light chain variable region as H:\aar\lnterwoven\NRPortbl\DCC\AAR\8378984_1.DOC-04.09.2015 2015224416 09 Sep 2015 -26- depicted in SEQ ID NO:2. This demonstrates the effectiveness of the antibodies in the treatment of OA.
Example 5: 5 Therapeutic effectiveness of a GM-CSF specific antibody comprising SEQ ID NOs. 3 or 4 [0072] Example 3 is repeated. As GM-CSF antagonist, a GM-CSF specific antibody comprising an amino acid sequence of a heavy chain variable region as depicted in SEQ 10 ID NO:3 or comprising an amino acid sequence of a light chain variable region as depicted in SEQ ID NO:4 is used. Another species than mouse may be used, in particular a species to which the antibody used in this experiment is cross reactive. Preferably the animal species used in this experiment is rat. 15 [0073] The animals, e.g. rat, treated with the isotype control antibody show significant increased signs of osteoarthritis as compared to the animals which received a GM-CSF specific antibody comprising an amino acid sequence of a heavy chain variable region as depicted in SEQ ID NO:3 or comprising an amino acid sequence of a light chain variable region as depicted in SEQ ID NO:4. This demonstrates the effectiveness of the antibodies 20 in the treatment of OA.
Example 6:
Therapeutic effectiveness of a GM-CSF specific antibodies comprising SEQ ID NOs. 5-20 25 [0074] Example 3 is repeated. As GM-CSF antagonist, a GM-CSF specific antibody comprising a H-CDR3 sequence selected from any one of SEQ ID NOs. 5-16 is used. Preferably, said antibodies additionally comprise the H-CDR1 sequence of SEQ ID NO: 16, and/or the H-CDR2 sequence of SEQ ID NO: 17, and/or the L-CDR1 sequence of 30 SEQ ID NO: 18, and/or the L-CDR2 sequence of SEQ ID NO: 19), and/or the L-CDR3 sequence of SEQ ID NO:20. Another species than mouse may be used, in particular a species to which the antibody used in this experiment is cross reactive. Preferably the animal species used in this experiment is rat. 2015224416 09 Sep 2015 H:\aar\lnterwoven\NRPortbl\DCC\AAR\8378984_1.DOC-04.09.2015 - 27- [0075] The animals, e.g. rat, treated with the isotype control antibody show significant increased signs of osteoarthritis as compared to the animals which received a GM-CSF specific antibody according to the present example. This demonstrates the effectiveness of the antibodies in the treatment of OA.
Example 7:
Therapeutic effectiveness of a GM-CSF specific antibodies comprising SEQ ID NOs. 21-26 10 [0076] Example 3 is repeated. As GM-CSF antagonist, a GM-CSF specific antibody comprising the L-CDR1 sequence of SEQ ID NO:22, and/or the L-CDR2 sequence of SEQ ID NO:23, and/or the L-CDR3 sequence of SEQ ID NO:24, and/or the H-CDR1 sequence of SEQ ID NO:25, and/or the H-CDR2 sequence of SEQ ID NO:26, and/or the H-CDR3 sequence of SEQ ID NO:27 is used. Preferably said antibody comprise all the 15 CRDs of SEQ ID NOs. 22-27. Another species than mouse may be used, in particular a species to which the antibody used in this experiment is cross reactive. Preferably the animal species used in this experiment is rat.
[0077] The animals, e.g. rat, treated with the isotype control antibody show significant 20 increased signs of osteoarthritis as compared to the animals which received a GM-CSF specific antibody according to the present example. This demonstrates the effectiveness of the antibodies in the treatment of OA. H:\aar\lnlerwoven\NRPortbl\DCC\AAR\8378984_1.DOC-04.09-2015 2015224416 09 Sep 2015 -28-
Example 8:
Therapeutic effectiveness of antibodies specific for the GM-CSF receptor [0078] Example 3 is repeated with the difference that a monoclonal antibody specific for 5 the GM-CSF receptor is used instead of a monoclonal antibody specific for the GM-CSF.
[0079] As GM-CSF antagonist, a GM-CSF receptor specific antibody comprising an amino acid sequence of a H-CDR3 sequence depicted in any one of SEQ ID No’s.:27-45 is used. Another species than mouse may be used, in particular a species to which the 10 antibody used in this experiment is cross reactive. Preferably the animal species used in this experiment is rat.
[0080] The animals, e.g. rat, treated with the isotype control antibody show significant increased signs of osteoarthritis as compared to the animals which received a GM-CSF 15 receptor specific antibody according to the present example. This demonstrates the effectiveness of the antibodies in the treatment of OA.
Example 9:
Clinical trial 20 [0081] A clinical trial is performed in adult patients suffering from osteoarthritis of the knee The objective of the randomized, double-blind, placebo-controlled clinical trial is to determine the comparative differences between the GM-CSF antagonists of the present invention and placebo in overall pain relief and quality of life in a total sample of 30 25 patients with diagnosed osteoarthritis (OA) of the knee. Another objective is to determine the safety and tolerability of the GM-CSF antagonists of the present invention as determined by the adverse events, physical examination and vital signs.
Methods: 30 [0082] Thirty patients (about 15 adult males and 15 adult females), aged 40 and over, with a clinical diagnosis of osteoarthritis of the knee(s) and verified knee pain for at least 15 days in the month prior to testing are enrolled in the study. Patients receive a therapeutically effective amount of GM-CSF antagonists or a placebo (e.g. once every two weeks for about six months). H:\aar\lnterwoven\NRPortbl\DCC\AAR\8378984_1.DOC-04.09.2015 2015224416 09 Sep 2015 -29- [0083] The Western Ontario and McMaster Universities Osteoarthritis Index (WOMAC; Bellamy et al, J Rheumatol 15(12): 1833-40, 1988) and the SF-36v2 Quality of Life instrument scales (Quality Metric Health Outcomes Solutions, Lincoln, Rl) are used in the 5 study. The WOMAC is a disease-specific, self-administered, health status measure. It probes clinically-important symptoms in the areas of pain, stiffness and physical function in patients with osteoarthritis of the hip and/or knee. The index consists of 24 questions (5-pain, 2-stiffness and 17-physical function) and can be completed in less than 5 minutes. The WOMAC is a valid, reliable and sensitive instrument for the detection of 10 clinically important changes in health status following a variety of interventions (pharmacologic, nutritional, surgical, physiotherapy, etc.). The WOMAC questionnaire is valid for assessing the effects of intervention on hip or knee osteoarthritis. The SF-36v2 Quality of Life instrument is a multi-purpose, short-form health survey with 36 questions. It yields an 8-scale profile of functional health and well- being scores as well as 15 psychometrically-based physical and mental health summary measures and a preference-based health utility index. It is a generic measure, as opposed to one that targets a specific age, disease, or treatment group. Accordingly, the SF-36v2 has proven useful in surveys of general and specific populations, comparing the relative burden of diseases, and in differentiating the health benefits produced by a wide range of different treatments. 20 The SF-36v2 yields information on the following aspects and subsets of health; Physical Health (comprised of physical functioning, role-physical, bodily pain and general health) and Mental Health (comprised of vitality, social functioning, role- emotional and mental health). 25 Results: [0084] Change in bodily pain: The improvement in SF-36v2 bodily pain is statistically significant in patients treated with the GM-CSF antagonists of the present invention as compared with placebo. A higher score is better because it means the patient feels less pain after taking the product. There is a statistical significant improvement in the bodily- 30 pain score in the group that received the GM-CSF antagonists of the present invention versus the placebo group.
[0085] Change in role-phvsical score: The superior effect of the GM-CSF antagonists of the present invention compared with the placebo is statistically significant in week 8, week H:\aar\lnlerwoven\NRPortbl\DCC\AAR\8378984_1.DOC-04.09.2015 2015224416 09 Sep 2015 - 30- 12, and week 20 in terms of role limitations due to physical health (role physical). A higher score is better because it means that the patient noticed a physical improvement and a reduction in the limitations suffered in activities of daily living. There is a statistical significant improvement in the role-physical score in the group that received the GM-CSF 5 antagonists of the present invention versus the placebo group.
[0086] Change in the total WOMAC score: The total WOMAC score of the group treated with the GM-CSF antagonists of the present invention is statistical significantly better than the total WOMAC score of the placebo group (a lower score is better). 10
[0087] Change in WOMAC ADL: The improvement in activities of daily living (measured as a WOMAC ADL sub-score) is greater in the group treated with the GM-CSF antagonists of the present invention than in the placebo group. There is an statistically significant improvement in the WOMAC ADL score in the group treated with the GM-CSF 15 antagonists of the present invention compared to the placebo group (a lower score is better).
Conclusions: [0088] The clinical trial shows the efficacy of the GM-CSF antagonists of the present 20 invention in improving the quality of life of patients with osteoarthritis of the knee. The results of the clinical trial also show the product's safety and tolerance, given that no serious adverse effects were found.
[0089] The efficacy of the GM-CSF antagonists of the present invention can also be 25 established through studies in other species to which the GM-CSF antagonists of the present invention are cross-reactive (e.g. on horses in order to evaluate joint movement); and by using in vitro studies to determine the ability of GM-CSF antagonists of the present invention to inhibit IL-1 -induced agrecan degradation, conducting the assay on condrocyte cultures. 30 [0090] Those skilled in the art will appreciate that the invention described herein is susceptible to variations and modifications other than those specifically described. It is to be understood that the invention includes all such variations and modifications. The invention also includes all of the steps, features, compositions and compounds referred to 2015224416 09 Sep 2015 H:\aar\lnterwoven\NRPortbl\DCC\AAR\8378984_1.DOC-04.09.2015 -31 - or indicated in this specification, individually or collectively, and any and all combinations of any two or more of said steps or features.
[0091] Throughout this specification, unless the context requires otherwise, the word 5 "comprise", or variations such as "comprises" or "comprising", will be understood to imply the inclusion of a stated element or integer or method step or group of elements or integers or method steps but not the exclusion of any other element or integer or method steps or group of elements or integers or method steps. 10 [0092] The reference in this specification to any prior publication (or information derived from it), or to any matter which is known, is not, and should not be taken as an acknowledgement or admission or any form of suggestion that the prior publication (or information derived from it) or known matter forms part of the common general knowledge in the field of endeavour to which this specification relates. 2015224416 09 Sep 2015 H:\aar\lnterwoven\NRPortbl\DCC\AAR\8378984_1.DOC-04.09.2015 -32-
BIBLIOGRAPHY
Bellamy et al, J Rheumatol 15(12)-. 1833-40, 1988 Blom et al, Arthritis Rheum 56:147-157, 2007 Ghose etal, J Combin Chem: 1:55-68, 1999 Knappik et al, J. Mol. Biol. 296:57, 2000 Krebs et al, J. Immunol. Methods. 254:67, 2001 Lipinski etal, Adv Drug Del Rev 23:3-25, 1997 Mills etal, J Immunol 164:6166-6173, 2000 Pritzker etal, Osteoarthritis Cartilage 14:13-29, 2006 Rothe etal, J. Mol. Biol. 376:1182, 2008

Claims (4)

  1. CLAIMS:
    1. An antagonist of GM-CSF when used in the treatment of osteoarthritis, wherein the antagonist is an antibody specific for the GM-CSF receptor.
  2. 2. A composition comprising a GM-CSF antagonist when used in the treatment of osteoarthritis, wherein the antagonist is an antibody specific for the GM-CSF receptor and further wherein said composition further comprises one or more pharmaceutically acceptable carriers and/or diluents.
  3. 3. The antagonist of Claim 1 or composition of Claim 2, wherein the antibody is a chimeric, humanized or human antibody.
  4. 4. The antagonist or composition according to Claim 3, wherein the antibody is a humanized antibody.
AU2015224416A 2008-12-22 2015-09-09 Osteoarthritis treatment Active AU2015224416B2 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
AU2015224416A AU2015224416B2 (en) 2008-12-22 2015-09-09 Osteoarthritis treatment

Applications Claiming Priority (7)

Application Number Priority Date Filing Date Title
US13967908P 2008-12-22 2008-12-22
US61/139,679 2008-12-22
US16448609P 2009-03-30 2009-03-30
US61/164,486 2009-03-30
PCT/AU2009/001672 WO2010071924A1 (en) 2008-12-22 2009-12-21 Osteoarthritis treatment
AU2009329814A AU2009329814B2 (en) 2008-12-22 2009-12-21 Osteoarthritis treatment
AU2015224416A AU2015224416B2 (en) 2008-12-22 2015-09-09 Osteoarthritis treatment

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
AU2009329814A Division AU2009329814B2 (en) 2008-12-22 2009-12-21 Osteoarthritis treatment

Publications (2)

Publication Number Publication Date
AU2015224416A1 AU2015224416A1 (en) 2015-10-01
AU2015224416B2 true AU2015224416B2 (en) 2017-04-20

Family

ID=54251797

Family Applications (1)

Application Number Title Priority Date Filing Date
AU2015224416A Active AU2015224416B2 (en) 2008-12-22 2015-09-09 Osteoarthritis treatment

Country Status (1)

Country Link
AU (1) AU2015224416B2 (en)

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2007110631A1 (en) * 2006-03-27 2007-10-04 Medimmune Limited Binding member for gm-csf receptor
WO2008064321A2 (en) * 2006-11-21 2008-05-29 Kalobios Pharmaceuticals, Inc. Methods of treating chronic inflammatory diseases using a gm-csf antagonist

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2007110631A1 (en) * 2006-03-27 2007-10-04 Medimmune Limited Binding member for gm-csf receptor
WO2008064321A2 (en) * 2006-11-21 2008-05-29 Kalobios Pharmaceuticals, Inc. Methods of treating chronic inflammatory diseases using a gm-csf antagonist

Also Published As

Publication number Publication date
AU2015224416A1 (en) 2015-10-01

Similar Documents

Publication Publication Date Title
US20200247895A1 (en) Osteoarthritis treatment
US9834599B2 (en) Pain treatment
KR20170016501A (en) Methods of treating conditions with antibodies that bind colony stimulating factor 1 receptor (csf1r)
AU2015224416B2 (en) Osteoarthritis treatment

Legal Events

Date Code Title Description
FGA Letters patent sealed or granted (standard patent)