WO2017134292A1 - Anti-ox40 antagonistic antibodies for the treatment of atopic dermatitis - Google Patents

Anti-ox40 antagonistic antibodies for the treatment of atopic dermatitis Download PDF

Info

Publication number
WO2017134292A1
WO2017134292A1 PCT/EP2017/052504 EP2017052504W WO2017134292A1 WO 2017134292 A1 WO2017134292 A1 WO 2017134292A1 EP 2017052504 W EP2017052504 W EP 2017052504W WO 2017134292 A1 WO2017134292 A1 WO 2017134292A1
Authority
WO
WIPO (PCT)
Prior art keywords
amino acid
antibody
seq
acid sequence
heavy chain
Prior art date
Application number
PCT/EP2017/052504
Other languages
French (fr)
Inventor
Sachin Kumar DUBEY
Jonathan Albert BACK
Original Assignee
Glenmark Pharmaceuticals S.A.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Glenmark Pharmaceuticals S.A. filed Critical Glenmark Pharmaceuticals S.A.
Publication of WO2017134292A1 publication Critical patent/WO2017134292A1/en

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
    • C07K16/2878Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the NGF-receptor/TNF-receptor superfamily, e.g. CD27, CD30, CD40, CD95
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/39591Stabilisation, fragmentation
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0019Injectable compositions; Intramuscular, intravenous, arterial, subcutaneous administration; Compositions to be administered through the skin in an invasive manner
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • 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
    • C07K2317/70Immunoglobulins 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

Definitions

  • the present invention relates to anti OX40 antagonist antibody for the treatment of atopic dermatitis, including moderate-to-severe Atopic dermatitis.
  • the present application also relates to a method of treating Atopic dermatitis by the parenteral administration of an anti OX40 antagonist antibody to a patient in need thereof.
  • the present invention also relates to a pharmaceutical composition comprising an anti OX40 antagonist antibody and processes for preparing such a pharmaceutical composition.
  • Atopic Dermatitis is one of the most prevalent autoimmune disorders worldwide that affects around 4-7% of adults and 15-25% of children with a significant impact on the quality of patient life. It is a chronic, relapsing inflammatory skin disease characterized by intense pruritus (e.g., itchiness) and by scaly and dry eczematous lesions and often associated with other atopic disorders, such as allergic rhinitis and asthma.
  • specific therapies for AD are still limited, and the most commonly used therapies are not based on a mechanistic understanding of the disease.
  • OX40 is a member of the TNFR-superfamily of receptors and was first identified in 1987 as a 50 kDa glycoprotein expressed on activated CD4+ T cells from the rat (Paterson DJ et al., (1987) Mol. Immunol. 24: 1281-90).
  • the extracellular ligand binding domain of OX40 is composed of 3 full cysteine-rich domains (CRDs) and a partial, fourth C-terminal CRD (Bodmer JL et al., (2002) Trends Biochem. Sci. 27: 19-26).
  • the ligand for OX40 is OX40L (CD252) and 3 copies of OX40 bind to the trimeric ligand to form the OX40-OX40L complex (Compaan DM & Hymowitz SG (2006) Structure, 14: 1321-1330).
  • OX40 is a membrane -bound receptor; however a soluble isoform has also been detected (Taylor L & Schwarz H (2001) J. Immunol. Methods, 255: 67-72).
  • OX40 is not constitutively expressed on naive T cells but is induced after engagement of the T-Cell Receptor (TCR).
  • TCR T-Cell Receptor
  • OX40 is a secondary costimulatory molecule, expressed after 24 to 72 hours following activation; its ligand, OX40L, is also not expressed on resting antigen presenting cells, but is following their activation.
  • OX40 is expressed mainly by activated CD4+ T cells and to a limited extent, by activated CD8+ T cells (Salek-Ardakani S et al., (2006) Curr. Immunol. Rev.
  • the present invention provides an anti-OX40 antagonist antibody for use in the treatment or prevention of atopic dermatitis, including moderate -to-severe atopic dermatitis.
  • the anti-OX40 antagonist antibody comprises: a heavy chain CDR1 comprising the amino acid sequence of SEQ ID NO: 1 , and/or a heavy chain CDR2 comprising the amino acid sequence of SEQ ID NO: 2, and/or a heavy chain CDR3 comprising the amino acid sequence of SEQ ID NO: 3; and/or comprising a light chain CDR1 comprising the amino acid sequence of SEQ ID NO: 4, and/or a light chain CDR2 comprising the amino acid sequence of SEQ ID NO: 5 and/or a light chain CDR3 comprising the amino acid sequence of SEQ ID NO: 6.
  • the anti-OX40 antagonist antibody comprises: a heavy chain sequence comprising amino acid sequence of SEQ ID NO: 7 or 8 and light chain sequence comprising the amino acid sequence of SEQ ID NO: 9.
  • the present invention provides uses, compositions or methods which cause an improvement in one or more AD efficacy parameter(s) in a patient.
  • Improvements in AD related efficacy parameters include, for instance a decrease in Investigator's Global Assessment (IGA) score; a decrease in Body Surface Area (BSA) score; a decrease in Eczema Area and Severity Index (EASI) score; a decrease in SCORAD score; a decrease in Pruritus Numeric Rating Scale (NRS) score, and/or improvement of Dermatology Life quality index( DLQI).
  • AD efficacy parameter also may mean a change in the transcriptome or in a change in the cyto/chemokine profile of the patient or a biopsy from the site of AD on the patient.
  • the present invention provides a method for treating or preventing AD in a subject, the methods comprising: (a) selecting a subject who exhibits an abberant level of at least one AD-associated biomarker or is diagnosed clinically with AD; and (b) administering to the subject a pharmaceutical composition comprising a therapeutically effective amount of an anti-OX 40 antagonist antibody.
  • said AD-associated biomarker is selected from the group, Exe but are not limited to, Serum Thymus and activation-regulated chemokine (TARC/CCL17), eotaxin-3, total Immunoglobulin E (IgE), Immunohistochemistry and trans -epidermal water loss (TEWL).
  • the present application also relates to a method of treating Atopic dermatitis in a subject by the parenteral administration to a patient in need of a therapeutically effective amount of an anti OX40 antagonist antibody.
  • the invention provides a method of treating of atopic dermatitis comprising administering to a subject a composition comprising a therapeutically effective amount of an anti OX40 antagonist antibody and a pharmaceutically acceptable carrier.
  • the invention provides a method of treating atopic dermatitis comprising administering to a subject a therapeutically effective amount of anti OX40 antagonist comprising a heavy chain CDR1 comprising the amino acid sequence of SEQ ID NO: 1 , and/or a heavy chain CDR2 comprising the amino acid sequence of SEQ ID NO: 2, and/or a heavy chain CDR3 comprising the amino acid sequence of SEQ ID NO: 3; and/or comprising a light chain CDR1 comprising the amino acid sequence of SEQ ID NO: 4, and/or a light chain CDR2 comprising the amino acid sequence of SEQ ID NO: 5 and/or a light chain CDR3 comprising the amino acid sequence of SEQ ID NO: 6.
  • the present invention relates to a method of treating atopic dermatitis comprising administering to a subject a therapeutically effective amount of anti OX40 antagonist comprising a heavy chain sequence comprising amino acid sequence of SEQ ID NO: 7 or 8 and light chain sequence comprising the amino acid sequence of SEQ ID NO: 9.
  • the anti OX40 antagonist antibody dosage ranges from 0.0001 to 100 mg kg, and more preferably between 0.01 to 10 mg/kg, of the host body weight.
  • the dose is selected from the group comprising 0.001 , 0.002, 0.003, 0.004, 0.005, 0.006, 0.007, 0.008, 0.009, 0.01 , 0.01 , 0.02, 0.03, 0.04, 0.05, 0.06, 0.07, 0.08, 0.09, 0.1 , 0.2, 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, 1 , 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 45, 50, 60, 70, 80, 90, 100 mg/kg.
  • this relates to fixed dose preparations of 100, 200, 300, 400, 500, 600, 700, 800, 900 mg per dose. In accordance with another aspect of the present invention this relates to a dose administered to a patient at an interval of daily, weekly biweekly, monthly, bimonthly or any suitable interval in between.
  • the anti-OX40 antagonist antibody comprises: a heavy chain CDR1 comprising the amino acid sequence of SEQ ID NO: 1 , and/or a heavy chain CDR2 comprising the amino acid sequence of SEQ ID NO: 2, and/or a heavy chain CDR3 comprising the amino acid sequence of SEQ ID NO: 3; and/or comprising a light chain CDR1 comprising the amino acid sequence of SEQ ID NO: 4, and/or a light chain CDR2 comprising the amino acid sequence of SEQ ID NO: 5 and/or a light chain CDR3 comprising the amino acid sequence of SEQ ID NO: 6.
  • the anti-OX40 antagonist antibody comprises: a heavy chain sequence comprising amino acid sequence of SEQ ID NO: 7 or 8 and light chain sequence comprising the amino acid sequence of SEQ ID NO: 9.
  • the present invention relates to a method of treating atopic dermatitis in a patient said method comprising the parental administration to the subject of 10 mg/kg of an anti OX40 antagonist antibody and its acceptable carrier, two doses administered at an interval of once every 4 weeks wherein the said method is further characterized by monitoring the effect of the antagonist anti-OX40 antibody in the patient for changes from baseline in the active AD mRNA expression signature and the pathologic epidermal phenotype measures obtained from skin biopsies.
  • the anti-OX40 antagonist antibody comprises: a heavy chain CDR1 comprising the amino acid sequence of SEQ ID NO: 1 , and/or a heavy chain CDR2 comprising the amino acid sequence of SEQ ID NO: 2, and/or a heavy chain CDR3 comprising the amino acid sequence of SEQ ID NO: 3; and/or comprising a light chain CDR1 comprising the amino acid sequence of SEQ ID NO: 4, and/or a light chain CDR2 comprising the amino acid sequence of SEQ ID NO: 5 and/or a light chain CDR3 comprising the amino acid sequence of SEQ ID NO: 6.
  • the anti-OX40 antagonist antibody comprises: a heavy chain sequence comprising amino acid sequence of SEQ ID NO: 7 or 8 and light chain sequence comprising the amino acid sequence of SEQ ID NO: 9.
  • the anti-OX40 antagonist antibody is present at a concentration of at least 0.1 mg/ml.
  • the anti-OX40 antagonist antibody is present at a concentration of at least 1 mg/ml. In accordance with this aspect of the present invention the anti-OX40 antagonist antibody is present at a concentration of at least 10 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 6.25.
  • the formulation may further comprise one or more excipients and bulking agents.
  • a low concentration aqueous formulation of an anti-OX40 antagonist antibody adjusted to pH 6.25 comprising: lOmg/ml of an anti-OX40 antagonist antibody; 15mM Histidine;150mM NaCl;0.01% Tween 80.
  • the inventors whilst developing these formulations have overcome issues associated with aqueous liquid formulations comprising proteins in general and antibodies in particular.
  • Several alternative formulations tried during the course of this research program lack long term stability at 5°C, 25°C or 40°C, in particular the antibody was subject to degradation and apparent instability in several of the alternative formulations not comprising the specific combination of components listed above.
  • 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 ELIS A 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-SEC analysis to determine changes in the antibody in the formulation.
  • FIG 1 Stability data for IV formulation at 5°C.
  • FIG 2 Stability data for IV formulation at 25°C.
  • FIG 3 Stability data for IV formulation at 40°C. Definitions
  • human OX40 as used herein includes variants, isoforms, and species homologs of human OX40. Accordingly, antibodies of this disclosure may, in certain cases, cross-react with OX40 from species other than human. In certain embodiments, the antibodies may be completely specific for one or more human OX40 proteins and may not exhibit species or other types of non-human cross-reactivity.
  • the complete amino acid sequence of an exemplary human OX40 has Swiss-Prot accession number P43489.
  • OX40 is also known as CD134, TNFRSF4, ACT35 or TXGP1 L.
  • Human OX40 is designated GenelD: 7293 by Entrez Gene, and HGNC: 1 1918 by HGNC.
  • OX40 has also been designated CD 134 (cluster of differentiation 134).
  • OX40 can be encoded by the gene designated TNFRSF4 /OX40.
  • human OX40 encompasses all known or as yet undiscovered alleles and polymorphic forms of human OX40.
  • the terms "human OX40”, “OX40” or “OX40 Receptor” are used herein equivalently and mean “human OX40” if not otherwise specifically indicated.
  • OX40 ligand or “OX40L” are used herein equivalently and include OX40 ligand, specifically human OX40 ligand.
  • OX40L is a member of the TNF superfamily and is also known as gp34 or CD252.
  • OX40L has also been designated CD252 (cluster of differentiation 252) and has the sequence database accession number P23510 (Swiss-Prot) or Q6FGS4 (Uniprot).
  • OX40L is expressed on the surface of activated B cells, T cells, dendritic cells and endothelial cells.
  • antibody or fragment thereof that binds to human OX40 includes antibodies or a fragment thereof that binds to human OX40 e.g. human OX40 in isolated form, with an affinity (KD) of 500 nM or less, preferably 200nM or less, more preferably 150 nM or less, more preferably 120 nM or less, even more preferably 1 10 nM or less.
  • affinity 500 nM or less, preferably 200nM or less, more preferably 150 nM or less, more preferably 120 nM or less, even more preferably 1 10 nM or less.
  • antibody or fragment thereof that binds to human OX40 includes antibodies or antigenic binding fragments thereof.
  • an antibody that is capable of inhibiting and/or neutralising the biological signalling activity of OX40, for example by blocking binding or substantially reducing binding of OX40 to OX40 ligand and thus inhibiting or reducing the signalisation pathway triggered by OX40 and/or inhibiting or reducing an OX40-mediated cell response like lymphocyte proliferation, cytokine expression, or lymphocyte survival.
  • the term “antibody” as referred to herein includes whole antibodies and any antigen binding fragments or single chains thereof.
  • An “antibody” refers to a glycoprotein comprising at least two heavy (H) chains and two light (L) chains inter-connected by disulfide bonds, or an antigen binding fragment thereof.
  • Each heavy chain is comprised of a heavy chain variable region (abbreviated herein as VH) and a heavy chain constant region.
  • the heavy chain constant region is comprised of three domains, CHI, CH2 and CH3.
  • Each light chain is comprised of a light chain variable region (abbreviated herein as VL) and a light chain constant region.
  • the light chain constant region is comprised of one domain, CL.
  • the VH and VL regions can be further subdivided into regions of hypervariability, termed complementarity determining regions (CDR) with are hypervariable in sequence and/or involved in antigen recognition and/or usually form structurally defined loops, interspersed with regions that are more conserved, termed framework regions (FR or FW).
  • CDR complementarity determining regions
  • Each VH and VL is composed of three CDRs and four FWs, arranged from amino- terminus to carboxy-terminus in the following order: FWl , CDR1 , FW2, CDR2, FW3, CDR3, FW4.
  • the amino acid sequences of FWl , FW2, FW3, and FW4 all together constitute the "non-CDR region" or "non-extended CDR region" of VH or VL as referred to herein.
  • heavy chain variable framework region may comprise one or more (e.g., one, two, three and/or four) heavy chain framework region sequences (e.g., framework 1 (FWl), framework 2 (FW2), framework 3 (FW3) and/or framework 4 (FW4)).
  • the heavy chain variable region framework comprises FWl , FW2 and/or FW3, more preferably FWl , FW2 and FW3.
  • the term "light chain variable framework region” as referred herein may comprise one or more (e.g., one, two, three and/or four) light chain framework region sequences (e.g., framework 1 (FWl), framework 2 (FW2), framework 3 (FW3) and/or framework 4 (FW4)).
  • the light chain variable region framework comprises FWl , FW2 and/or FW3, more preferably FWl, FW2 and FW3.
  • variable regions of the heavy and light chains contain a binding domain that interacts with an antigen.
  • the constant regions of the antibodies may mediate the binding of the immunoglobulin to host tissues or factors, including various cells of the immune system (e.g., effector cells) and the First component (Cl q) of the classical complement system.
  • 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 (IGHAl), 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 al., (1988) 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 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 “humanized anti-OX40 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 C gamma 2 and C gamma 3 (Cy2 and Cy3) and the hinge between C gamma 1 (Cyl) and C gamma 2 (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.
  • the Fc region is herein defined to comprise residue P232 to its carboxyl-terminus, wherein the numbering is according to the EU numbering system (Edelman GM et al., (1969) Proc Natl Acad Sci USA, 63(1): 78-85).
  • 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 '3 ⁇ 4inge 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 (Edelman GM et al., supra).
  • parent antibody or “parent immunoglobulin” as used herein includes 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.
  • parent anti-OX40 antibody as used herein is meant an antibody or immunoglobulin that binds human OX40 and is modified to generate a variant.
  • corresponding murine antibody as used herein is meant a murine antibody or immunoglobulin that bind to human OX40 and that can be modified to generate a variant, specifically the murine antibody 1D4 as disclosed herein.
  • 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. These families include 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), nonpolar side chains (e.g., alanine, valine, leucine, isoleucine, proline, phenylalanine, methionine), beta-branched side chains (e.g., threonine, valine, isoleucine) and aromatic side chains (e.g., tyrosine, phenylalanine, tryptophan, histidine).
  • 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
  • IGLC7 human lambda immunoglobulin light chain constant domains
  • 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
  • Dariavach P et al. (1987) Proc Natl Acad Sci USA, 84(24): 9074-8 and Frangione B et al, (1985) Proc Natl Acad Sci USA, 82(10): 3415-9.
  • 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 (VH) and the other located in the light chain (VL).
  • VH heavy chain
  • VL light chain
  • 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 1 10 amino acids long, and consist of relatively invariant stretches of amino acid sequence called framework regions (FRs) of 15-30 amino acids separated by shorter regions of extreme variability called "hypervariable regions” that are 9-12 amino acids long.
  • FRs framework regions
  • variable domains of native heavy and light chains comprise four FRs, largely adopting a beat-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., supra).
  • 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., supra).
  • 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., supra). Chothia refers instead to the location of the structural loops (Chothia C & Lesk AM (1987) J. 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) Proc. Natl Acad. Sci. USA, 86: 9268-72; 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 the international ImMunoGeneTics information system®
  • Lefranc MP the international ImMunoGeneTics information system®
  • CDRs Complementarity Determining Regions
  • EVIGT® Complementarity Determining Regions
  • the variable domain residues for each of these CDRs are as follows (numbering according to Kabat EA, et al., supra): LCDR1 : 27-32, LCDR2: 50-52, LCDR3: 89-97, HCDRl : 26-35, HCDR2: 51-57 and HCDR3: 93-102.
  • non-CDR region of the VL region as used herein comprise the amino acid sequences: 1-26 (FR1), 33-49 (FR2), 53- 88 (FR3), and 98- approximately 107 (FR4).
  • the "non-CDR region” of the VH region as used herein comprise the amino acid sequences: 1 -25 (FR1), 36-50 (FR2), 58-92 (FR3), and 103- approximately 113 (FR4).
  • the CDRs of the present invention may comprise "extended CDRs" which are based on the aforementioned definitions and have variable domain residues as follows: LCDR1 : 24-36, LCDR2: 46-56, LCDR3:89-97, HCDRl : 26-36, HCDR2:47-65, HCDR3: 93-102. These extended CDRs are numbered as well according to Kabat et al., supra.
  • the "non-extended CDR region" of the VL region as used herein comprise the amino acid sequences: 1-23 (FR1), 37-45 (FR2), 57-88 (FR3), and 98- approximately 107 (FR4).
  • the "non-extended CDR region” of the VH region as used herein comprise the amino acid sequences: 1 -25 (FR1), 37-46 (FR2), 66-92 (FR3), and 103- approximately 113 (FR4).
  • full length antibody 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 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 ES et al, (1989) Nature, 341 : 544-546) which consists of a single variable, (v) F(ab')2 fragments, a bivalent fragment comprising two linked Fab fragments (vi) single chain Fv molecules (scFv), wherein a VH domain and a VL domain are linked by a peptide linker which allows the two domains to associate to form an antigen binding site (Bird RE et al, (1988) Science 242: 423-426; Huston JS et al, (1988) Proc.
  • 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, e.g. enhancing or suppressing various reactions of the immune system, with possible beneficial effects in diagnosis and therapy.
  • OX40-mediated disorder includes conditions such as allergy, asthma, COPD, rheumatoid arthritis, psoriasis and diseases associated with autoimmunity and inflammation.
  • the term “subject” includes any human or nonhuman animal.
  • nonhuman animal includes all vertebrates, e.g., mammals and non-mammals, such as nonhuman primates, sheep, dogs, cats, horses, cows, chickens, amphibians, reptiles, etc.
  • the subject is human.
  • Atopic dermatitis means an inflammatory skin disease characterized by intense pruritus (e.g., severe itch) and by scaly and dry eczematous lesions.
  • the term "atopic dermatitis” includes, but is not limited to, AD caused by or associated with epidermal barrier dysfunction, allergy (e.g., allergy to certain foods, pollen, mold, dust mite, animals, etc.), radiation exposure, and/or asthma.
  • the present invention encompasses methods to treat patients with mild, moderate -to-severe or severe AD.
  • Moderate-to-severe AD is characterized by intensely pruritic, widespread skin lesions that are often complicated by persistent bacterial, viral or fungal infections.
  • Moderate -to-severe AD also includes chronic AD in patients.
  • the chronic lesions include thickened plaques of skin, lichenification and fibrous papules.
  • Patients affected by moderate-to-severe AD also, in general, have more than 10% of the body's skin affected, or 10% of skin area in addition to involvement of the eyes, hands and body folds.
  • Moderate -to-severe AD is also considered to be present in patients who require frequent treatment with topical corticosteroids.
  • a patient may also be said to have moderate-to -severe AD when the patient is resistant or refractory to treatment by either a topical corticosteroid or a calcineurin inhibitor or any other commonly used therapeutic agent known in the art.
  • AD related efficacy parameters include: (a) Scoring of Atopic Dermatitis -SCORAD (b) Investigators Global Assessment (IGA); (c) Pruritus Numerical rating scale( NRS) (d) Dermatology Life Quality Index-DLQI (e)Body Surface Area (BSA); (f) Eczema Area and Severity Index (EASI); (h) and trans - epidermal water loss( TEWL).An “improvement in an AD- related efficacy parameters " means a decrease from baseline of one or more of IGA, BSA, EASI, SCORAD, TEWL, DLQI or NRS.
  • baseline means the numerical value of the AD- related efficacy parameters for a subject prior to or at the time of administration of a pharmaceutical composition of the present invention SCORAD.
  • the SCORAD is a validated tool used in clinical research and clinical practice that was developed to standardize the evaluation of the extent and severity of AD (Dermatology 1993). The extent of AD is assessed as a percentage of each defined body area and reported as the sum of all areas, with a maximum score of 100% (assigned as "A" in the overall SCORAD calculation).
  • AD The severity of 6 specific symptoms of AD is assessed using the following scale: none (0), mild (1), moderate (2), or severe (3) (for a maximum of 18 total points, assigned as "B” in the overall SCORAD calculation).
  • Subjective assessment of itch and sleeplessness is recorded for each symptom by the patient or relative on a visual analogue scale (VAS), where 0 is no itch (or sleeplessness) and 10 is the worst imaginable itch (or sleeplessness), with a maximum possible score of 20. This parameter is assigned as "C” in the overall SCORAD calculation.
  • VAS visual analogue scale
  • the SCORAD is calculated as: A 5 + 7B/2 + C (Kunz et al, 1997).Investigators Global Assessment (IGA);The IGA is an assessment scale used in clinical studies to determine severity of AD and clinical response to treatment based on a 5 -point scale ranging from 0 (clear) to 4 (severe/very severe).
  • Pruritus Numerical rating scale (NRS)Pruritus Numerical rating scale (NRS): The Pruritus NRS is a single-question assessment tool that is used to assess a subject's worst itch, on a scale of 1 to 10, as a result of AD in the previous 12 hours. Patients will record once daily and respond to the following question, "On a scale of 0 - 10, with 0 being no itch and 10 being the worst itch imaginable, how would you rate your worst degree of itch during the previous 24 hours?" Patient compliance on the pruritus NRS will be followed at each clinic visit.
  • the DLQI is a simple, patient-administered, 10- question, validated, quality-of-life questionnaire that covers 6 domains including symptoms and feelings, daily activities, leisure, work and school, personal relationships, and treatment. Response categories include "a little,” “a lot,” and “very much” with corresponding scores of 1 , 2, and 3 respectively and “not at all", “not relevant” responses scored as “0.” Totals range from 0 to 30 (less to more impairment) and a 5-point change from baseline is considered clinically relevant (Basra et al, 2008; Finlay et al, 1994).
  • BSA Body Surface Area
  • EASI Eczema Area and Severity Index
  • the EASI is a validated measure used in clinical practice and clinical trials to assess the severity and extent of AD.
  • Four AD disease characteristics will be assessed for severity by the investigator or designee on a scale of "0" (absent) through "3" (severe).
  • the area of AD involvement will be assessed as a percentage by body area of head, trunk, arms, and legs and converted to a score of 0 to 6 (Hanifin, 2001).
  • Atopic dermatitis biomarker parameters The present invention also includes methods involving the use, quantification, and analysis of Atopic dermatitis biomarker parameters.
  • Atopic dermatitis biomarker parameters means any biological response, cell type, parameter, protein, polypeptide, enzyme, enzyme activity, metabolite, nucleic acid, carbohydrate, or other biomolecule which is present or detectable in an AD patient at a level or amount that is different from (e.g., greater than or less than) the level or amount of the marker present or detectable in a non-AD patient.
  • the term “Atopic dermatitis biomarker parameters” includes a biomarker associated with Type 2 helper T-cell Th2)-driven inflammation.
  • AD transcriptome In order to evaluate for the drug effect or how much of the disease profile has been reversed by treatment as measured changes in the AD transcriptome using gene arrays consisting of differentially expressed genes between lesional and non lesional AD skin as defined by fold changes (typically a fold change of more than 2)
  • the AD disease phenotype is the integration of cellular and molecular markers that define the epidermal pathology (hyperplasia, differentiation abnormalities), and Th2, and Th22 immune activation. The changes or reversal of these immune and barrier defects will be assessed by IHC and RT-PCR.
  • AD-associated biomarkers include a panel of Thl , Th2, Th22 cytokines and chemokines that are shown as elevated in AD blood and to decrease with treatment will be assessed, including but not limited to, e.g., Serum Thymus and activation-regulated chemokine (TARC/CCL17), eotaxin-3, total Immunoglobulin E (IgE), Thymus and activation-regulated chemokine is a chemokine, shown to be strongly associated with disease severity in AD, and may be involved in pathogenesis of the disease. Baseline TARC levels will be assessed for potential predictive value for treatment response.
  • TARC/CCL17 Serum Thymus and activation-regulated chemokine
  • IgE total Immunoglobulin E
  • Thymus and activation-regulated chemokine is a chemokine, shown to be strongly associated with disease severity in AD, and may be involved in pathogenesis of the disease.
  • Baseline TARC levels will be assessed for potential predictive value for treatment response.
  • Eotaxin-3 (CCL26), Eotaxin-3 is a chemokine, shown to be associated with disease severity in AD, and may be involved in pathogenesis of the disease. Baseline eotaxin-3 levels will be assessed for potential predictive value for treatment response. Post-treatment samples will be evaluated for effects of anti OX40 antagonist antibody on eotaxin-3., Total Immunoglobulin E (IgE), Patients with AD often have elevated IgE. Total IgE levels have been found to modestly correlate with AD severity and may be involved in the pathogenesis of the disease. Changes in total IgE reflects not only on AD, but atopy in general. Baseline IgE levels will be assessed for potential predictive value for treatment response.
  • IgE Total Immunoglobulin E
  • Transepidermal water loss is a skin barrier function test that measures perspiration or water loss through the skin. This procedure involves the non-invasive application of a probe on the surface of the skin on the arm or leg. Affected and non-affected areas of skin will be tested.
  • Example 1 - IV formulation The inventors have generated and tested a low concentration liquid formulation of an anti-Ox40 antagonist antibody adjusted to pH 6.25, comprising: lOmg/ml of an anti-ox40 antagonist antibody comprising a light chain variable sequence comprising: SEQ ID NO: 9 and a heavy chain variable region comprising SEQ ID NO: 7 or 8; 15mM Histidine; 150mM NaCl;0.01% Tween 80.
  • the inventors have characterised the stability of the anti-Ox40 antagonist 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, Tl 8, T24, T30.
  • the antibody present in the formulation was characterized 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 15 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-SEC analysis so as to determine changes in the antibody in the formulation.Each of the 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
  • T-cell driven autoimmune and inflammatory diseases often display a relapse - remitting pattern.
  • the relapses are driven mainly by reactivation of autoimmune memory T lymphocytes. Therefore inhibiting such memory T cell immune responses represents a very promising therapeutic strategy for autoimmune diseases such as rheumatoid arthritis, systemic lupus erythematosus, multiple sclerosis, psoriasis and others.
  • the antigen selected for MS was human Myelin Basic protein (MBP) purified from human brain.
  • the antigen selected for lupus was the SmDl 83-119 peptide as described in Riemekasten et al. Ann Rheum Dis 2002;61 :779-785.
  • the antigen selected for uveitis was the human soluble antigen (S-Ag), as described in Smet et al, Invest Opthalmol Vis Sci. 2001 Dec;42(13):3233-8. Briefly the PBMCs from patients were incubated during 5 to 9 days with or without relevant antigens in presence or not of the claimed or control antibodies. Each condition was performed at least in triplicates. Tritiated thymidine incorporation was measured to quantify the proliferation happening in each condition.
  • the proliferation is described to be a direct consequence of T lymphocyte reactivation to an antigen provided.
  • a stimulation index was calculated for each donor and antigen tested, as the ratio between the condition with the antigen and without antigen. If the stimulation index was superior or equal to 2, the donor was considered as a responder to that antigen.
  • the claimed antibody potently suppressed the T cell reactivation response to the relevant antigens, in essentially all responders.
  • Example 3 Antagonistic anti-OX40 antibody suppresses a physiological memory T cell response in an in vitro Tetanus Toxoid re-stimulation assay
  • PBMCs peripheral blood mononuclear cells
  • TT purified tetanus toxoid
  • 3 H-T tritiated-thymidine incorporation
  • PBMC isolation was performed by Ficoll density gradient.
  • the PBMCs were washed 3 times with PBS, counted and resuspended into cell culture medium.
  • the PBMCs were then distributed into 96-well U bottom plates at 10 5 or 2x10 5 cells/well.
  • TT was added to relevant wells at 0.17 ⁇ g/ml final.
  • the plates were incubated for 3-7 days at 37°C in a C0 2 incubator. The day before harvesting, 0.5 ⁇ / ⁇ of 3 H-T was added to each well and the plates were incubated at 37°C for an additional 15-24 h.
  • the plates were harvested using a Packard filtermate harvester.
  • a stimulation index was defined as the ratio between the condition "PBMC only” (no TT) and the condition "with TT only”. Donors were considered as responders if the SI was superior to 3.
  • Table 2 Summary of the effect of an antagonistic anti-OX40 antibody and abatacept on TT recall response
  • SI stimulation index
  • Example 4 Phase la clinical trial to establish safety and tolerability of antagonistic anti- OX40 antibody A phase la clinical study was undertaken to evaluate the safety and tolerability of single ascending doses of an antagonistic anti-OX40 antibody (0.3, 1.0, 3.0 and 10.0 mg kg) in healthy volunteers (HVs). Subject disposition: 32 subjects were enrolled to receive single IV doses of the antagonistic anti- OX40 antibody (24 HVs) or matching placebo (8 HVs). All subjects completed the study and were included in the final analysis for PK and safety analysis.
  • Example 5 Pharmacokinetic data from Phase lb, Randomized, Placebo-controlled, Single Ascending Dose Study in Adult Healthy Volunteers
  • Serum samples were collected at scheduled time points uptolO weeks post dosing.
  • concentrations of the claimed antagonistic anti-OX40 antibody in serum were estimated using a validated ELISA method (LLOQ: 97.7 ng/mL).
  • LLOQ 97.7 ng/mL
  • the interim serum concentration data along with nominal sampling time points for individual subjects were subjected to non-compartmental analysis using Phoenix® WinNonlin version 6.4.
  • the summary of PK parameters is presented in Table 3.

Abstract

The present invention relates to anti OX40 antagonist antibody for the treatment of atopic dermatitis, including moderate-to-severe Atopic dermatitis and stable liquid formulations suitable for the treatment of atopic dermatitis and other autoimmune diseases.

Description

Anti-OX40 antagonistic antibodies for the treatment of Atopic Dermatitis
Technical Field
The present invention relates to anti OX40 antagonist antibody for the treatment of atopic dermatitis, including moderate-to-severe Atopic dermatitis. The present application also relates to a method of treating Atopic dermatitis by the parenteral administration of an anti OX40 antagonist antibody to a patient in need thereof. The present invention also relates to a pharmaceutical composition comprising an anti OX40 antagonist antibody and processes for preparing such a pharmaceutical composition. Background of Invention
Atopic Dermatitis (AD) is one of the most prevalent autoimmune disorders worldwide that affects around 4-7% of adults and 15-25% of children with a significant impact on the quality of patient life. It is a chronic, relapsing inflammatory skin disease characterized by intense pruritus (e.g., itchiness) and by scaly and dry eczematous lesions and often associated with other atopic disorders, such as allergic rhinitis and asthma. Despite its increasing prevalence worldwide, and the burden on society, specific therapies for AD are still limited, and the most commonly used therapies are not based on a mechanistic understanding of the disease. Approximately 20% of patients with AD have moderate to severe symptoms and generally require systemic therapy, as outlined in several international and regional treatment guidelines (Sidbury et al, Guidelines of care for the management of atopic dermatitis; Section 3. Management and treatment with phototherapy and systemic agents; J Am Acad Dermatol 2014;71 :327-49, Ring et al, Guidelines for treatment of atopic eczema (atopic dermatitis) Part II; Journal of the European Academy of Dermatology and Venereology 2012, 1468-3083).
Although there are multiple systemic agents recommended for the treatment of AD, many of the patients still do not achieve optimal efficacy and existing treatments have limited efficacy in moderate to severe disease. Drug-specific safety concerns are another major limitation (Levin et al, Biologic fatigueJ Dermatolog Treat. 2014 Feb; 25(l):78-82). Thus, there remains a significant unmet patient need for new agents with unique mechanisms that can provide improved and sustained skin clearance, and a safety profile that allows for chronic use. Biologic agents hold the promise of a more targeted, effective and less toxic approach to systemic therapy.
OX40 is a member of the TNFR-superfamily of receptors and was first identified in 1987 as a 50 kDa glycoprotein expressed on activated CD4+ T cells from the rat (Paterson DJ et al., (1987) Mol. Immunol. 24: 1281-90).
The extracellular ligand binding domain of OX40 is composed of 3 full cysteine-rich domains (CRDs) and a partial, fourth C-terminal CRD (Bodmer JL et al., (2002) Trends Biochem. Sci. 27: 19-26). The ligand for OX40 is OX40L (CD252) and 3 copies of OX40 bind to the trimeric ligand to form the OX40-OX40L complex (Compaan DM & Hymowitz SG (2006) Structure, 14: 1321-1330). OX40 is a membrane -bound receptor; however a soluble isoform has also been detected (Taylor L & Schwarz H (2001) J. Immunol. Methods, 255: 67-72). Unlike CD28, OX40 is not constitutively expressed on naive T cells but is induced after engagement of the T-Cell Receptor (TCR). OX40 is a secondary costimulatory molecule, expressed after 24 to 72 hours following activation; its ligand, OX40L, is also not expressed on resting antigen presenting cells, but is following their activation. OX40 is expressed mainly by activated CD4+ T cells and to a limited extent, by activated CD8+ T cells (Salek-Ardakani S et al., (2006) Curr. Immunol. Rev. 2: 37-53).Blocking the OX40/OX40L pathway has been shown to be protective in several animal models of human disease such as asthma, irritable bowel disease, transplant rejection, autoimmune diabetes, graft versus host disease (GvHD) and experimental autoimmune encephalomyelitis, thus validating OX40 as a highly attractive pathway to antagonize in autoimmune diseases. Unlike conventional immunotherapies, blockade of OX40-OX40L interactions seems to specifically inhibit disease-responsible effector T-cell function, and therefore not cause widespread immunosuppression.
Summary of invention The present invention provides an anti-OX40 antagonist antibody for use in the treatment or prevention of atopic dermatitis, including moderate -to-severe atopic dermatitis.
In accordance with this aspect of the present invention the anti-OX40 antagonist antibody comprises: a heavy chain CDR1 comprising the amino acid sequence of SEQ ID NO: 1 , and/or a heavy chain CDR2 comprising the amino acid sequence of SEQ ID NO: 2, and/or a heavy chain CDR3 comprising the amino acid sequence of SEQ ID NO: 3; and/or comprising a light chain CDR1 comprising the amino acid sequence of SEQ ID NO: 4, and/or a light chain CDR2 comprising the amino acid sequence of SEQ ID NO: 5 and/or a light chain CDR3 comprising the amino acid sequence of SEQ ID NO: 6.
In accordance with this aspect of the present invention the anti-OX40 antagonist antibody comprises: a heavy chain sequence comprising amino acid sequence of SEQ ID NO: 7 or 8 and light chain sequence comprising the amino acid sequence of SEQ ID NO: 9.
Preferably, the present invention provides uses, compositions or methods which cause an improvement in one or more AD efficacy parameter(s) in a patient. Improvements in AD related efficacy parameters include, for instance a decrease in Investigator's Global Assessment (IGA) score; a decrease in Body Surface Area (BSA) score; a decrease in Eczema Area and Severity Index (EASI) score; a decrease in SCORAD score; a decrease in Pruritus Numeric Rating Scale (NRS) score, and/or improvement of Dermatology Life quality index( DLQI). AD efficacy parameter also may mean a change in the transcriptome or in a change in the cyto/chemokine profile of the patient or a biopsy from the site of AD on the patient.
According to other embodiments, the present invention provides a method for treating or preventing AD in a subject, the methods comprising: (a) selecting a subject who exhibits an abberant level of at least one AD-associated biomarker or is diagnosed clinically with AD; and (b) administering to the subject a pharmaceutical composition comprising a therapeutically effective amount of an anti-OX 40 antagonist antibody. Wherein said AD-associated biomarker is selected from the group, Exe but are not limited to, Serum Thymus and activation-regulated chemokine (TARC/CCL17), eotaxin-3, total Immunoglobulin E (IgE), Immunohistochemistry and trans -epidermal water loss (TEWL). The present application also relates to a method of treating Atopic dermatitis in a subject by the parenteral administration to a patient in need of a therapeutically effective amount of an anti OX40 antagonist antibody. In a further aspect, the invention provides a method of treating of atopic dermatitis comprising administering to a subject a composition comprising a therapeutically effective amount of an anti OX40 antagonist antibody and a pharmaceutically acceptable carrier.
Accordingly in one aspect, the invention provides a method of treating atopic dermatitis comprising administering to a subject a therapeutically effective amount of anti OX40 antagonist comprising a heavy chain CDR1 comprising the amino acid sequence of SEQ ID NO: 1 , and/or a heavy chain CDR2 comprising the amino acid sequence of SEQ ID NO: 2, and/or a heavy chain CDR3 comprising the amino acid sequence of SEQ ID NO: 3; and/or comprising a light chain CDR1 comprising the amino acid sequence of SEQ ID NO: 4, and/or a light chain CDR2 comprising the amino acid sequence of SEQ ID NO: 5 and/or a light chain CDR3 comprising the amino acid sequence of SEQ ID NO: 6.
Accordingly in one aspect the present invention relates to a method of treating atopic dermatitis comprising administering to a subject a therapeutically effective amount of anti OX40 antagonist comprising a heavy chain sequence comprising amino acid sequence of SEQ ID NO: 7 or 8 and light chain sequence comprising the amino acid sequence of SEQ ID NO: 9.
According to one aspect for administration the anti OX40 antagonist antibody dosage ranges from 0.0001 to 100 mg kg, and more preferably between 0.01 to 10 mg/kg, of the host body weight. In particular the dose is selected from the group comprising 0.001 , 0.002, 0.003, 0.004, 0.005, 0.006, 0.007, 0.008, 0.009, 0.01 , 0.01 , 0.02, 0.03, 0.04, 0.05, 0.06, 0.07, 0.08, 0.09, 0.1 , 0.2, 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, 1 , 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 45, 50, 60, 70, 80, 90, 100 mg/kg. In accordance with another aspect of the present invention this relates to fixed dose preparations of 100, 200, 300, 400, 500, 600, 700, 800, 900 mg per dose. In accordance with another aspect of the present invention this relates to a dose administered to a patient at an interval of daily, weekly biweekly, monthly, bimonthly or any suitable interval in between.
In accordance with this aspect of the present invention the anti-OX40 antagonist antibody comprises: a heavy chain CDR1 comprising the amino acid sequence of SEQ ID NO: 1 , and/or a heavy chain CDR2 comprising the amino acid sequence of SEQ ID NO: 2, and/or a heavy chain CDR3 comprising the amino acid sequence of SEQ ID NO: 3; and/or comprising a light chain CDR1 comprising the amino acid sequence of SEQ ID NO: 4, and/or a light chain CDR2 comprising the amino acid sequence of SEQ ID NO: 5 and/or a light chain CDR3 comprising the amino acid sequence of SEQ ID NO: 6. In accordance with this aspect of the present invention the anti-OX40 antagonist antibody comprises: a heavy chain sequence comprising amino acid sequence of SEQ ID NO: 7 or 8 and light chain sequence comprising the amino acid sequence of SEQ ID NO: 9.
In another embodiment, the present invention relates to a method of treating atopic dermatitis in a patient said method comprising the parental administration to the subject of 10 mg/kg of an anti OX40 antagonist antibody and its acceptable carrier, two doses administered at an interval of once every 4 weeks wherein the said method is further characterized by monitoring the effect of the antagonist anti-OX40 antibody in the patient for changes from baseline in the active AD mRNA expression signature and the pathologic epidermal phenotype measures obtained from skin biopsies.
Wherein the change from baseline expression levels of cytokines in serum: Interleukin (IL)-13 and IL-22, Chemokine ligands (CCL) CCL2, CCL3, CCL4, CCL5, CCL18, CCL20, CCL22, CCL13, CXCL9, CXCL10, CXCL11 are monitored., Leukocyte sub-population cell counts (Total T, T helper, Cytotoxic T, Tregs, Memory T cells, OX40 T cells)., Cellular infiltrates (T- cells, Dendritic cells) as assessed by CD3, FcEpsilon RI, and OX40L., Serum Thymus and activation-regulated chemokine (TARC/CCL17), eotaxin-3, total Immunoglobulin E (IgE), and circulating eosinophil counts , Percentage OX40 receptor occupancy (RO)According to one aspect of the present invention there is provided a stable aqueous pharmaceutical formulation comprising a therapeutically effective amount of an anti-ox40 antagonist antibody, a buffer, a surfactant and a tonicifying agent, wherein the pH of the formulation is from about 5 to about 7.
In accordance with this aspect of the present invention the anti-OX40 antagonist antibody comprises: a heavy chain CDR1 comprising the amino acid sequence of SEQ ID NO: 1 , and/or a heavy chain CDR2 comprising the amino acid sequence of SEQ ID NO: 2, and/or a heavy chain CDR3 comprising the amino acid sequence of SEQ ID NO: 3; and/or comprising a light chain CDR1 comprising the amino acid sequence of SEQ ID NO: 4, and/or a light chain CDR2 comprising the amino acid sequence of SEQ ID NO: 5 and/or a light chain CDR3 comprising the amino acid sequence of SEQ ID NO: 6.
In accordance with this aspect of the present invention the anti-OX40 antagonist antibody comprises: a heavy chain sequence comprising amino acid sequence of SEQ ID NO: 7 or 8 and light chain sequence comprising the amino acid sequence of SEQ ID NO: 9.
In accordance with this aspect of the present invention the anti-OX40 antagonist antibody is present at a concentration of at least 0.1 mg/ml.
In accordance with this aspect of the present invention the anti-OX40 antagonist antibody is present at a concentration of at least 1 mg/ml. In accordance with this aspect of the present invention the anti-OX40 antagonist antibody is present at a concentration of at least 10 mg/ml.
In accordance with this aspect of the present invention the buffer is selected from the group comprising: citrate, acetate, histidine, phosphate, tris (tris(hydroxymethyl)aminomethane). In accordance with this aspect of the present invention 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. In accordance with this aspect of the present invention the surfactant is selected from the group comprising: tween 20/40/80, polysorbate 20/80, poloxamer, sodium lauryl sulphate.
In accordance with this aspect of the present invention the pH of the formulation is 6.25. In accordance with this aspect of the present invention, the formulation may further comprise one or more excipients and bulking agents.
In accordance with a preferred embodiment of the present invention there is provided a low concentration aqueous formulation of an anti-OX40 antagonist antibody adjusted to pH 6.25 comprising: lOmg/ml of an anti-OX40 antagonist antibody; 15mM Histidine;150mM NaCl;0.01% Tween 80.The inventors whilst developing these formulations have overcome issues associated with aqueous liquid formulations comprising proteins in general and antibodies in particular. Several alternative formulations tried during the course of this research program lack long term stability at 5°C, 25°C or 40°C, in particular the antibody was subject to degradation and apparent instability in several of the alternative formulations not comprising the specific combination of components listed above.
In accordance with a further aspect of the present invention the formulation is stable at 5°C for at least 1 year and preferably at least 2 years.
In accordance with a further aspect of the present invention the formulation is stable at 25°C for at least 3 months, preferably 6 months and most preferably 1 year.
In accordance with a further aspect of the present invention the formulation is stable at 40°C for at least 1 month and preferably 3 months.
Wherein 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 ELIS A 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-SEC analysis to determine changes in the antibody in the formulation.
Figures
FIG 1 : Stability data for IV formulation at 5°C. FIG 2: Stability data for IV formulation at 25°C. FIG 3: Stability data for IV formulation at 40°C. Definitions
The term "human OX40" as used herein includes variants, isoforms, and species homologs of human OX40. Accordingly, antibodies of this disclosure may, in certain cases, cross-react with OX40 from species other than human. In certain embodiments, the antibodies may be completely specific for one or more human OX40 proteins and may not exhibit species or other types of non-human cross-reactivity. The complete amino acid sequence of an exemplary human OX40 has Swiss-Prot accession number P43489. OX40 is also known as CD134, TNFRSF4, ACT35 or TXGP1 L. Human OX40 is designated GenelD: 7293 by Entrez Gene, and HGNC: 1 1918 by HGNC. OX40 has also been designated CD 134 (cluster of differentiation 134). OX40 can be encoded by the gene designated TNFRSF4 /OX40.
The use of "human OX40" herein encompasses all known or as yet undiscovered alleles and polymorphic forms of human OX40. The terms "human OX40", "OX40" or "OX40 Receptor" are used herein equivalently and mean "human OX40" if not otherwise specifically indicated.
The term "OX40 ligand" or "OX40L" are used herein equivalently and include OX40 ligand, specifically human OX40 ligand. OX40L is a member of the TNF superfamily and is also known as gp34 or CD252. OX40L has also been designated CD252 (cluster of differentiation 252) and has the sequence database accession number P23510 (Swiss-Prot) or Q6FGS4 (Uniprot). OX40L is expressed on the surface of activated B cells, T cells, dendritic cells and endothelial cells.
The term "antibody or fragment thereof that binds to human OX40" as used herein includes antibodies or a fragment thereof that binds to human OX40 e.g. human OX40 in isolated form, with an affinity (KD) of 500 nM or less, preferably 200nM or less, more preferably 150 nM or less, more preferably 120 nM or less, even more preferably 1 10 nM or less. The term "antibody or fragment thereof that binds to human OX40" includes antibodies or antigenic binding fragments thereof.
The terms "antagonistic antibody" or "antagonist antibody" are used herein equivalently and include an antibody that is capable of inhibiting and/or neutralising the biological signalling activity of OX40, for example by blocking binding or substantially reducing binding of OX40 to OX40 ligand and thus inhibiting or reducing the signalisation pathway triggered by OX40 and/or inhibiting or reducing an OX40-mediated cell response like lymphocyte proliferation, cytokine expression, or lymphocyte survival. The term "antibody" as referred to herein includes whole antibodies and any antigen binding fragments or single chains thereof. An "antibody" refers to a glycoprotein comprising at least two heavy (H) chains and two light (L) chains inter-connected by disulfide bonds, or an antigen binding fragment thereof. Each heavy chain is comprised of a heavy chain variable region (abbreviated herein as VH) and a heavy chain constant region. The heavy chain constant region is comprised of three domains, CHI, CH2 and CH3. Each light chain is comprised of a light chain variable region (abbreviated herein as VL) and a light chain constant region. The light chain constant region is comprised of one domain, CL. The VH and VL regions can be further subdivided into regions of hypervariability, termed complementarity determining regions (CDR) with are hypervariable in sequence and/or involved in antigen recognition and/or usually form structurally defined loops, interspersed with regions that are more conserved, termed framework regions (FR or FW). Each VH and VL is composed of three CDRs and four FWs, arranged from amino- terminus to carboxy-terminus in the following order: FWl , CDR1 , FW2, CDR2, FW3, CDR3, FW4. The amino acid sequences of FWl , FW2, FW3, and FW4 all together constitute the "non-CDR region" or "non-extended CDR region" of VH or VL as referred to herein. The term "heavy chain variable framework region" as referred herein may comprise one or more (e.g., one, two, three and/or four) heavy chain framework region sequences (e.g., framework 1 (FWl), framework 2 (FW2), framework 3 (FW3) and/or framework 4 (FW4)). Preferably the heavy chain variable region framework comprises FWl , FW2 and/or FW3, more preferably FWl , FW2 and FW3. The term "light chain variable framework region" as referred herein may comprise one or more (e.g., one, two, three and/or four) light chain framework region sequences (e.g., framework 1 (FWl), framework 2 (FW2), framework 3 (FW3) and/or framework 4 (FW4)). Preferably the light chain variable region framework comprises FWl , FW2 and/or FW3, more preferably FWl, FW2 and FW3.
The variable regions of the heavy and light chains contain a binding domain that interacts with an antigen. The constant regions of the antibodies may mediate the binding of the immunoglobulin to host tissues or factors, including various cells of the immune system (e.g., effector cells) and the First component (Cl q) of the classical complement system.
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 (μ), delta (δ), gamma (γ), alpha (a), or epsilon (ε), and define the antibody's isotype as IgM, IgD, IgG, IgA, and IgE, respectively. Thus, "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 (IGHAl), 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 al., (1988) Nucleic Acids Res. 16(7): 3108). In spite of having an altered switch region, 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.
The term "chimeric 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.
The term "humanized antibody" or "humanized anti-OX40 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.
The term "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.
The term "Fc" or "Fc region", as used herein includes the polypeptide comprising the constant region of an antibody excluding the first constant region immunoglobulin domain. Thus 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. For IgA and IgM, Fc may include the J chain. For IgG, Fc comprises immunoglobulin domains C gamma 2 and C gamma 3 (Cy2 and Cy3) and the hinge between C gamma 1 (Cyl) and C gamma 2 (Cy2). Although the boundaries of the Fc region may vary, 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. For human IgGl the Fc region is herein defined to comprise residue P232 to its carboxyl-terminus, wherein the numbering is according to the EU numbering system (Edelman GM et al., (1969) Proc Natl Acad Sci USA, 63(1): 78-85). 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 '¾inge 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. Thus for IgG 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 (Edelman GM et al., supra).
The term "parent antibody" or "parent immunoglobulin" as used herein includes 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. By "parent anti-OX40 antibody" as used herein is meant an antibody or immunoglobulin that binds human OX40 and is modified to generate a variant. By "corresponding murine antibody" as used herein is meant a murine antibody or immunoglobulin that bind to human OX40 and that can be modified to generate a variant, specifically the murine antibody 1D4 as disclosed herein.
The term "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.
The term "amino acid modification" herein includes an amino acid substitution, insertion, and/or deletion in a polypeptide sequence. By "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. For example, the 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. For the preceding example, 94K indicates the substitution of position 94 with a lysine. For the purposes herein, multiple substitutions are typically separated by a slash. For example, R94K/L78V refers to a double variant comprising the substitutions R94K and L78V. By "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. For example, insert -94 designates an insertion at position 94. By "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. For example, R94- designates the deletion of arginine at position 94.
As used herein, the term "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. These families include 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), nonpolar side chains (e.g., alanine, valine, leucine, isoleucine, proline, phenylalanine, methionine), beta-branched side chains (e.g., threonine, valine, isoleucine) and aromatic side chains (e.g., tyrosine, phenylalanine, tryptophan, histidine). Thus, 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.
For all human immunoglobulin heavy chain constant domains numbering is according to the "EU numbering system" (Edelman GM et al, (1969) Proc Natl Acad Sci USA, 63(1): 78-85).
For the human kappa immunoglobulin light chain constant domain (IGKC), numbering is according to the "EU numbering system" (Edelman GM et al., supra).
For the human lambda immunoglobulin light chain constant domains (IGLCl, IGLC2, IGLC3, IGLC6, and IGLC7), 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) Proc Natl Acad Sci USA, 84(24): 9074-8 and Frangione B et al, (1985) Proc Natl Acad Sci 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. In naturally occurring antibodies, the antigen-binding site consists of two variable domains that define specificity: one located in the heavy chain (VH) and the other located in the light chain (VL). In some cases, 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 1 10 amino acids long, and consist of relatively invariant stretches of amino acid sequence called framework regions (FRs) of 15-30 amino acids separated by shorter regions of extreme variability called "hypervariable regions" that are 9-12 amino acids long. The variable domains of native heavy and light chains comprise four FRs, largely adopting a beat-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., supra). 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., supra).
A number of 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., supra). Chothia refers instead to the location of the structural loops (Chothia C & Lesk AM (1987) J. 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) Proc. Natl Acad. Sci. USA, 86: 9268-72; Martin ACR et al, (1991) Methods Enzymol. 203: 121-153; Pedersen JT et al, (1992) Immunomethods, 1 : 126-136; Rees AR et al, (1996) In Sternberg M.J.E. (ed.), Protein Structure Prediction. Oxford University Press, Oxford, 141-172). The contact definition has been recently introduced (MacCallum RM et al., (1996) J. Mol. Biol. 262: 732-745) and is based on an analysis of the available complex structures available in the Protein Databank. The definition of the CDR by 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., (1991) 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(1): 207-9; Lefranc MP (2003) Nucleic Acids Res. 31(1): 307-10; Lefranc MP et al, (2005) Dev. Comp. Immunol. 29(3): 185-203; Kaas Q et al, (2007) Briefings in Functional Genomics & Proteomics, 6(4): 253-64). All Complementarity Determining Regions (CDRs) discussed in the present invention, are defined preferably according to EVIGT®. The variable domain residues for each of these CDRs are as follows (numbering according to Kabat EA, et al., supra): LCDR1 : 27-32, LCDR2: 50-52, LCDR3: 89-97, HCDRl : 26-35, HCDR2: 51-57 and HCDR3: 93-102. The "non-CDR region" of the VL region as used herein comprise the amino acid sequences: 1-26 (FR1), 33-49 (FR2), 53- 88 (FR3), and 98- approximately 107 (FR4). The "non-CDR region" of the VH region as used herein comprise the amino acid sequences: 1 -25 (FR1), 36-50 (FR2), 58-92 (FR3), and 103- approximately 113 (FR4).
The CDRs of the present invention may comprise "extended CDRs" which are based on the aforementioned definitions and have variable domain residues as follows: LCDR1 : 24-36, LCDR2: 46-56, LCDR3:89-97, HCDRl : 26-36, HCDR2:47-65, HCDR3: 93-102. These extended CDRs are numbered as well according to Kabat et al., supra. The "non-extended CDR region" of the VL region as used herein comprise the amino acid sequences: 1-23 (FR1), 37-45 (FR2), 57-88 (FR3), and 98- approximately 107 (FR4). The "non-extended CDR region" of the VH region as used herein comprise the amino acid sequences: 1 -25 (FR1), 37-46 (FR2), 66-92 (FR3), and 103- approximately 113 (FR4).
The term "full length antibody" as used herein includes the structure that constitutes the natural biological form of an antibody, including variable and constant regions. For example, in most mammals, including humans and mice, 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). In some mammals, for example in camels and llamas, IgG antibodies may consist of only two heavy chains, each heavy chain comprising a variable domain attached to the Fc region. Antibody 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 ES et al, (1989) Nature, 341 : 544-546) which consists of a single variable, (v) F(ab')2 fragments, a bivalent fragment comprising two linked Fab fragments (vi) single chain Fv molecules (scFv), wherein a VH domain and a VL domain are linked by a peptide linker which allows the two domains to associate to form an antigen binding site (Bird RE et al, (1988) Science 242: 423-426; Huston JS et al, (1988) Proc. Natl. Acad. Sci. USA, 85: 5879-83), (vii) bispecific single chain Fv dimers (PCT/US92/09965), (viii) "diabodies" or "triabodies", multivalent or multispecific fragments constructed by gene fusion (Tomlinson I & Hollinger P (2000) Methods Enzymol. 326: 461-79; WO94/13804; Holliger P et al, (1993) Proc. Natl. Acad. Sci. USA, 90: 6444-48) and (ix) scFv genetically fused to the same or a different antibody (Coloma MJ & Morrison SL (1997) Nature Biotechnology, 15(2): 159-163).
The term "effector function" as used herein 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). 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, e.g. enhancing or suppressing various reactions of the immune system, with possible beneficial effects in diagnosis and therapy.
As used herein, the term "OX40-mediated disorder" includes conditions such as allergy, asthma, COPD, rheumatoid arthritis, psoriasis and diseases associated with autoimmunity and inflammation.
As used herein, the term "subject" includes any human or nonhuman animal. The term "nonhuman animal" includes all vertebrates, e.g., mammals and non-mammals, such as nonhuman primates, sheep, dogs, cats, horses, cows, chickens, amphibians, reptiles, etc. Preferably the subject is human.
Atopic dermatitis, Atopic dermatitis" (AD), as used herein, means an inflammatory skin disease characterized by intense pruritus (e.g., severe itch) and by scaly and dry eczematous lesions. The term "atopic dermatitis" includes, but is not limited to, AD caused by or associated with epidermal barrier dysfunction, allergy (e.g., allergy to certain foods, pollen, mold, dust mite, animals, etc.), radiation exposure, and/or asthma. The present invention encompasses methods to treat patients with mild, moderate -to-severe or severe AD. As used herein, "moderate-to-severe AD", is characterized by intensely pruritic, widespread skin lesions that are often complicated by persistent bacterial, viral or fungal infections. Moderate -to-severe AD also includes chronic AD in patients. In many cases, the chronic lesions include thickened plaques of skin, lichenification and fibrous papules. Patients affected by moderate-to-severe AD also, in general, have more than 10% of the body's skin affected, or 10% of skin area in addition to involvement of the eyes, hands and body folds. Moderate -to-severe AD is also considered to be present in patients who require frequent treatment with topical corticosteroids. A patient may also be said to have moderate-to -severe AD when the patient is resistant or refractory to treatment by either a topical corticosteroid or a calcineurin inhibitor or any other commonly used therapeutic agent known in the art.
The present invention provides materials and methods for improving one or more Atopic dermatitis efficacy parameter(s) in a subject. Examples of " AD related efficacy parameters " include: (a) Scoring of Atopic Dermatitis -SCORAD (b) Investigators Global Assessment (IGA); (c) Pruritus Numerical rating scale( NRS) (d) Dermatology Life Quality Index-DLQI (e)Body Surface Area (BSA); (f) Eczema Area and Severity Index (EASI); (h) and trans - epidermal water loss( TEWL).An "improvement in an AD- related efficacy parameters " means a decrease from baseline of one or more of IGA, BSA, EASI, SCORAD, TEWL, DLQI or NRS. As used herein, the term "baseline," with regard to an AD- related efficacy parameters, means the numerical value of the AD- related efficacy parameters for a subject prior to or at the time of administration of a pharmaceutical composition of the present invention SCORAD. The SCORAD is a validated tool used in clinical research and clinical practice that was developed to standardize the evaluation of the extent and severity of AD (Dermatology 1993). The extent of AD is assessed as a percentage of each defined body area and reported as the sum of all areas, with a maximum score of 100% (assigned as "A" in the overall SCORAD calculation). The severity of 6 specific symptoms of AD is assessed using the following scale: none (0), mild (1), moderate (2), or severe (3) (for a maximum of 18 total points, assigned as "B" in the overall SCORAD calculation). Subjective assessment of itch and sleeplessness is recorded for each symptom by the patient or relative on a visual analogue scale (VAS), where 0 is no itch (or sleeplessness) and 10 is the worst imaginable itch (or sleeplessness), with a maximum possible score of 20. This parameter is assigned as "C" in the overall SCORAD calculation. The SCORAD is calculated as: A 5 + 7B/2 + C (Kunz et al, 1997).Investigators Global Assessment (IGA);The IGA is an assessment scale used in clinical studies to determine severity of AD and clinical response to treatment based on a 5 -point scale ranging from 0 (clear) to 4 (severe/very severe).
Pruritus Numerical rating scale (NRS)Pruritus Numerical rating scale (NRS): The Pruritus NRS is a single-question assessment tool that is used to assess a subject's worst itch, on a scale of 1 to 10, as a result of AD in the previous 12 hours. Patients will record once daily and respond to the following question, "On a scale of 0 - 10, with 0 being no itch and 10 being the worst itch imaginable, how would you rate your worst degree of itch during the previous 24 hours?" Patient compliance on the pruritus NRS will be followed at each clinic visit.
Dermatology Life Quality Index (DLQI): The DLQI is a simple, patient-administered, 10- question, validated, quality-of-life questionnaire that covers 6 domains including symptoms and feelings, daily activities, leisure, work and school, personal relationships, and treatment. Response categories include "a little," "a lot," and "very much" with corresponding scores of 1 , 2, and 3 respectively and "not at all", "not relevant" responses scored as "0." Totals range from 0 to 30 (less to more impairment) and a 5-point change from baseline is considered clinically relevant (Basra et al, 2008; Finlay et al, 1994).
Body Surface Area (BSA) BSA is assessed for each major section of the body (head, trunk, arms and legs) and is reported as a percentage of all major body sections combined.
Eczema Area and Severity Index (EASI), The EASI is a validated measure used in clinical practice and clinical trials to assess the severity and extent of AD. Four AD disease characteristics will be assessed for severity by the investigator or designee on a scale of "0" (absent) through "3" (severe). In addition, the area of AD involvement will be assessed as a percentage by body area of head, trunk, arms, and legs and converted to a score of 0 to 6 (Hanifin, 2001). Atopic dermatitis biomarker parameters, The present invention also includes methods involving the use, quantification, and analysis of Atopic dermatitis biomarker parameters. As used herein, the term " Atopic dermatitis biomarker parameters " means any biological response, cell type, parameter, protein, polypeptide, enzyme, enzyme activity, metabolite, nucleic acid, carbohydrate, or other biomolecule which is present or detectable in an AD patient at a level or amount that is different from (e.g., greater than or less than) the level or amount of the marker present or detectable in a non-AD patient. In some embodiments, the term "Atopic dermatitis biomarker parameters" includes a biomarker associated with Type 2 helper T-cell Th2)-driven inflammation. In order to evaluate for the drug effect or how much of the disease profile has been reversed by treatment as measured changes in the AD transcriptome using gene arrays consisting of differentially expressed genes between lesional and non lesional AD skin as defined by fold changes (typically a fold change of more than 2)
The AD disease phenotype is the integration of cellular and molecular markers that define the epidermal pathology (hyperplasia, differentiation abnormalities), and Th2, and Th22 immune activation. The changes or reversal of these immune and barrier defects will be assessed by IHC and RT-PCR.
Other exemplary AD-associated biomarkers include a panel of Thl , Th2, Th22 cytokines and chemokines that are shown as elevated in AD blood and to decrease with treatment will be assessed, including but not limited to, e.g., Serum Thymus and activation-regulated chemokine (TARC/CCL17), eotaxin-3, total Immunoglobulin E (IgE), Thymus and activation-regulated chemokine is a chemokine, shown to be strongly associated with disease severity in AD, and may be involved in pathogenesis of the disease. Baseline TARC levels will be assessed for potential predictive value for treatment response. , Eotaxin-3 (CCL26), Eotaxin-3 is a chemokine, shown to be associated with disease severity in AD, and may be involved in pathogenesis of the disease. Baseline eotaxin-3 levels will be assessed for potential predictive value for treatment response. Post-treatment samples will be evaluated for effects of anti OX40 antagonist antibody on eotaxin-3., Total Immunoglobulin E (IgE), Patients with AD often have elevated IgE. Total IgE levels have been found to modestly correlate with AD severity and may be involved in the pathogenesis of the disease. Changes in total IgE reflects not only on AD, but atopy in general. Baseline IgE levels will be assessed for potential predictive value for treatment response. Trans -epidermal water loss( TEWL)., Transepidermal water loss is a skin barrier function test that measures perspiration or water loss through the skin. This procedure involves the non-invasive application of a probe on the surface of the skin on the arm or leg. Affected and non-affected areas of skin will be tested.
Example 1 - IV formulation The inventors have generated and tested a low concentration liquid formulation of an anti-Ox40 antagonist antibody adjusted to pH 6.25, comprising: lOmg/ml of an anti-ox40 antagonist antibody comprising a light chain variable sequence comprising: SEQ ID NO: 9 and a heavy chain variable region comprising SEQ ID NO: 7 or 8; 15mM Histidine; 150mM NaCl;0.01% Tween 80. The inventors have characterised the stability of the anti-Ox40 antagonist 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, Tl 8, T24, T30.In particular the antibody present in the formulation was characterized 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 15 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-SEC analysis so as to determine changes in the antibody in the formulation.Each of the 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 1, 25°C in figure 2 and 40°C in figure 3. Example 2 - T Cell restimulation
T-cell driven autoimmune and inflammatory diseases often display a relapse - remitting pattern. The relapses are driven mainly by reactivation of autoimmune memory T lymphocytes. Therefore inhibiting such memory T cell immune responses represents a very promising therapeutic strategy for autoimmune diseases such as rheumatoid arthritis, systemic lupus erythematosus, multiple sclerosis, psoriasis and others.
Experimental Rationale and Set up - T-cell driven autoimmune and inflammatory diseases often display a relapse - remitting pattern. The relapses are driven mainly by reactivation of autoimmune memory T lymphocytes. Therefore inhibiting such memory T cell immune responses represents a very promising therapeutic strategy for autoimmune diseases such as rheumatoid arthritis, systemic lupus erythematosus, multiple sclerosis, psoriasis and others. Previous data have shown that blocking OX40 with the claimed antibody can potently suppress a T-cell memory response to tetanus toxoid with peripheral blood mononuclear cells from healthy volunteers. Extending that proof of concept to different auto-antigen specific T cell memory responses with cells from autoimmune patients would therefore provide a strong rationale for a broad potential effect of the claimed antibody on different types of autoimmune diseases. , The potency of the claimed antibody to suppress the proliferation of peripheral blood mononuclear cells (PBMCs) from MS, RA, lupus and Uveitis patients, to selected auto-antigens relevant for each disease, was assessed in vitro. The antigen selected for MS was human Myelin Basic protein (MBP) purified from human brain. The antigens selected for RA were citrulinated peptides from aggrecan, vimentin and fibrinogen proteins, as described in Law et al. Arthritis Research & Therapy 2012, 14:R118. The antigen selected for lupus was the SmDl 83-119 peptide as described in Riemekasten et al. Ann Rheum Dis 2002;61 :779-785. The antigen selected for uveitis was the human soluble antigen (S-Ag), as described in Smet et al, Invest Opthalmol Vis Sci. 2001 Dec;42(13):3233-8. Briefly the PBMCs from patients were incubated during 5 to 9 days with or without relevant antigens in presence or not of the claimed or control antibodies. Each condition was performed at least in triplicates. Tritiated thymidine incorporation was measured to quantify the proliferation happening in each condition. The proliferation is described to be a direct consequence of T lymphocyte reactivation to an antigen provided. A stimulation index was calculated for each donor and antigen tested, as the ratio between the condition with the antigen and without antigen. If the stimulation index was superior or equal to 2, the donor was considered as a responder to that antigen.
The effect of the claimed antibody was assessed on all the responders and the results are shown in the table below. Irrespective of the disease type, the claimed antibody potently suppressed the T cell reactivation response to the relevant antigens, in essentially all responders. These observations highlight that the claimed can display an immunosuppressive effect on a broad range of autoimmune antigen-mediated T cell reactivation responses.
Figure imgf000023_0001
Table 1 : Summary of T cell suppression
Example 3 - Antagonistic anti-OX40 antibody suppresses a physiological memory T cell response in an in vitro Tetanus Toxoid re-stimulation assay
The effect of an antagonistic anti-OX40 antibody on a memory T cell response was assessed using an in vitro physiological assay in which the recall response of peripheral blood mononuclear cells (PBMCs) to a known antigen, tetanus toxoid, was tested. For that, PBMCs from donors with historical tetanus vaccinations were cultured in the presence of purified tetanus toxoid (TT) and tritiated-thymidine incorporation (3H-T) was used as a readout of T cell proliferation.
Peripheral blood from healthy volunteers was collected in citrate S-monovette® collection tubes. PBMC isolation was performed by Ficoll density gradient. The PBMCs were washed 3 times with PBS, counted and resuspended into cell culture medium. The PBMCs were then distributed into 96-well U bottom plates at 105 or 2x105 cells/well. TT was added to relevant wells at 0.17 μg/ml final. The plates were incubated for 3-7 days at 37°C in a C02 incubator. The day before harvesting, 0.5 μθι/ννεΐΐ of 3H-T was added to each well and the plates were incubated at 37°C for an additional 15-24 h. The plates were harvested using a Packard filtermate harvester. The filters were dried (50°C 3 h or RT 24h), sealed in a bag with scintillation liquid and immediately read on a Trilux MicroBeta beta counter (Wallac). A stimulation index (SI) was defined as the ratio between the condition "PBMC only" (no TT) and the condition "with TT only". Donors were considered as responders if the SI was superior to 3.
In the first experiment, on the total of 8 donors included, 6 of them showed a positive response to TT (SI>3). The antagonistic anti-OX40 antibody was able to inhibit the proliferative response on 5 out of these 6 donors as shown by Table 2. Only for the donor GDU was considered that the antagonistic anti-OX40 antibody did not produce a significant inhibition as the IgGl control produced a similar effect. Abatacept also inhibited the response on 4 out of these 6 donors. When considering the maximum response or the concentration at which the treatments did lost efficacy, the antagonistic anti-OX40 antibody showed a superior efficacy to abatacept on 3 out of the 6 responders (VDU, SHO and SLE).
This experiment was reproduced on 7 of the donors tested in the first experiment plus 3 additional donors. In this second experiment, only 3 donors were considered as responders (SI>3, Table 3 in appendix 3) and the antagonistic anti-OX40 antibody did inhibit the TT response on all of them. Similarly to the first experiment, the antagonistic anti-OX40 antibody on the donor SHO did produce a better inhibition than abatacept. On the 2 new responders (ASI and ROL), the antagonistic anti-OX40 antibody also showed a better inhibition than abatacept on one of them.
These data indicate that targeting OX40 with the antagonistic anti-OX40 antibody can potently inhibit a physiologic memory T helper cell response. Additionally the comparison with CD28 blockade (abatacept) suggests that the claimed antibody may be more potent than abatacept to supress T cell memory responses.
abatacept antagonistic anti-OX40 antibody
Experiment # Donor ID Maximum Minimal Maximum Minimal active effect (%) active effect (%) concentration concentration SLE 76 4 80 0.8
SHO 14 20 51 <0.8
EXP 1. FAB 79 0.8 87 0.8
GDU NI NA NI NA
VDU NI NA 67 4
SZA 100 4 99 4
SHO 50 0.16 87 <0.16
EXP 2.
ASI 100 >0.16 100 <0.16
ROL 61 0.16 83 <0.16
Table 2: Summary of the effect of an antagonistic anti-OX40 antibody and abatacept on TT recall response
Table Legend: The table shows the maximum blocking effect (%) and the approximate minimal active concentration values for each responder defined by a stimulation index (SI)>3. SI was calculated as the ratio between the condition "with TT only" and the condition "PBMC only" (no TT). Each donor is represented by a 3 letter code. NI: no inhibition observed compared to IgG control. NA: not applicable.
Example 4 - Phase la clinical trial to establish safety and tolerability of antagonistic anti- OX40 antibody A phase la clinical study was undertaken to evaluate the safety and tolerability of single ascending doses of an antagonistic anti-OX40 antibody (0.3, 1.0, 3.0 and 10.0 mg kg) in healthy volunteers (HVs). Subject disposition: 32 subjects were enrolled to receive single IV doses of the antagonistic anti- OX40 antibody (24 HVs) or matching placebo (8 HVs). All subjects completed the study and were included in the final analysis for PK and safety analysis.
Safety data: In the study 69% of subjects experienced at least one TEAE. There were no meaningful differences in the frequency (63% in placebo in 71% in antibody) of TEAEs between the study groups. None of these TEAEs were related. There were two cases of tooth abscess seen in the study and were reported as AEs of moderate intensity. One of this was seen in placebo group and one was reported in the antibody 10.0 mg/kg group. Other than this, there were no other AEs of moderate intensity. There were no deaths or other serious and significant AEs in this part of the study.
Conclusion: The antagonistic anti-OX40 antibody is safe and well tolerated in this study.
Example 5 - Pharmacokinetic data from Phase lb, Randomized, Placebo-controlled, Single Ascending Dose Study in Adult Healthy Volunteers
Serum samples were collected at scheduled time points uptolO weeks post dosing. The concentrations of the claimed antagonistic anti-OX40 antibody in serum were estimated using a validated ELISA method (LLOQ: 97.7 ng/mL).The interim serum concentration data along with nominal sampling time points for individual subjects (blinded by alias codes) were subjected to non-compartmental analysis using Phoenix® WinNonlin version 6.4. The summary of PK parameters is presented in Table 3.
Part 1 Part 2
PK
Unit 0.3 mg/kg 1 mg/kg 3 mg/kg 10 mg/kg 10 mg/kg parameter
N=6 N=6 N=6 N=6 N=10
94.7 270.9
Cmax ^g/mL) 9.5 (26.6) 33.8 (11.8) 339 (22)
(12.9) (1 1.4)
1 max (h) 2 (1-6) 4 (1 -6) 1.75 (1 -8) 2 (1.5-4) 2 (1.5-4)
AUCo-tlast ^g.h/mL) 1957.1 8022.9 24650.8 80637.4 70429.1
Figure imgf000027_0001
Summary of PK parameters following single IV infusion (1 hr) in healthy
Following single TV infusion, maximum serum concentrations were achieved at a median Tmax ranging from 1.75 to 4 hours. After Tmax, the serum concentrations generally declined in a bi- exponential manner with a short distribution phase followed by a longer terminal elimination phase. The antibody showed slow clearance and low volume of distribution, consistent with the general characteristics of monoclonal antibodies. The terminal elimination half-life ranged from 10 to 14 days, and appeared to be independent of dose. The PK profiles at 10 mg kg appeared comparable between Part 1 and Part 2. Both Cmax and AUCo-inf appeared to be nearly dose proportional across the tested dose range. Overall low to moderate inter-subject variability in PK parameters was observed. The data indicated that both CmaX and AUCo-inf increased in a near dose proportional manner from 0.3 to 10 mg/kg. There was no dose dependency in clearance, volume of distribution and terminal half-lives. The claimed antibody therefore possesses in human a PK profile characteristic of a human IgGl monoclonal antibody, with minimal target-mediated disposition. Altogether, these
pharmacokinetic characteristics validate further clinical studies with a dose dependent exposure of patients.

Claims

Claims
1. An anti-OX40 antagonist antibody for use in the treatment or prevention of atopic dermatitis, including moderate -to -severe atopic dermatitis, wherein said anti-OX40 antagonist antibody comprises: a heavy chain CDR1 comprising the amino acid sequence of SEQ ID NO: 1 , and/or a heavy chain CDR2 comprising the amino acid sequence of SEQ ID NO: 2, and/or a heavy chain CDR3 comprising the amino acid sequence of SEQ ID NO: 3; and/or comprising a light chain CDR1 comprising the amino acid sequence of SEQ ID NO: 4, and/or a light chain CDR2 comprising the amino acid sequence of SEQ ID NO: 5 and/or a light chain CDR3 comprising the amino acid sequence of SEQ ID NO: 6.
2. The antibody according to claim 1 wherein said antibody is administered at a dose of between 10 to 40 mg/kg, of the patient body weight.
3. The antibody according to claim 1 or 2 wherein said dose is administered to a patient at an interval of daily, weekly biweekly, monthly, bimonthly or any suitable interval in between.
4. A stable aqueous pharmaceutical formulation comprising an anti-OX40 antagonist antibody comprising: a heavy chain CDR1 comprising the amino acid sequence of SEQ ID NO: 1 , and/or a heavy chain CDR2 comprising the amino acid sequence of SEQ ID NO: 2, and/or a heavy chain CDR3 comprising the amino acid sequence of SEQ ID NO: 3; and/or comprising a light chain CDR1 comprising the amino acid sequence of SEQ ID NO: 4, and/or a light chain CDR2 comprising the amino acid sequence of SEQ ID NO: 5 and/or a light chain CDR3 comprising the amino acid sequence of SEQ ID NO: 6, wherein said stable aqueous pharmaceutical formulation comprises a therapeutically effective amount of said anti-ox40 antagonist antibody, a buffer, a surfactant and a tonicifying agent, and wherein the pH of the formulation is from about 5 to about 7.
5. The stable aqueous pharmaceutical formulation according to claim 4, wherein said anti- OX40 antagonist antibody is present at a concentration of at least 0.1 mg/ml.
6. The stable aqueous pharmaceutical formulation according to claim 4, wherein said buffer is selected from the group comprising: citrate, acetate, histidine, phosphate, tris (tris(hydroxymethyl)aminomethane).
7. The stable aqueous pharmaceutical formulation according to claim 4, wherein said stabilizing or tonicifying agent 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.
8. The stable aqueous pharmaceutical formulation according to claim 4, wherein said surfactant is selected from the group comprising: tween 20/40/80, polysorbate 20/80, poloxamer, sodium lauryl sulphate.
9. The stable aqueous pharmaceutical formulation according to claim 4, wherein said pH is
6.25.
10. The stable aqueous pharmaceutical formulation according to claim 4, comprising: lOmg/ml of an anti-OX40 antagonist antibody; 15mM histidine; 150mM NaCl; 0.01% tween 80.
PCT/EP2017/052504 2016-02-04 2017-02-06 Anti-ox40 antagonistic antibodies for the treatment of atopic dermatitis WO2017134292A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
EP16154324 2016-02-04
EPEP16154324.4 2016-02-04

Publications (1)

Publication Number Publication Date
WO2017134292A1 true WO2017134292A1 (en) 2017-08-10

Family

ID=55300423

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2017/052504 WO2017134292A1 (en) 2016-02-04 2017-02-06 Anti-ox40 antagonistic antibodies for the treatment of atopic dermatitis

Country Status (1)

Country Link
WO (1) WO2017134292A1 (en)

Cited By (26)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20180044428A1 (en) * 2015-03-11 2018-02-15 Providence Health & Services-Oregon Compositions and methods for enhancing the efficacy of cancer therapy
WO2019089921A1 (en) 2017-11-01 2019-05-09 Bristol-Myers Squibb Company Immunostimulatory agonistic antibodies for use in treating cancer
WO2019191133A1 (en) 2018-03-27 2019-10-03 Bristol-Myers Squibb Company Real-time monitoring of protein concentration using ultraviolet signal
WO2019219978A1 (en) * 2018-05-18 2019-11-21 Glenmark Pharmaceuticals S.A. Anti-ox40 antagonistic antibodies for the treatment of autoimmune diseases
WO2019229155A1 (en) * 2018-05-31 2019-12-05 Glenmark Pharmceuticals S.A. Anti-ox40 antagonistic antibodies and dosage for the treatment of ox40-mediated disorders
WO2020014583A1 (en) 2018-07-13 2020-01-16 Bristol-Myers Squibb Company Ox-40 agonist, pd-1 pathway inhibitor and ctla-4 inhibitor combination for use in a mehtod of treating a cancer or a solid tumor
WO2020016417A1 (en) * 2018-07-19 2020-01-23 Ichnos Sciences S.A. Liquid antibody formulation
WO2020172658A1 (en) 2019-02-24 2020-08-27 Bristol-Myers Squibb Company Methods of isolating a protein
WO2020237221A1 (en) 2019-05-23 2020-11-26 Bristol-Myers Squibb Company Methods of monitoring cell culture media
WO2020243563A1 (en) 2019-05-30 2020-12-03 Bristol-Myers Squibb Company Multi-tumor gene signatures for suitability to immuno-oncology therapy
WO2020243570A1 (en) 2019-05-30 2020-12-03 Bristol-Myers Squibb Company Cell localization signature and combination therapy
WO2020243568A1 (en) 2019-05-30 2020-12-03 Bristol-Myers Squibb Company Methods of identifying a subject suitable for an immuno-oncology (i-o) therapy
EP3766481A1 (en) * 2019-07-19 2021-01-20 Ichnos Sciences SA Liquid antibody formulation
WO2021013689A1 (en) * 2019-07-19 2021-01-28 Ichnos Sciences SA Lyophilized antibody formulation
US11242393B2 (en) 2018-03-23 2022-02-08 Bristol-Myers Squibb Company Antibodies against MICA and/or MICB and uses thereof
WO2022047412A1 (en) 2020-08-31 2022-03-03 Bristol-Myers Squibb Company Cell localization signature and immunotherapy
WO2022076318A1 (en) 2020-10-05 2022-04-14 Bristol-Myers Squibb Company Methods for concentrating proteins
WO2022120179A1 (en) 2020-12-03 2022-06-09 Bristol-Myers Squibb Company Multi-tumor gene signatures and uses thereof
WO2022146948A1 (en) 2020-12-28 2022-07-07 Bristol-Myers Squibb Company Subcutaneous administration of pd1/pd-l1 antibodies
WO2022146947A1 (en) 2020-12-28 2022-07-07 Bristol-Myers Squibb Company Antibody compositions and methods of use thereof
WO2022212876A1 (en) 2021-04-02 2022-10-06 The Regents Of The University Of California Antibodies against cleaved cdcp1 and uses thereof
WO2023061424A1 (en) * 2021-10-15 2023-04-20 Inmagene Biopharmaceuticals (Hangzhou) Co., Ltd. Pharmaceutical formulation comprising anti-ox40 monoclonal antibody
WO2023173011A1 (en) 2022-03-09 2023-09-14 Bristol-Myers Squibb Company Transient expression of therapeutic proteins
WO2023178329A1 (en) 2022-03-18 2023-09-21 Bristol-Myers Squibb Company Methods of isolating polypeptides
WO2023235847A1 (en) 2022-06-02 2023-12-07 Bristol-Myers Squibb Company Antibody compositions and methods of use thereof
WO2024054992A1 (en) 2022-09-09 2024-03-14 Bristol-Myers Squibb Company Methods of separating chelator

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2008106116A2 (en) * 2007-02-27 2008-09-04 Genentech, Inc. Antagonist ox40 antibodies and their use in the treatment of inflammatory and autoimmune diseases
WO2010096418A2 (en) * 2009-02-17 2010-08-26 Ucb Pharma S.A. Antibody molecules having specificity for human ox40
WO2013008171A1 (en) * 2011-07-11 2013-01-17 Glenmark Pharmaceuticals S.A. Antibodies that bind to ox40 and their uses

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2008106116A2 (en) * 2007-02-27 2008-09-04 Genentech, Inc. Antagonist ox40 antibodies and their use in the treatment of inflammatory and autoimmune diseases
WO2010096418A2 (en) * 2009-02-17 2010-08-26 Ucb Pharma S.A. Antibody molecules having specificity for human ox40
WO2013008171A1 (en) * 2011-07-11 2013-01-17 Glenmark Pharmaceuticals S.A. Antibodies that bind to ox40 and their uses

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
ANONYMOUS: "Press Release For Immediate Dissemination", 3 September 2015 (2015-09-03), pages 1 - 3, XP055362667, Retrieved from the Internet <URL:http://www.glenmark.de/files/dokumente/pressemitteilungen/Press_Release_Glenmarks_novel_monoclonal_antibody_GBR_830_to_enter_Phase_2_clinical_studies_in_Atopic_Dermatitis_and_Celiac_Disease_in_US_and_Europe_.pdf> [retrieved on 20170406] *
JONATHAN BACK ET AL: "GBR 830: An antagonistic anti-human OX40 mAb with potent suppressive effect on pathological immune responses", 1 January 2015 (2015-01-01), XP055363012, Retrieved from the Internet <URL:http://www.glenmarkpharma.com/sites/default/files/GBR%20830%20-%20Jonathan%20%20PEGS%20Boston%202014.pdf> [retrieved on 20170407] *
T. ILVES ET AL: "OX40 ligand and OX40 are increased in atopic dermatitis lesions but do not correlate with clinical severity", JEADV. JOURNAL OF THE EUROPEAN ACADEMY OF DERMATOLOGY AND VENEREOLOGY., vol. 27, no. 2, 1 February 2013 (2013-02-01), NL, pages e197 - e205, XP055319135, ISSN: 0926-9959, DOI: 10.1111/j.1468-3083.2012.04587.x *

Cited By (28)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20180044428A1 (en) * 2015-03-11 2018-02-15 Providence Health & Services-Oregon Compositions and methods for enhancing the efficacy of cancer therapy
WO2019089921A1 (en) 2017-11-01 2019-05-09 Bristol-Myers Squibb Company Immunostimulatory agonistic antibodies for use in treating cancer
US11242393B2 (en) 2018-03-23 2022-02-08 Bristol-Myers Squibb Company Antibodies against MICA and/or MICB and uses thereof
WO2019191133A1 (en) 2018-03-27 2019-10-03 Bristol-Myers Squibb Company Real-time monitoring of protein concentration using ultraviolet signal
WO2019219978A1 (en) * 2018-05-18 2019-11-21 Glenmark Pharmaceuticals S.A. Anti-ox40 antagonistic antibodies for the treatment of autoimmune diseases
WO2019229155A1 (en) * 2018-05-31 2019-12-05 Glenmark Pharmceuticals S.A. Anti-ox40 antagonistic antibodies and dosage for the treatment of ox40-mediated disorders
WO2020014583A1 (en) 2018-07-13 2020-01-16 Bristol-Myers Squibb Company Ox-40 agonist, pd-1 pathway inhibitor and ctla-4 inhibitor combination for use in a mehtod of treating a cancer or a solid tumor
WO2020016417A1 (en) * 2018-07-19 2020-01-23 Ichnos Sciences S.A. Liquid antibody formulation
WO2021013372A1 (en) * 2018-07-19 2021-01-28 Ichnos Sciences SA Liquid antibody formulation
WO2021013385A1 (en) * 2018-07-19 2021-01-28 Ichnos Sciences SA Processes for obtaining a highly concentrated antibody solution
WO2020172658A1 (en) 2019-02-24 2020-08-27 Bristol-Myers Squibb Company Methods of isolating a protein
WO2020237221A1 (en) 2019-05-23 2020-11-26 Bristol-Myers Squibb Company Methods of monitoring cell culture media
WO2020243563A1 (en) 2019-05-30 2020-12-03 Bristol-Myers Squibb Company Multi-tumor gene signatures for suitability to immuno-oncology therapy
WO2020243570A1 (en) 2019-05-30 2020-12-03 Bristol-Myers Squibb Company Cell localization signature and combination therapy
WO2020243568A1 (en) 2019-05-30 2020-12-03 Bristol-Myers Squibb Company Methods of identifying a subject suitable for an immuno-oncology (i-o) therapy
WO2021013689A1 (en) * 2019-07-19 2021-01-28 Ichnos Sciences SA Lyophilized antibody formulation
EP3766481A1 (en) * 2019-07-19 2021-01-20 Ichnos Sciences SA Liquid antibody formulation
WO2022047412A1 (en) 2020-08-31 2022-03-03 Bristol-Myers Squibb Company Cell localization signature and immunotherapy
WO2022076318A1 (en) 2020-10-05 2022-04-14 Bristol-Myers Squibb Company Methods for concentrating proteins
WO2022120179A1 (en) 2020-12-03 2022-06-09 Bristol-Myers Squibb Company Multi-tumor gene signatures and uses thereof
WO2022146948A1 (en) 2020-12-28 2022-07-07 Bristol-Myers Squibb Company Subcutaneous administration of pd1/pd-l1 antibodies
WO2022146947A1 (en) 2020-12-28 2022-07-07 Bristol-Myers Squibb Company Antibody compositions and methods of use thereof
WO2022212876A1 (en) 2021-04-02 2022-10-06 The Regents Of The University Of California Antibodies against cleaved cdcp1 and uses thereof
WO2023061424A1 (en) * 2021-10-15 2023-04-20 Inmagene Biopharmaceuticals (Hangzhou) Co., Ltd. Pharmaceutical formulation comprising anti-ox40 monoclonal antibody
WO2023173011A1 (en) 2022-03-09 2023-09-14 Bristol-Myers Squibb Company Transient expression of therapeutic proteins
WO2023178329A1 (en) 2022-03-18 2023-09-21 Bristol-Myers Squibb Company Methods of isolating polypeptides
WO2023235847A1 (en) 2022-06-02 2023-12-07 Bristol-Myers Squibb Company Antibody compositions and methods of use thereof
WO2024054992A1 (en) 2022-09-09 2024-03-14 Bristol-Myers Squibb Company Methods of separating chelator

Similar Documents

Publication Publication Date Title
WO2017134292A1 (en) Anti-ox40 antagonistic antibodies for the treatment of atopic dermatitis
JP7254699B2 (en) Anti-BCMA heavy chain only antibody
JP7303126B2 (en) Anti-BCMA heavy chain only antibody
JP7240335B2 (en) Anti-BCMA heavy chain only antibody
KR102194568B1 (en) Anti-mcam antibodies and associated methods of use
AU2018311055A1 (en) Anti-TREM2 antibodies and methods of use thereof
KR20160065981A (en) Use of semaphorin-4d binding molecules for treating neurodegenerative disorders
US10918716B2 (en) Triple combination therapy for treating Crohn&#39;s disease
DK2970467T3 (en) ANTI-CD52 ANTIBODIES
EP3814377A2 (en) Anti-sirp-beta1 antibodies and methods of use thereof
JP2022529340A (en) Antibodies targeting human CD47
CA3158565A1 (en) Methods of use of anti-trem2 antibodies
CN111263771A (en) anti-CD 40 antibodies for the treatment of sjogren&#39;s syndrome
US20230272097A1 (en) Therapeutic antibodies
US20200002422A1 (en) Method of preventing graft versus host disease
RU2816994C2 (en) Bcma heavy chain antibodies
US11746154B2 (en) CD1a antibodies and uses thereof
US20230365681A1 (en) Bispecific antibody treatment of lymphoid malignant neoplasm conditions
KR20240026185A (en) Anti-CD40 antibodies, antigen-binding fragments and medical uses thereof
KR20230154239A (en) Anti-CD38 antibody for use in the treatment of antibody-mediated transplant rejection
KR20230045613A (en) SARS-COV-2 Antibodies for Treatment and Prevention of COVID-19
TW202311293A (en) Combinations of immunotherapies and uses thereof
NZ762893B2 (en) Binding molecules specific for il-21 and uses thereof

Legal Events

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

Ref document number: 17703960

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 17703960

Country of ref document: EP

Kind code of ref document: A1