WO2022207785A1 - Anticorps dirigés contre gfral - Google Patents

Anticorps dirigés contre gfral Download PDF

Info

Publication number
WO2022207785A1
WO2022207785A1 PCT/EP2022/058556 EP2022058556W WO2022207785A1 WO 2022207785 A1 WO2022207785 A1 WO 2022207785A1 EP 2022058556 W EP2022058556 W EP 2022058556W WO 2022207785 A1 WO2022207785 A1 WO 2022207785A1
Authority
WO
WIPO (PCT)
Prior art keywords
seq
domain
quel
antibody
gfral
Prior art date
Application number
PCT/EP2022/058556
Other languages
English (en)
Inventor
E-Chiang Lee
Hui Liu
Adam Carpenter
Original Assignee
Kymab Limited
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from GBGB2104553.9A external-priority patent/GB202104553D0/en
Priority claimed from GBGB2107330.9A external-priority patent/GB202107330D0/en
Application filed by Kymab Limited filed Critical Kymab Limited
Publication of WO2022207785A1 publication Critical patent/WO2022207785A1/fr

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2863Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against receptors for growth factors, growth regulators
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/21Immunoglobulins specific features characterized by taxonomic origin from primates, e.g. man
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/30Immunoglobulins specific features characterized by aspects of specificity or valency
    • C07K2317/33Crossreactivity, e.g. for species or epitope, or lack of said crossreactivity
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/76Antagonist effect on antigen, e.g. neutralization or inhibition of binding
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/90Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
    • C07K2317/92Affinity (KD), association rate (Ka), dissociation rate (Kd) or EC50 value

Definitions

  • This invention relates to antibodies that bind to and inhibit the activity of glial cell-derived neurotrophic factor receptor alpha like (GFRAL) protein.
  • GFRAL glial cell-derived neurotrophic factor receptor alpha like
  • the cell surface receptor known as "glial cell line-derived neurotrophic factor family receptor alpha like” is encoded by the GFRAL gene which is located on human chromosome 6p12.1 and has 9 exons encoding a sequence of 394 amino acids.
  • GFRAL transcripts have been found primarily in the central nervous system. Its expression was found to be restricted to certain neurons of the brainstem, specifically in the area postrema (AP) and the nucleus of the solitary tract (nucleus tractus solitaries, NTS), constituting part of an emergency circuit that controls feeding behaviour in response to stressful conditions. Later on, GFRAL expression was also detected at low levels in testis and adipose tissue [2, 3, 4, 5].
  • the ligand for GFRAL has been identified as the hormone GDF15 (growth and differentiation factor 15) [2, 3, 4, 5, 6].
  • GDF15 growth and differentiation factor 15
  • RET a tyrosine protein-kinase receptor
  • MAPK a tyrosine protein-kinase receptor
  • AKT a tyrosine protein-kinase receptor
  • PLC-y signalling pathways This signalling has been shown to have the downstream effect of regulating food intake, energy expenditure and body weight [2, 3, 4, 5].
  • the active signalling complex is an assembly of six polypeptides, comprising a homodimer of GDF15-GFRAL-RET.
  • GDF15 is a distant member of the TGF-b family. It is a stress-induced cytokine and can be upregulated by tissue injury [7], ionising radiation [8], hypoxia [9], inflammation [10], chemical toxins [11 , 12] and various disease states, including cancer [13], rheumatoid arthritis [14], chronic renal failure [15], cardiovascular diseases [16], obesity and diabetes [17] It is also induced by tissue-damaging toxins such as chemotherapeutic agents [18, 19].
  • GDF15 is believed to represent a “sentinel” hormone response to cellular stress, its protective effects including limiting systemic exposure to recently ingested toxins (e.g., through emesis) and, through its induction of conditioned aversion, promoting the avoidance of future exposures to agents which have previously led to cellular stress [11].
  • GDF15 High levels of GDF15 are linked with cachexia in cancer patients.
  • the presence of cancer in the body may increase the level of GDF15, driving cancer cachexia.
  • Cachexia is a multifactorial disease characterised by weight loss via skeletal muscle and adipose tissue loss, an imbalance in metabolic regulation, and reduced food intake [22]
  • Cachexia is estimated to affect up to 74% of patients with many types of cancer globally, with the highest incidence in head and neck, pancreatic, gastric, and hepatic cancer [23].
  • Cancer cachexia not only negatively affects the quality of life of patients with cancer [24, 25], but also reduces the effectiveness of anti-cancer chemotherapy [26, 27] and increases its toxicity [28, 29, 30], leading to increased cancer-related mortality [30, 31 , 32] and increased expenditure of medical resources.
  • a range of conditions other than cancer are also associated with cachexia, including pulmonary and cardiac conditions (e.g., congestive heart failure, chronic obstructive pulmonary disease), as well as chronic kidney disease, acquired immune deficiency syndrome (AIDS), and in the advanced states of cystic fibrosis, multiple sclerosis, motor neuron disease, Parkinson's disease, dementia, tuberculosis, multiple system atrophy, mercury poisoning, Crohn's disease, rheumatoid arthritis and celiac disease.
  • pulmonary and cardiac conditions e.g., congestive heart failure, chronic obstructive pulmonary disease
  • chronic kidney disease e.g., chronic kidney disease
  • AIDS acquired immune deficiency syndrome
  • GFRAL-/- mice on a chow diet appear normal, and they become obese on a high-fat diet just like wild-type mice.
  • Wild-type mice treated with GDF15 exhibit significantly attenuated food intake and sustained weight loss in comparison to vehicle-treated mice, whereas GFRAL-/- mice are refractory to the effects of GDF15 [3, 4] This illustrates the suppression of body weight gain by GDF15-GFRAL signalling.
  • GDF15-transgenic mice GDF15 overexpression has been shown to protect mice from the development of obesity and improve their glucose tolerance on a high-fat diet [13, 36]. In mice or rats that are fed chow or high-fat diet, GDF15 administration reduces food intake and body weight.
  • Cynomolgus monkeys with spontaneous obesity show decreased food intake resulting in significant weight loss after 4 weeks of exposure to recombinant HAS-GDF15 [2, 3, 4, 5, 6].
  • serum GDF15 level was positively correlated with tumour volume and negatively correlated with food intake, and it was shown that GDF15 induces anorexia through nausea and emesis [37, 38].
  • GDF15-GFRAL-RET complex regulates food intake and body weight, and its activity plays an important role in conditions such as cancer cachexia, anorexia and hyperemesis gravidarum.
  • antagonists offer a route to therapeutic treatment of these and other conditions.
  • the present invention provides antibodies to human GFRAL.
  • Antibody binding to the GFRAL extracellular domain may inhibit formation of the GDF15-GFRAL-RET complex, e.g., by inhibiting association of RET with GDF15-GFRAL.
  • Exemplary antibodies of the present invention are named QUEL-0101 , QUEL-0201 and QUEL-0301. Further, related antibodies of the present invention are named QUEL-0102, QUEL- 0103, QUEL-0104, QUEL-0105, QUEL-0302, QUEL-0303 and QUEL-0304.
  • the present invention extends to anti-GFRAL binding molecules incorporating antigen-binding sequences of QUEL-0101 , QUEL-0201 or QUEL-0301 , or of any of QUEL-0102, QUEL-0103, QUEL-0104, QUEL-0105, QUEL-0302, QUEL-0303 or QUEL-0304, such as their complementarity determining regions (CDRs), e.g., heavy chain CDRs (FICDRs) FICDR1 , FICDR2 and/or FICDR3 and/or light chain CDRs (LCDRs) LCDR1 , LCDR2 and/or LCDR3 and optionally the heavy and/or light chain variable (VFI and/or VL) domain, and variants thereof.
  • CDRs complementarity determining regions
  • a binding molecule may comprise the HCDR1 , HCDR2 and/or HCDR3 of QUEL-0101 , QUEL-0102, QUEL-0103, QUEL- 0104, QUEL-0105, QUEL-0201 , QUEL-0301 , QUEL-0302, QUEL-0303 or QUEL-0304 in a polypeptide scaffold and/or it may comprise the LCDR1 , LCDR2 and/or LCDR3 of QUEL-0101 , QUEL-0102, QUEL-0103, QUEL-0104, QUEL-0105, QUEL-0201 , QUEL-0301 , QUEL-0302, QUEL-0303 or QUEL-0304 in a polypeptide scaffold.
  • the binding molecule may comprise the HCDR3 of QUEL-0201 , optionally the QUEL-0201 set of HCDRs, and optionally it may comprise the HCDRs and LCDRs of QUEL
  • Embodiments of the invention include antibodies comprising a VH domain and a VL domain, the VH domain comprising a set of HCDRs HCDR1 , HCDR2 and HCDR3 and the VL domain comprising a set of LCDRs LCDR1 , LCDR2 and LCDR3, wherein
  • the set of HCDRs is the QUEL-0101 set of HCDRs defined wherein HCDR1 is SEQ ID NO: 3, HCDR2 is SEQ ID NO: 4 and HCDR3 is SEQ ID NO: 5,
  • the set of HCDRs is the QUEL-0201 set of HCDRs defined wherein HCDR1 is SEQ ID NO: 13, HCDR2 is SEQ ID NO: 14 and HCDR3 is SEQ ID NO: 15, or
  • the set of HCDRs is the QUEL-0301 set of HCDRs defined wherein HCDR1 is SEQ ID NO: 23, HCDR2 is SEQ ID NO: 24 and HCDR3 is SEQ ID NO: 25.
  • FIG. 1 For embodiments of the invention, antibodies comprising a VH domain and a VL domain, the VH domain comprising a set of HCDRs HCDR1 , HCDR2 and HCDR3 and the VL domain comprising a set of LCDRs LCDR1 , LCDR2 and LCDR3, wherein
  • the set of HCDRs is the QUEL-0102 set of HCDRs defined wherein HCDR1 is SEQ ID NO: 99, HCDR2 is SEQ ID NO: 100 and HCDR3 is SEQ ID NO: 101 ,
  • the set of HCDRs is the QUEL-0103 set of HCDRs defined wherein HCDR1 is SEQ ID NO: 107, HCDR2 is SEQ ID NO: 108 and HCDR3 is SEQ ID NO: 5,
  • the set of HCDRs is the QUEL-0104 set of HCDRs defined wherein HCDR1 is SEQ ID NO: 107, HCDR2 is SEQ ID NO: 108 and HCDR3 is SEQ ID NO: 113, or
  • the set of HCDRs is the QUEL-0105 set of HCDRs defined wherein HCDR1 is SEQ ID NO: 118, HCDR2 is SEQ ID NO: 108 and HCDR3 is SEQ ID NO: 119.
  • FIG. 1 For embodiments of the invention, antibodies comprising a VH domain and a VL domain, the VH domain comprising a set of HCDRs HCDR1 , HCDR2 and HCDR3 and the VL domain comprising a set of LCDRs LCDR1 , LCDR2 and LCDR3, wherein
  • the set of HCDRs is the QUEL-0302 set of HCDRs defined wherein HCDR1 is SEQ ID NO: 23, HCDR2 is SEQ ID NO: 124 and HCDR3 is SEQ ID NO: 125,
  • the set of HCDRs is the QUEL-0303 set of HCDRs defined wherein HCDR1 is SEQ ID NO: 133, HCDR2 is SEQ ID NO: 134 and HCDR3 is SEQ ID NO: 135, or
  • the set of HCDRs is the QUEL-0304 set of HCDRs defined wherein HCDR1 is SEQ ID NO: 133, HCDR2 is SEQ ID NO: 142 and HCDR3 is SEQ ID NO: 143.
  • An antibody of the present invention may comprise a VH domain and a VL domain, the VH domain comprising a set of HCDRs HCDR1 , HCDR2 and HCDR3, and the VL domain comprising a set of LCDRs LCDR1 , LCDR2 and LCDR3, wherein HCDR1 is SEQ ID NO: 3, SEQ ID NO: 99, SEQ ID NO: 107 or SEQ ID NO: 118, HCDR2 is SEQ ID NO: 4, SEQ ID NO: 100 or SEQ ID NO: 108 and HCDR3 is SEQ ID NO: 5, SEQ ID NO: 101 , SEQ ID NO: 113 or SEQ ID NO: 119, and optionally wherein
  • LCDR1 is SEQ ID NO: 8
  • LCDR2 is SEQ ID NO: 9 and
  • LCDR3 is SEQ ID NO: 10 or SEQ ID NO: 104.
  • An antibody of the present invention may comprise a VH domain and a VL domain, the VH domain comprising the QUEL-0101 set of HCDRs HCDR1 SEQ ID NO: 3, HCDR2 SEQ ID NO: 4 and HCDR3 SEQ ID NO: 5, and the VL domain comprising the QUEL-0101 set of LCDRs LCDR1 SEQ ID NO: 8, LCDR2 SEQ ID NO: 9 and LCDR3 SEQ ID NO: 10.
  • the antibody comprises a VH domain and a VL domain, the VH domain comprising the QUEL-0102 set of HCDRs HCDR1 SEQ ID NO: 99, HCDR2 SEQ ID NO: 100 and HCDR3 SEQ ID NO: 101 , and the VL domain comprising the QUEL-0102 set of LCDRs LCDR1 SEQ ID NO: 8, LCDR2 SEQ ID NO: 9 and LCDR3 SEQ ID NO: 104.
  • the antibody comprises a VH domain and a VL domain, the VH domain comprising the QUEL-0103 set of HCDRs HCDR1 SEQ ID NO: 107, HCDR2 SEQ ID NO: 108 and HCDR3 SEQ ID NO: 5, and the VL domain comprising the QUEL-0103 set of LCDRs LCDR1 SEQ ID NO: 8, LCDR2 SEQ ID NO: 9 and LCDR3 SEQ ID NO: 104.
  • the antibody comprises a VH domain and a VL domain, the VH domain comprising the QUEL-0104 set of HCDRs HCDR1 SEQ ID NO: 107, HCDR2 SEQ ID NO: 108 and HCDR3 SEQ ID NO: 113, and the VL domain comprising the QUEL-0104 set of LCDRs LCDR1 SEQ ID NO: 8, LCDR2 SEQ ID NO: 9 and LCDR3 SEQ ID NO: 104.
  • An amino acid alteration may be a substitution, deletion or insertion of an amino acid. Such alterations may optionally be in the variable domain framework, outside the CDRs. For example, there may be one or two amino acid alterations in the VH and/or VL domain framework. Alterations may be conservative substitutions and/or may represent germlining of framework residues.
  • an amino acid residue means to revert it to the residue that occurs at that position as encoded by the germline gene segment from which the variable domain was produced.
  • Corresponding germline gene segments may be identified by comparing the variable domain sequence against human v, d and j (for VH) or human v and j (for VL) gene segments and identifying the closest match.
  • Variants of the QUEL- 0101 VH domain include VH domains that have one or more alterations that are present in the QUEL-0102, QUEL-0103, QUEL-0104 or QUEL-0105 VH domain, e.g., that are present in the framework of one or more of these VH domains.
  • An antibody of the present invention optionally comprises the QUEL-0102 VH domain SEQ ID NO: 98, the QUEL-0103 VH domain SEQ ID NO: 106, the QUEL-0104 VH domain SEQ ID NO: 112 or the QUEL-0105 VH domain SEQ ID NO: 117.
  • the VH domain may be encoded by a nucleotide sequence produced by recombination of heavy chain v gene segment IGHV3-30 (e.g., IGHV3-30 * 18) with a d gene segment and a heavy chain j gene segment, e.g., IGHJ6 (e.g., IGHJ6 * 02).
  • IGHV6 heavy chain j gene segment
  • it may comprise a VH domain framework produced by recombination of IGHV3-30 and IGHJ6 ("an IGHV3-30 IGHJ6 framework").
  • the VH domain is produced by recombination of IGHV3-30, IGHD3-10 and IGHJ6.
  • the antibody may comprise a VH domain having at least 90 % amino acid sequence identity to the QUEL-0101 VH domain SEQ ID NO: 2, optionally at least 95 %, at least 98 % or at least 99 % sequence identity.
  • the VH domain may comprise or consist of SEQ ID NO: 2.
  • the VH domain may comprise or consist of a VH domain encoded by SEQ ID NO: 1 expressed in a mammalian cell, e.g., CHO.
  • the VL domain may be encoded by a nucleotide sequence produced by recombination of light chain v gene segment IGKV1 -27 (e.g., IGKV1 -27 * 01 ) with a light chain j gene segment, e.g., a kappa j segment such as IGKJ4 (e.g., IGKJ4 * 01).
  • IGKJ4 e.g., IGKJ4 * 01
  • it may comprise a VL domain framework produced by recombination of IGKV1-27 and IGKJ4 ("an IGKV1-27 IGKJ4 framework").
  • the antibody may comprise a VL domain having at least 90 % amino acid sequence identity to the QUEL-0101 VH domain SEQ ID NO: 7, optionally at least 95 %, at least 98 % or at least 99 % sequence identity.
  • the VL domain may comprise or consist of SEQ ID NO: 7.
  • the VL domain may comprise or consist of a VL domain encoded by SEQ ID NO: 6 expressed in a mammalian cell, e.g., CHO.
  • An anti-GFRAL antibody of the invention may comprise the QUEL-0101 VH domain SEQ ID NO: 2 and the QUEL-0101 VL domain SEQ ID NO: 7.
  • it comprises the QUEL-0102 VH domain SEQ ID NO: 98 and the QUEL-0102 VL domain SEQ ID NO: 103.
  • it comprises the QUEL-0103 VH domain SEQ ID NO: 106 and the QUEL-0103 VH domain SEQ ID NO: 110.
  • it comprises the QUEL-0104 VH domain SEQ ID NO: 112 and the QUEL-0104 VL domain SEQ ID NO: 115.
  • it comprises the QUEL-0105 VH domain SEQ ID NO: 117 and the QUEL-0105 VH domain SEQ ID NO: 121.
  • An antibody of the present invention may comprise a VH domain and a VL domain, the VH domain comprising the QUEL-0201 set of heavy chain complementarity determining regions (HCDRs) HCDR1 SEQ ID NO: 13, HCDR2 SEQ ID NO: 14 and HCDR3 SEQ ID NO: 15, and the VL domain comprising the QUEL-0201 set of light chain complementarity determining regions (LCDRs) LCDR1 SEQ ID NO: 18, LCDR2 SEQ ID NO: 19 and LCDR3 SEQ ID NO: 20.
  • HCDRs heavy chain complementarity determining regions
  • LCDRs light chain complementarity determining regions
  • An antibody of the present invention may comprise the QUEL-0201 VH domain SEQ ID NO: 12 or a variant thereof in which there are one or more amino acid alterations and/or the QUEL-0201 VL domain SEQ ID NO: 17 in which there are one or more amino acid alterations.
  • An amino acid alteration may be a substitution, deletion or insertion of an amino acid. Such alterations may optionally be in the variable domain framework, outside the CDRs. For example, there may be one or two amino acid alterations in the VH and/or VL domain framework. Alterations may be conservative substitutions and/or may represent germlining of framework residues.
  • the VH domain may be encoded by a nucleotide sequence produced by recombination of heavy chain v gene segment IGHV1 -3 (e.g., IGHV1 -3 * 01 ) with a d gene segment and a heavy chain j gene segment, e.g., IGHJ6 (e.g., IGHJ6 * 02).
  • IGHJ6 e.g., IGHJ6 * 02
  • it may comprise a VH domain framework produced by recombination of IGHV1-3 and IGHJ6 ("an IGHV1-3 IGHJ6 framework").
  • the VH domain is produced by recombination of IGHV1 -3, IGHD5-18 and IGHJ6.
  • the antibody may comprise a VH domain having at least 90 % amino acid sequence identity to the QUEL-0201 VH domain SEQ ID NO: 12, optionally at least 95 %, at least 98 % or at least 99 % sequence identity.
  • the VH domain may comprise or consist of SEQ ID NO: 12.
  • the VH domain may comprise or consist of a VH domain encoded by SEQ ID NO: 11 expressed in a mammalian cell, e.g., CHO.
  • the VL domain may be encoded by a nucleotide sequence produced by recombination of light chain v gene segment IGLV1 -40 (e.g., IGLV1 -40 * 01 ) with a light chain j gene segment, e.g., a lambda j segment such as IGLJ3 (e.g., IGLJ3 * 02).
  • IGLV1 -40 * 01 a light chain j gene segment
  • IGLJ3 e.g., IGLJ3 * 02
  • VL domain framework produced by recombination of IGLV1-40 and IGLJ3
  • An antibody of the present invention may comprise the QUEL-0201 VL domain SEQ ID NO: 17 or a variant thereof having one or more amino acid alterations.
  • the antibody may comprise a VL domain having at least 90 % amino acid sequence identity to the QUEL-0201 VH domain SEQ ID NO: 17, optionally at least 95 %, at least 98 % or at least 99 % sequence identity.
  • the VL domain may comprise or consist of SEQ ID NO: 17.
  • the VL domain may comprise or consist of a VL domain encoded by SEQ ID NO: 16 expressed in a mammalian cell, e.g., CHO.
  • An anti-GFRAL antibody of the invention may comprise the QUEL-0201 VH domain SEQ ID NO: 12 and the QUEL-0201 VL domain SEQ ID NO: 17.
  • An antibody of the present invention may comprise a VH domain and a VL domain, the VH domain comprising a set of HCDRs HCDR1 , HCDR2 and HCDR3, and the VL domain comprising a set of LCDRs LCDR1 , LCDR2 and LCDR3, wherein HCDR1 is SEQ ID NO: 23 or SEQ ID NO: 133,
  • HCDR2 is SEQ ID NO: 24, SEQ ID NO: 124, SEQ ID NO 134 or SEQ ID NO: 142
  • HCDR3 is SEQ ID NO: 25, SEQ ID NO: 125, SEQ ID NO: 135 or SEQ ID NO: 143, and optionally wherein
  • LCDR1 is SEQ ID NO: 28, SEQ ID NO: 128 or SEQ ID NO: 138,
  • LCDR2 is SEQ ID NO: 29 or SEQ ID NO: 129.
  • LCDR3 is SEQ ID NO: 30, SEQ ID NO: 130, SEQ ID NO: 139 or SEQ ID NO: 146.
  • An antibody of the present invention may comprise a VH domain and a VL domain, the VH domain comprising the QUEL-0301 set of heavy chain complementarity determining regions (HCDRs) HCDR1 SEQ ID NO: 23, HCDR2 SEQ ID NO: 24 and HCDR3 SEQ ID NO: 25, and the VL domain comprising the QUEL-0301 set of light chain complementarity determining regions (LCDRs) LCDR1 SEQ ID NO: 28, LCDR2 SEQ ID NO: 29 and LCDR3 SEQ ID NO: 30.
  • HCDRs heavy chain complementarity determining regions
  • LCDRs light chain complementarity determining regions
  • the antibody comprises a VH domain and a VL domain, the VH domain comprising the QUEL-0302 set of HCDRs HCDR1 SEQ ID NO: 23, HCDR2 SEQ ID NO: 124 and HCDR3 SEQ ID NO: 125, and the VL domain comprising the QUEL-0302 set of LCDRs LCDR1 SEQ ID NO: 128, LCDR2 SEQ ID NO: 129 and LCDR3 SEQ ID NO: 130.
  • the antibody comprises a VH domain and a VL domain, the VH domain comprising the QUEL-0303 set of HCDRs HCDR1 SEQ ID NO: 133, HCDR2 SEQ ID NO: 134 and HCDR3 SEQ ID NO: 135, and the VL domain comprising the QUEL-0303 set of LCDRs LCDR1 SEQ ID NO: 138, LCDR2 SEQ ID NO: 29 and LCDR3 SEQ ID NO: 139.
  • the antibody comprises a VH domain and a VL domain, the VH domain comprising the QUEL-0304 set of HCDRs HCDR1 SEQ ID NO: 133, HCDR2 SEQ ID NO: 142 and HCDR3 SEQ ID NO: 143, and the VL domain comprising the QUEL-0304 set of LCDRs LCDR1 SEQ ID NO: 138, LCDR2 SEQ ID NO: 29 and LCDR3 SEQ ID NO: 146.
  • An antibody of the present invention may comprise the QUEL-0301 VH domain SEQ ID NO: 22 or a variant thereof in which there are one or more amino acid alterations and/or the QUEL-0301 VL domain SEQ ID NO: 27 in which there are one or more amino acid alterations.
  • An amino acid alteration may be a substitution, deletion or insertion of an amino acid. Such alterations may optionally be in the variable domain framework, outside the CDRs. For example, there may be one or two amino acid alterations in the VH and/or VL domain framework. Alterations may be conservative substitutions and/or may represent germlining of framework residues.
  • Variants of the QUEL-0301 VH domain include VH domains that have one or more alterations that are present in the QUEL-0302, QUEL-0303 or QUEL-0304 VH domain, e.g., that are present in the framework of one or more of these VH domains.
  • An antibody of the present invention optionally comprises the QUEL-0302 VH domain SEQ ID NO: 123, the QUEL-0303 VH domain SEQ ID NO: 132 or the QUEL-0304 VH domain SEQ ID NO: 141.
  • the VH domain may be encoded by a nucleotide sequence produced by recombination of heavy chain v gene segment IGHV3-7 (e.g., IGHV3-7 * 01 ) with a d gene segment and a heavy chain j gene segment, e.g., IGHJ4 (e.g., IGHJ4 * 02).
  • IGHV3-7 heavy chain v gene segment
  • IGHJ4 heavy chain j gene segment
  • the VH domain is produced by recombination of IGHV3-7, IGHD1-7 and IGHJ4.
  • the VH domain is produced by recombination of IGHV3-7, IGHD1-20 and IGHJ4.
  • the antibody may comprise a VH domain having at least 90 % amino acid sequence identity to the QUEL-0301 VH domain SEQ ID NO: 22, optionally at least 95 %, at least 98 % or at least 99 % sequence identity.
  • the VH domain may comprise or consist of SEQ ID NO: 22.
  • the VH domain may comprise or consist of a VH domain encoded by SEQ ID NO: 21 expressed in a mammalian cell, e.g., CHO.
  • the VL domain may be encoded by a nucleotide sequence produced by recombination of light chain v gene segment IGLV1 -44 (e.g., IGLV1 -44 * 01 ) or IGLV1 -47 (e.g., IGLV1 -47 * 01 ) with a light chain j gene segment, e.g., a lambda j segment such as IGLJ3 (e.g., IGLJ3 * 02).
  • the light chain v gene segment is IGLV1-44.
  • the light chain v gene segment is IGLV1-47.
  • it may comprise a VL domain framework produced by recombination of IGLV1-44 and IGLJ3 ("an IGLV1-44 IGLJ3 framework").
  • it may comprise a VL domain framework produced by recombinatino of IGLV1-47 and IGLJ3 ("an IGLV1-47 IGLJ3 framework").
  • the antibody may comprise a VL domain having at least 90 % amino acid sequence identity to the QUEL-0301 VL domain SEQ ID NO: 27, optionally at least 95 %, at least 98 % or at least 99 % sequence identity.
  • the VL domain may comprise or consist of SEQ ID NO: 27.
  • the VL domain may comprise or consist of a VL domain encoded by SEQ ID NO: 26 expressed in a mammalian cell, e.g., CHO.
  • An anti-GFRAL antibody of the invention may comprise the QUEL-0301 VH domain SEQ ID NO: 22 and the QUEL-0301 VL domain SEQ ID NO: 27.
  • it comprises the QUEL-0302 VH domain SEQ ID NO: 123 and the QUEL-0302 VL domain SEQ ID NO: 127.
  • it comprises the QUEL-0303 VH domain SEQ ID NO: 132 and the QUEL-0303 VL domain SEQ ID NO: 137.
  • it comprises the QUEL-0304 VH domain SEQ ID NO: 141 and the QUEL-0304 VL domain SEQ ID NO: 145.
  • An anti-GFRAL antibody according to the present invention may be one that competes for binding to human GFRAL with QUEL-0101 , QUEL-0201 or QUEL-0301.
  • it may compete with QUEL-0201 IgG comprising QUEL-0201 VH domain SEQ ID NO: 12 and QUEL- 0201 VL domain comprising SEQ ID NO: 17.
  • it may compete with QUEL-0101 IgG comprising QUEL-0101 VH domain SEQ ID NO: 2 and QUEL-0101 VL domain SEQ ID NO: 7 and/or it may compete with QUEL-0301 IgG comprising QUEL-0301 VH domain SEQ ID NO: 22 and QUEL-0301 VL domain SEQ ID NO: 27.
  • the ability of a binding molecule to compete with a reference molecule for binding to GFRAL may be determined in vitro, e.g., by surface plasmon resonance (SPR) in a sandwich assay.
  • An anti-GFRAL antibody according to the present invention may be one which does not compete with GDF15 for binding to GFRAL.
  • Ability of a binding molecule to compete with GDF15 for binding to GFRAL may be also determined by SPR sandwich assay.
  • An anti-GFRAL antibody preferably binds human GFRAL with an affinity (KD) of 1 nanomolar (nM) or stronger (i.e., 1 nM or less than 1 nM) considering the limited expression of GRFAL protein. Affinity may be determined by SPR, e.g., as described in Example 2.
  • the KD may be 0.5 nM (500 picomolar, pM) or less, 400 pM or less, 300 pM or less, 200 pM or less, or 100 pM or less.
  • the KD may be 50 pM or less, e.g., 10 pM or less.
  • the KD may be approximately 100 pM, e.g., in the range 50 pM to 200 pM.
  • the KD may be 5 pM or less, 4 pM or less, 3 pM or less, 2 pM or less or 1 pM or less.
  • the KD may be approximately 1 pM (e.g., 0.5 pM - 2 pM).
  • KD may be at least 0.1 pM.
  • the KD may be at least 0.5 pM.
  • the KD may be at least 1 pM, e.g., in the range 1 pM to 1 nM.
  • An anti-GFRAL antibody preferably also binds non-human GFRAL (e.g., mouse GFRAL, rat GFRAL and/or cynomolgus GFRAL) in addition to human GFRAL.
  • non-human GFRAL e.g., mouse GFRAL, rat GFRAL and/or cynomolgus GFRAL
  • an antibody will be cross-reactive for mouse and cynomolgus GFRAL (within 10-fold affinity/potency).
  • the amino acid sequence identity between human GFRAL ( Figure 1) and mouse GFRAL ( Figure 2) is 70%, and between human and cynomolgus monkey is 94%.
  • the degree of cross-reactivity of an antibody may be quantified as a fold difference in KD for binding human and non-human GFRAL.
  • an antibody will have an affinity for mouse GFRAL within 100-fold, within 10-fold, within 5-fold or more preferably within 2-fold of its affinity for human GFRAL.
  • An anti-GFRAL antibody may bind mouse GFRAL with an affinity (KD) of 10 nM or stronger, preferably 5 nM or stronger. Affinity for mouse GFRAL is optionally 100 pM or less.
  • Anti-GFRAL antibodies described herein are inhibitors of GDF15 signalling. Specifically, they inhibit GDF15 signalling through GFRAL, by binding to GFRAL extracellular domain and inhibiting formation of the cell surface GDF15-GFRAL-RET signalling complex. Potency of inhibition by anti-GFRAL antagonist antibodies may be determined in an in vitro assay, such as an ERK phosphorylation assay. This is a cell-based assay which determines the ability of a candidate antagonist molecule to inhibit the RET signalling that is triggered by addition of GDF15. Potency can be quantified as IC50 in the assay.
  • an antibody inhibits GFRAL activation with a potency (IC50) of 15 nM or stronger (i.e., 15 nM or less than 15 nM) in the ERK phosphorylation assay.
  • IC50 is 10 nM or less, e.g., 5 nM or less.
  • Nucleic acid encoding antibodies as described herein is also provided, as are cells comprising said nucleic acid.
  • a host cell in vitro may comprise the nucleic acid, optionally integrated into its cellular (e.g., genomic) DNA, or transiently transfected (e.g., plasmid DNA).
  • Anti-GFRAL antagonist antibodies are suitable for medical use. Antagonistic antibodies that target GFRAL, inhibiting signalling of the hormone GDF15 via the GDF15-GFRAL-RET pathway, represent therapeutic agents for conditions such as cachexia (e.g., cancer cachexia), anorexia and hyperemesis gravidarum.
  • Anti-GFRAL antibodies may be administered to patients to increase food intake (e.g., for patients with cachexia or anorexia relating to cancer).
  • Anti-GFRAL antibodies or their encoding nucleic acids may be used as an adjuvant therapeutic drug, in combination with other anti-cancer interventions such as surgery, radiotherapy and/or administration of anti-neoplastic drugs.
  • Figure 1 shows the amino acid sequence of human GFRAL SEQ ID NO: 31 , NCBI NP 997293.2, comprising 394 amino acids of which residues 1 - 18 represent a signal peptide. Boxed asparagine residues are sites of N-linked glycosylation (GlcNAc) at N23, N50, N62, N67, N103 and N 116.
  • GlcNAc N-linked glycosylation
  • GFRAL contains two GDNF/GAS1 domains, formed by residues 131 - 210 and 220 - 316 respectively, boxed.
  • a region believed to be required for interaction with GDF15 is residues 149 - 228, italicised.
  • a transmembrane domain spans residues 352 - 371 , emboldened.
  • residue R is present at ⁇ 62 % and C is present at ⁇ 38 %.
  • Residue P386 in the cytoplasmic tail is also variable and may be S.
  • Figure 2 shows the amino acid sequence of mouse GFRAL SEQ ID NO: 32, NCBI NP 995316.2. Sequence feature annotations use the same convention as for Figure 1 .
  • Figure 3 shows AF % plotted against log human GDF15 molar concentration in an ERK phosphorylation assay.
  • Figure 4 shows change in fluorescence for antagonist titrations in the ERK phosphorylation assay with 4 nM (0.05 pg/ml) GDF15.
  • Figure 5 shows sensorgrams from SPR analysis of antibodies binding to human GFRAL.
  • A QUEL-0101.
  • B QUEL-0201.
  • C QUEL-0301.
  • Figure 6 shows sensorgrams from SPR analysis of antibodies binding to mouse GFRAL.
  • A QUEL-0101.
  • B QUEL-0201.
  • C QUEL-0301.
  • aspects of the present invention include: methods of treating patients with an inhibitor of GDF15 signalling, an inhibitor of GDF15 signalling for use in treating patients, and use of an inhibitor of GDF15 signalling for the manufacture of a medicament for treating patients.
  • an inhibitor of GDF15 signalling as described herein may be used to treat any medical condition associated with the GDF15-GFRAL pathway. Examples of such conditions are described herein, and include
  • Treatment may comprise preventative treatment, wherein the therapeutic composition is administered in advance of emergence of the condition to be treated, in order to prevent or at least ameliorate the effects of the condition. Treatment may also be given after emergence of the condition, e.g., the treatment may be prescribed following diagnosis of the condition.
  • Conditions (i) - (vi) may be side effects of other medical interventions such as treatment with cytotoxic agents (e.g., anti-neoplastic agents used in treating cancer) or radiotherapy.
  • the patient may have an elevated physiological level of GDF15, detectable in the blood.
  • the patient to be treated may be a cancer patient.
  • a cancer patient As is evident from the published literature in the field, many types of cancer are associated with an increase in GDF15, which will drive cancer cachexia.
  • the cancer may be a solid tumour (optionally metastatic) or a blood cancer.
  • the cancer may be bladder cancer, brain cancer, breast cancer, colorectal cancer, head and neck cancer, kidney cancer, lung cancer (e.g., non-small cell lung cancer), lymphoma (e.g., non-Hodgkin lymphoma), melanoma, mesothelioma, neuroblastoma, oesophageal cancer, oral or oropharyngeal cancer, gastrointestinal cancer (e.g., gastric cancer), pancreatic cancer, prostate cancer, testicular cancer, thyroid cancer or uterine cancer.
  • lung cancer e.g., non-small cell lung cancer
  • lymphoma e.g., non-Hodgkin lymphoma
  • melanoma mesothelioma
  • neuroblastoma e.g., oesophageal cancer
  • oral or oropharyngeal cancer e.g., gastrointestinal cancer
  • pancreatic cancer e.g., prostate cancer, testicular cancer, thyroid cancer or
  • treatment may comprise treating cachexia in a patient with a pulmonary and/or cardiac condition (e.g., congestive heart failure, chronic obstructive pulmonary disease), chronic kidney disease, acquired immune deficiency syndrome (AIDS), cystic fibrosis, multiple sclerosis, motor neuron disease, Parkinson's disease, dementia, tuberculosis, multiple system atrophy, mercury poisoning, Crohn's disease, rheumatoid arthritis or celiac disease.
  • a pulmonary and/or cardiac condition e.g., congestive heart failure, chronic obstructive pulmonary disease), chronic kidney disease, acquired immune deficiency syndrome (AIDS), cystic fibrosis, multiple sclerosis, motor neuron disease, Parkinson's disease, dementia, tuberculosis, multiple system atrophy, mercury poisoning, Crohn's disease, rheumatoid arthritis or celiac disease.
  • Treatment may result in reduction of the condition, and increased food intake and/or stabilisation or gain of weight by the patient may be observed.
  • a patient may be treated for cancer-associated cachexia, wherein treatment increases body weight or reduces loss of body weight.
  • a patient treated in accordance with the present invention may be one who has been exposed or will be exposed to a cytotoxic chemical agent.
  • Cytotoxic agents include those that are genotoxic (e.g., by damaging or cross-linking DNA, and/or by inhibiting DNA synthesis or DNA replication), or which induce ER stress (e.g., by inhibiting protein folding).
  • a patient treated in accordance with the present invention may be one who has been exposed or will be exposed to ionising radiation.
  • Cytotoxic agents include those that induce cell death (e.g., by apoptosis).
  • the cytotoxic agent may be an anti-neoplastic agent, examples of which are presented below and elsewhere herein.
  • Treatment may comprise combination therapy, in which a composition of the present invention is administered to a patient who is also receiving treatment with an anti-neoplastic agent, such as cancer chemotherapy.
  • the composition of the present invention may be administered before or after the anti-neoplastic agent, or simultaneously. It will generally be administered in a separate formulation, although a single medicament comprising both agents is a possibility where they can be formulated together.
  • Treatment according to the present invention may enhance the effect of the chemotherapy.
  • the treatment may extend survival of the patient. It is also expected to significantly improve patients' quality of life.
  • the patient to be treated may be one who has received, or who will receive, treatment with one or more of the following cancer chemotherapeutic agents:
  • Alkylating agents e.g., bifunctional alkylators such as cyclophosphamide, mechlorethamine, chlorambucil, melphalan, or monofunctional alkylators such as dacarbazine, nitrosoureas, temozolomide;
  • Anthracyclines e.g., daunorubicin, doxorubicin, epirubicin, idarubicin, mitoxantrone, valrubicin; Cytoskeletal disruptors (e.g., taxanes such as paclitaxel, docetaxel, abraxane, taxotere)
  • Epothilones e.g., epothilone A, B, C, D, E or F
  • Histone deacetylase inhibitors e.g., vorinostat, romidepsin
  • Inhibitors of topoisomerase I e.g., irinotecan, topotecan, tafluposide
  • Inhibitors of topoisomerase II e.g., etoposide, teniposide, tafluposide
  • kinase inhibitors e.g., bortezomib, erlotinib, gefitinib, imatinib, vemurafenib, vismodegib
  • Nucleotide analogs and precursor analogs e.g., azacitidine, azathioprine, capecitabine, cytarabine, doxifluridine, fluorouracil, gemcitabine, hydroxyurea, mercaptopurine, methotrexate, tioguanine
  • Peptide antibiotics e.g., bleomycin, actinomycin
  • Platinum-based agents e.g., carboplatin, cisplatin, oxaliplatin
  • Retinoids e.g., alitretinoin, bexarotene, tretinoin
  • Vinca alkaloids and derivatives e.g., vinblastine, vincristine, vindesine, vinorelbine.
  • Antibodies e.g., vinblastine, vincristine, vindesine, vinorelbine.
  • Antibodies according to the present invention are immunoglobulins or molecules comprising immunoglobulin domains, whether natural or partly or wholly synthetically produced.
  • Antibodies may be IgG, IgM, IgA, IgD or IgE molecules or antigen-specific antibody fragments thereof (including, but not limited to, a Fab, F(ab')2, Fv, disulphide linked Fv, scFv, single domain antibody, closed conformation multispecific antibody, disulphide-linked scfv, diabody), whether derived from any species that naturally produces an antibody, or created by recombinant DNA technology; whether isolated from serum, B-cells, hybridomas, transfectomas, yeast or bacteria.
  • Antibodies can be humanised using routine technology.
  • antibody covers any polypeptide or protein comprising an antibody antigen-binding site.
  • An antigen-binding site is the part of an antibody that binds to and is complementary to the epitope of its target antigen, e.g., GFRAL.
  • epitope refers to a region of an antigen that is bound by an antibody.
  • Epitopes may be defined as structural or functional. Functional epitopes are generally a subset of the structural epitopes and have those residues that directly contribute to the affinity of the interaction. Epitopes may also be conformational, that is, composed of non-linear amino acids.
  • epitopes may include determinants that are chemically active surface groupings of molecules such as amino acids, sugar side chains, phosphoryl groups, or sulfonyl groups, and, in certain embodiments, may have specific three-dimensional structural characteristics, and/or specific charge characteristics.
  • the antigen binding site is a polypeptide or domain that comprises one or more CDRs of an antibody and is capable of binding the antigen.
  • the polypeptide comprises a CDR3 (e.g., HCDR3).
  • the polypeptide comprises CDRs 1 and 2 (e.g., HCDR1 and 2) or CDRs 1 -3 of a variable domain of an antibody (e.g., HCDRsl -3).
  • An antibody antigen-binding site may be provided by one or more antibody variable domains.
  • the antibody binding site is provided by a single variable domain, e.g., a heavy chain variable domain (VH domain) or a light chain variable domain (VL domain).
  • the binding site comprises a VH/VL pair or two or more of such pairs.
  • an antibody antigen-binding site may comprise a VH and a VL.
  • the antibody may be a whole immunoglobulin, including constant regions, or may be an antibody fragment.
  • An antibody fragment is a portion of an intact antibody, for example comprising the antigen binding and/or variable region of the intact antibody. Examples of antibody fragments include:
  • Fab fragment a monovalent fragment consisting of the VL, VH, CL and CH1 domains
  • F(ab')2 fragment a bivalent fragment including two Fab fragments linked by a disulfide bridge at the hinge region
  • CDR complementarity determining region
  • antibodies are H2 antibodies that comprise a dimer of a heavy chain (5’-VH-(optional hinge)-CH2-CH3-3’) and are devoid of a light chain.
  • Single-chain antibodies e.g., scFv
  • Multispecific antibodies may be formed from antibody fragments.
  • An antibody of the invention may employ any such format, as appropriate.
  • binder polypeptides, or antibody immunoglobulin domains thereof may be fused or conjugated to additional polypeptide sequences and/or to labels, tags, toxins or other molecules. Binder polypeptides may be fused or conjugated to one or more different antigen binding regions, providing a molecule that is able to bind a second antigen in addition to GFRAL.
  • an antibody of the present invention may be a multispecific antibody, e.g., a bispecific antibody, comprising (i) an antibody antigen binding site for GFRAL and (ii) a further antigen binding site (optionally an antibody antigen binding site, as described herein) which recognises another antigen.
  • An antibody normally comprises an antibody VH and/or VL domain.
  • Isolated VH and VL domains of antibodies are also part of the invention.
  • the antibody variable domains are the portions of the light and heavy chains of antibodies that include amino acid sequences of complementarity determining regions (CDRs; ie., CDR1 , CDR2, and CDR3), and framework regions (FRs).
  • CDRs complementarity determining regions
  • FRs framework regions
  • a VFI domain comprises a set of FICDRs
  • a VL domain comprises a set of LCDRs.
  • VFI refers to the variable domain of the heavy chain.
  • VL refers to the variable domain of the light chain.
  • Each VFI and VL is typically composed of three CDRs and four FRs, arranged from amino-terminus to carboxy-terminus in the following order: FR1 , CDR1 , FR2, CDR2, FR3, CDR3, FR4.
  • FR1 , CDR1 , FR2, CDR2, FR3, CDR3, FR4 amino acid positions assigned to CDRs and FRs are defined according to IMGT nomenclature.
  • An antibody may comprise an antibody VFI domain comprising a VFI CDR1 , CDR2 and CDR3 and a framework. It may alternatively or also comprise an antibody VL domain comprising a VL CDR1 , CDR2 and CDR3 and a framework. Examples of antibody VFI and VL domains and CDRs according to the present invention are as listed in Table A. All VFI and VL sequences, CDR sequences, sets of CDRs and sets of FICDRs and sets of LCDRs disclosed herein represent aspects and embodiments of the invention. As described herein, a "set of CDRs" comprises CDR1 , CDR2 and CDR3.
  • a set of FICDRs refers to FICDR1 , FICDR2 and FICDR3, and a set of LCDRs refers to LCDR1 , LCDR2 and LCDR3. Unless otherwise stated, a "set of CDRs" includes FICDRs and LCDRs.
  • QUEL-0101 As described in more detail in the Examples, we isolated and characterised antibodies of particular interest, designated QUEL-0101 , QUEL-0201 and QUEL-0301. Subsequently, we identified structural variants of QUEL-0101 and QUEL-0301 respectively, namely QUEL-0102, QUEL-0103, QUEL-0104, QUEL-0105, QUEL-0302, QUEL-0303 and QUEL-0304.
  • an anti-GFRAL antibody may optionally be selected from QUEL-0101 , QUEL-0102, QUEL-0103, QUEL-0104, QUEL-0105, QUEL-0201 , QUEL-0301 , QUEL-0302, QUEL-0303 and QUEL-0304.
  • it is selected from QUEL-0101 , QUEL-0201 and QUEL-0301.
  • the present invention encompasses anti-GFRAL antibodies having the VFI and/or VL domain sequences of these antibodies, as shown in the appended sequence listing, as well as antibodies comprising the FICDRs and/or LCDRs of those antibodies, and optionally having the full heavy chain and/or full light chain amino acid sequence.
  • an antibody VFI domain or VL domain comprises one or more residues in a framework region which differ from the germline gene segment from which it was obtained by recombination
  • the non-germline residue may be retained or may be mutated to a different residue, e.g., it may be reverted to the germline residue.
  • Corresponding germline gene segments may be identified as the gene segment to which the sequence of the variable domain is most closely aligned, and the germline gene segments corresponding to each of QUEL-0101 , QUEL-0201 and QUEL-0301 VH and VL domains, and to their corresponding related antibodies, are shown in Table G herein.
  • An antibody according to the present invention may comprise one or more CDRs as described herein, e.g. a CDR3, and optionally also a CDR1 and CDR2 to form a set of CDRs.
  • the CDR or set of CDRs may be a CDR or set of CDRs of any of QUEL-0101 , QUEL-0102, QUEL-0103, QUEL-0104, QUEL-0105, QUEL-0201 , QUEL-0301 , QUEL-0302, QUEL-0303 and QUEL-0304.
  • the invention provides antibodies comprising an HCDR1 , HCDR2 and/or HCDR3 of any of antibodies QUEL-0101 , QUEL-0102, QUEL-0103, QUEL-0104, QUEL-0105, QUEL-0201 , QUEL-0301 , QUEL-0302, QUEL-0303 and QUEL-0304 and/or an LCDR1 , LCDR2 and/or LCDR3 of any of these antibodies, e.g. a set of CDRs.
  • the antibody may comprise a set of VH CDRs of one of these antibodies.
  • it may also comprise a set of VL CDRs of one of these antibodies, and the VL CDRs may be from the same or a different antibody as the VH CDRs.
  • a VH domain comprising a disclosed set of HCDRs, and/or a VL domain comprising a disclosed set of LCDRs, are also provided by the invention.
  • a VH domain is paired with a VL domain to provide an antibody antigen binding site, although as discussed further below a VH or VL domain alone may be used to bind antigen.
  • the QUEL-0201 VH domain may be paired with the QUEL-0201 VL domain, so that an antibody antigen-binding site is formed comprising both the QUEL-0201 VH and VL domains. Analogous embodiments are provided for the other VH and VL domains disclosed herein.
  • the QUEL-0201 VH is paired with a different VL domain, e.g., a l VL domain, e.g., the QUEL-0301 VL domain.
  • the QUEL-0301 VH may be paired with a different VL domain, e.g., a l VL domain, e.g., the QUEL-0201 VL domain.
  • Light-chain promiscuity is well established in the art. For example:
  • the QUEL-0101 VH domain can be paired with the VL domain of any of QUEL-0101 , QUEL-0102, QUEL-0103, QUEL-0104 and QUEL-0105.
  • the QUEL-0102 VH domain can be paired with the VL domain of any of QUEL-0101 , QUEL-0102, QUEL-0103, QUEL-0104 and QUEL-0105.
  • the QUEL-0103 VH domain can be paired with the VL domain of any of QUEL-0101 , QUEL-0102, QUEL-0103, QUEL-0104 and QUEL-0105.
  • the QUEL-0104 VH domain can be paired with the VL domain of any of QUEL-0101 , QUEL-0102, QUEL-0103, QUEL-0104 and QUEL-0105.
  • the QUEL-0105 VH domain can be paired with the VL domain of any of QUEL-0101 , QUEL-0102, QUEL-0103, QUEL-0104 and QUEL-0105.
  • the QUEL-0301 VH domain can be paired with the VL domain of any of QUEL-0301 , QUEL-0302, QUEL-0303 and QUEL-0304.
  • the QUEL-0302 VH domain can be paired with the VL domain of any of QUEL-0301 , QUEL-0302, QUEL-0303 and QUEL-0304.
  • the QUEL-0303 VH domain can be paired with the VL domain of any of QUEL-0301 , QUEL-0302, QUEL-0303 and QUEL-0304.
  • the QUEL-0304 VH domain can be paired with the VL domain of any of QUEL-0301 , QUEL-0302, QUEL-0303 and QUEL-0304.
  • An antibody may comprise one or more CDRs, e.g. a set of CDRs, within an antibody framework.
  • the framework regions may be of human germline gene segment sequences.
  • the antibody may be a human antibody having a VH domain comprising a set of HCDRs in a human germline framework.
  • the antibody also has a VL domain comprising a set of LCDRs, e.g. in a human germline framework.
  • an antibody “gene segment”, e.g., a VH gene segment, D gene segment, or JH gene segment refers to oligonucleotide having a nucleic acid sequence from which that portion of an antibody is derived, e.g., a VH gene segment is an oligonucleotide comprising a nucleic acid sequence that corresponds to a polypeptide VH domain from FR1 to part of CDR3.
  • Human V, D and J gene segments recombine to generate the VH domain, and human V and J segments recombine to generate the VL domain.
  • the D domain or region refers to the diversity domain or region of an antibody chain.
  • J domain or region refers to the joining domain or region of an antibody chain.
  • Somatic hypermutation may result in an antibody VH or VL domain having framework regions that do not exactly match or align with the corresponding gene segments, but sequence alignment can be used to identify the closest gene segments and thus identify from which particular combination of gene segments a particular VH or VL domain is derived.
  • the antibody amino acid sequence may be aligned with the amino acid sequence encoded by the gene segment, or the antibody nucleotide sequence may be aligned directly with the nucleotide sequence of the gene segment.
  • An antibody of the invention may be a human antibody or a chimaeric antibody comprising human variable regions and non-human (e.g., mouse) constant regions.
  • the antibody of the invention for example has human variable regions, and optionally also has human constant regions.
  • antibodies optionally include constant regions or parts thereof, e.g., human antibody constant regions or parts thereof.
  • a VL domain may be attached at its C- terminal end to antibody light chain kappa or lambda constant domains.
  • an antibody VH domain may be attached at its C-terminal end to all or part (e.g. a CH1 domain or Fc region) of an immunoglobulin heavy chain constant region derived from any antibody isotype, e.g. IgG, IgA, IgE and IgM and any of the isotype sub-classes, such as lgG1 or lgG4.
  • a preferred example is lgG4PE, e.g., SEQ ID NO: 60.
  • Table C Further examples of human heavy chain constant region sequences are shown in Table C.
  • the anti-GFRAL antibody is QUEL-0201 IgG comprising
  • a heavy chain comprising the QUEL-0201 VFI domain SEQ ID NO: 12 and human lgG4 heavy chain constant region (e.g., lgG4PE SEQ ID NO: 60), and
  • the anti-GFRAL antibody is QUEL-0301 IgG comprising
  • a heavy chain comprising the QUEL-0301 VFI domain SEQ ID NO: 22 and human lgG4 heavy chain constant region (e.g., lgG4PE SEQ ID NO: 60), and
  • the anti-GFRAL antibody is QUEL-0101 IgG comprising
  • a heavy chain comprising the QUEL-0101 VFI domain SEQ ID NO: 2 and human lgG4 heavy chain constant region (e.g., lgG4PE SEQ ID NO: 60), and
  • Constant regions of antibodies of the invention may alternatively be non-human constant regions.
  • chimaeric antibodies may be produced comprising human variable regions and non-human (host animal) constant regions.
  • Some transgenic animals generate fully human antibodies.
  • Others have been engineered to generate antibodies comprising chimaeric heavy chains and fully human light chains.
  • these may be replaced with human constant regions to provide antibodies more suitable for administration to humans as therapeutic compositions, as their immunogenicity is thereby reduced.
  • Fab antigen-binding fragments
  • Fc fragments having no antigen-binding activity but having the ability to crystallize.
  • Fab when used herein refers to a fragment of an antibody that includes one constant and one variable domain of each of the heavy and light chains.
  • Fc region herein is used to define a C-terminal region of an immunoglobulin heavy chain, including native-sequence Fc regions and variant Fc regions.
  • the "Fc fragment” refers to the carboxy-terminal portions of both FI chains held together by disulfides.
  • the effector functions of antibodies are determined by sequences in the Fc region, the region which is also recognised by Fc receptors (FcR) found on certain types of cells. Digestion of antibodies with the enzyme pepsin, results in a F(ab')2 fragment in which the two arms of the antibody molecule remain linked and comprise two-antigen binding sites. The F(ab')2 fragment has the ability to crosslink antigen.
  • Fv when used herein refers to the minimum fragment of an antibody that retains both antigen-recognition and antigen-binding sites. This region consists of a dimer of one heavy and one light chain variable domain in tight, non-covalent or covalent association. It is in this configuration that the three CDRs of each variable domain interact to define an antigen-binding site on the surface of the VFI-VL dimer. Collectively, the six CDRs confer antigen-binding specificity to the antibody. Flowever, even a single variable domain (or half of an Fv comprising only three CDRs specific for an antigen) has the ability to recognise and bind antigen, although at a lower affinity than the entire binding site.
  • Antibodies disclosed herein may be modified to increase or decrease serum half-life.
  • one or more of the following mutations: T252L, T254S or T256F are introduced to increase biological half-life of the antibody.
  • Biological half-life can also be increased by altering the heavy chain constant region CHi domain or CL region to contain a salvage receptor binding epitope taken from two loops of a CFI2 domain of an Fc region of an IgG, as described in U.S. Patent Numbers. 5,869,046 and 6,121 ,022, the modifications described therein are incorporated herein by reference.
  • the Fc hinge region of an antibody or antigen-binding fragment of the invention is mutated to decrease the biological half-life of the antibody or fragment.
  • the antibody or fragment is PEGylated.
  • the antibody or fragment is fused to an albumin-biding domain, e.g. an albumin binding single domain antibody (dAb).
  • the antibody or fragment is PASylated (i.e. genetic fusion of polypeptide sequences composed of PAS (XL-Protein GmbH) which forms uncharged random coil structures with large hydrodynamic volume).
  • the antibody or fragment is XTENylated ® /rPEGylated (i.e. genetic fusion of non exact repeat peptide sequence (Amunix, Versartis) to the therapeutic peptide).
  • the antibody or fragment is ELPylated (i.e. genetic fusion to ELP repeat sequence (PhaseBio)).
  • antibodies can be provided in various isotypes and with different constant regions.
  • the Fc region of antibodies is recognised by Fc receptors and determines the ability of the antibody to mediate cellular effector functions, including antibody-dependent cell- mediated cytotoxicity (ADCC) activity, complement dependent cytotoxicity (CDC) activity and antibody-dependent cell phagocytosis (ADCP) activity.
  • ADCC antibody-dependent cell- mediated cytotoxicity
  • CDC complement dependent cytotoxicity
  • ADCP antibody-dependent cell phagocytosis
  • antibodies according to the present invention may lack Fc effector function, for example they may contain Fc regions that do not mediate ADCC, ADCP and/or CDC, or they may lack Fc regions or lack antibody constant regions entirely.
  • An antibody may have a constant region which is effector null.
  • An antibody may have a heavy chain constant region that binds one or more types of Fc receptor but does not induce cellular effector functions, i.e., does not mediate ADCC, CDC or ADCP activity. Such a constant region may be unable to bind the particular Fc receptor(s) responsible for triggering ADCC, CDC or ADCP activity.
  • an antibody may have a heavy chain constant region that does not bind Fey receptors, for example the constant region may comprise a Leu235Glu mutation (i.e., where the wild type leucine residue is mutated to a glutamic acid residue), which may be referred to as an ⁇ ” mutation, e.g., lgG4-E.
  • Another optional mutation for a heavy chain constant region is Ser228Pro (“P” mutation), which increases stability by reducing Fab arm exchange.
  • a heavy chain constant region may be an lgG4 comprising both the Leu235Glu mutation and the Ser228Pro mutation. This “lgG4-PE” heavy chain constant region is effector null.
  • An alternative effector null human constant region is a disabled lgG1 .
  • lgG4PE is a preferred antibody isotype for the present invention.
  • a binder polypeptide may be an lgG4PE antibody comprising the sequence of an lgG4PE constant region shown in Table C.
  • Antibody constant regions may be engineered to have an extended half life in vivo. Examples include ⁇ TE” mutations and other half-life extending mutations (Dall’Acqua, Kiener & Wu, JBC 281 (33):23514-235242006 and W002/060919, incorporated by reference herein).
  • the triple mutation YTE is a substitution of 3 amino acids in the IgG CH2 domain, these mutations providing tyrosine at residue 252, threonine at residue 254 and glutamic acid at residue 256, numbered according to the EU index of Kabat. As described in the referenced publications, the YTE modification increases the half-life of the antibody compared with the half- life of a corresponding antibody having a human CH2 wild type domain.
  • antibodies of the present invention may include antibody constant regions (e.g., IgG constant regions, e.g., IgG CH2 domains) that have one or more mutations that increase the half life of the antibody compared with the corresponding wild type human constant region (e.g., IgG, e.g., IgG CH2 domain).
  • Half-life may be determined by standard methods, such as are described in W002/060919.
  • An inhibitor of GDF15 signalling e.g., an anti-GFRAL antibody
  • an anti-GFRAL antibody may recognise an epitope within the extracellular domain of GFRAL. It may bind within the sequence of residues 19 - 351 as shown in Figure 1.
  • the epitope may comprise an N terminal region of GFRAL comprising residues upstream of the first GDNF/GAS1 domain.
  • the residues of GFRAL that bind to the antibody may be determined by structural resolution of the antibody:antigen complex, e.g., by cryo electron microscopy or by x-ray crystallography. Binding residues may include those that form salt bridges, hydrophobic interactions or hydrogen bonds with the antibody, via their side chain or the polypeptide backbone.
  • the epitope may further include a carbohydrate moiety on the glycosylated antigen.
  • An anti-GFRAL antibody may recognise an epitope of GFRAL which is the same as or overlaps with the epitope recognised by QUEL-0101 , QUEL-0201 or QUEL-0301 .
  • An anti- GFRAL antibody may for example bind an epitope comprising one or more, optionally all, of the residues bound by QUEL-0101 , QUEL-0201 or QUEL-0301. Recognition of these epitopes is associated with antagonist activity, i.e., inhibition of GDF15-GFRAL-RET signalling, and are thus valuable epitopes to target.
  • an anti-GFRAL binding agent may compete with an antibody (e.g., IgG or scFv) comprising the VH and VL domains, or an IgG comprising the full heavy and light chains, of QUEL-0101 , QUEL-0201 or QUEL-0301. It may for example compete with QUEL-0201 IgG.
  • an antibody e.g., IgG or scFv
  • Binder polypeptides or other agents indicates that they have epitopes in the same region of GFRAL, e.g., both may bind the same domain with an overlapping binding footprint. This may be confirmed by other techniques, e.g., by structural resolution of the anti-GFRAL molecule bound to GFRAL, which may be achieved for example using cryo electron microscopy or x-ray crystallography as mentioned.
  • Competition is optionally determined by a sandwich assay to assess the ability of the two binders to simultaneously bind GFRAL extracellular domain in solution.
  • a first binder e.g., anti-GFRAL antibody QUEL-0101 , QUEL-0201 or QUEL-0301 IgG
  • the GFRAL antigen is added in solution, allowing formation of an antibody- antigen (or other binder-antigen) complex.
  • the test antibody or other anti-GFRAL binding molecule is then added in solution. If binding of the test molecule is detected, this indicates that it does not compete with the first binder (e.g., reference (QUEL) antibody).
  • first binder e.g., reference (QUEL) antibody
  • the assay may be performed using SPR, wherein the first binder bound to the surface of a biosensor chip.
  • SPR For an IgG, coupling is commonly via the Fc region, e.g., using a chip coated with anti-Fc antibody. See Example 4 for further details of the SPR sandwich assay.
  • an anti-GFRAL binding agent does not inhibit binding of GDF15 to GFRAL. This may be determined in a sandwich assay using the principles described above, coupling the binder to a solid support and adding GFRAL antigen in solution, then determining binding or absence of binding of GDF15.
  • the affinity of a binder (e.g., anti-GFRAL antibody) for GFRAL may be quantified in terms of the equilibrium dissociation constant KD, which is the ratio Ka/Kd of the association or on-rate (Ka) and the dissociation or off-rate (kd) of the binding interaction.
  • KD, Ka and Kd for antigen binding can be measured using surface plasmon resonance SPR.
  • Example SPR procedure and conditions are set out in Example 2 and Example 3.
  • Affinity (KD) is a measure of how strong the interaction of the antibody with its antigen is.
  • Association rate (ka) shows how fast antigen is recognised.
  • Dissociation rate (ka) is a measure of stability of binding.
  • SPR may comprise coating or immobilising the anti-GFRAL binder on to a biosensor chip (directly or indirectly), exposing the binder to the antigen in buffered solution at a range of concentrations, detecting binding, and calculating the equilibrium dissociation constant KD for the binding interaction.
  • a biosensor chip directly or indirectly
  • exposing the binder to the antigen in buffered solution at a range of concentrations detecting binding, and calculating the equilibrium dissociation constant KD for the binding interaction.
  • coupling to the chip can conveniently be done via Fc capture on an anti-Fc-coated chip (a chip with anti-human Fc antibody on its surface, e.g., chemically immobilised at the chip surface).
  • the binding data can be fitted to a 1 :1 model using standard algorithms, which may be inherent to the instrument used.
  • SPR may be performed at 25 e C by capturing the binder on a chip for 60 seconds at 1 pg/ml concentration (e.g., approximately between 80 and 140 RU may be captured), and soluble GFRAL (analyte) injected for 120 sec (association time) at 30 pL/min and dissociation monitored for 1200 seconds.
  • Analyte may be injected at a dilution series, (e.g., 100, 25, 6.25, 1.56, 0.39, 0.098, 0.024 and 0 nM concentration).
  • a suitable running buffer is HBS-P+ buffer pH 7.4 with 1 mM CaCh.
  • Sensorgrams for the binder polypeptide are generated, and data may be fitted to a 1 :1 interaction model. KD and optionally other kinetic data are calculated.
  • Isolated purified GFRAL extracellular domain may conveniently be used in assays and is a suitable analyte for SPR (see Example 2).
  • the antibody must be cross-reactive with the corresponding antigen in the species of interest.
  • Antibodies of the present invention preferably bind mouse GFRAL, rat GFRAL and/or cynomolgus GFRAL in addition to human GFRAL.
  • the extent of species cross-reactivity of an anti-GFRAL antibody or other anti-GFRAL binding molecule is as the fold-difference in its affinity for antigen or one species compared with antigen of another species, e.g., fold difference in affinity for human antigen vs mouse antigen.
  • Affinity may be quantified as KD, referring to the equilibrium dissociation constant of the binding of the antigen to the antigen-binding molecule. KD may be determined by SPR as described elsewhere herein.
  • a species cross-reactive binding molecule may have a fold-difference in affinity for binding human and non-human antigen that is 100-fold or less, 50-fold or less, 30-fold or less, 25-fold or less, 20-fold or less, 15-fold or less, 10-fold or less, 5-fold or less, or 2-fold or less.
  • the KD of binding the extracellular domain of the human antigen may be within 100-fold, 50-fold, 30-fold, 25-fold, 20-fold, 15-fold, 10-fold, 5-fold or 2-fold of the KD of binding the extracellular domain of the non-human antigen.
  • the binding affinities of human and non-human antigen are within a range of 10-fold or less, more preferably within 5-fold or within 2-fold.
  • KD for binding mouse GFRAL e.g., as determined by SPR, may be up to 10-fold (preferably up to 5-fold or up to 2-fold) greater or up to 10-fold lower (preferably up to 5-fold or up to 2-fold lower) than the KD for binding human GFRAL.
  • Binding molecules can also be considered species cross-reactive if the KD for binding antigen of both species meets a threshold value, e.g., if the KD of binding human antigen and the KD of binding non-human (e.g., mouse) antigen are both 10 mM or less, preferably 5 mM or less, more preferably 1 mM or less.
  • the KD may be 100 nM or less, 50 nM or less, 25 nM or less, 10 nM or less, 5 nM or less, 2 nM or less, or 1 nM or less.
  • a binding molecule may have a measurable capacity to inhibit GDF15 signalling in a cell based assay with GFRAL from multiple species, (e.g., one or more, or all, of human and mouse, rat and cynomolgus GFRAL). It may exhibit dose-dependent inhibition of GDF15-induced GFRAL signalling activity in an assay described herein with human and non-human (e.g., mouse, rat or cynomolgus) GFRAL, e.g., in the ERK phosphorylation assay.
  • GFRAL is preferably the only antigen bound by the antigen binding site of the binder polypeptide.
  • a binder polypeptide may optionally be engineered to comprise further binding sites, and an antibody comprising an antibody constant region may for example optionally bind one or more Fc receptors.
  • an inhibitor of GDF15 signalling e.g., an anti-GFRAL antibody
  • a suitable assay is the ERK phosphorylation assay.
  • ERK phosphorylation assay In this assay, cells co expressing GFRAL and RET at their cell surface are incubated with GDF15, resulting in formation of the GDF15-GFRAL-RET signalling complex and consequent downstream signalling including phosphorylation of ERK.
  • ERK phosphorylation may be quantified in lysed cells, e.g., by using FITRF and detecting change in fluorescence.
  • addition of GDF15 in a dilution series generates a sigmoid curve ( Figure 3).
  • the effect of adding inhibitor in this assay may be quantified by its effect on the change in fluorescence detected by FITRF.
  • the skilled person will include appropriate controls and will calibrate the assay to establish suitable concentrations of reagents.
  • the GFRAL/RET expressing cells may be exposed to a range of GDF15 concentrations and inhibition at each concentration determined, before selecting a suitable concentration. For example, we found a concentration of 4 nM produced maximal response in our experiments and thus selected 4 nM as the GDF15 concentration to use for this assay. More details are provided in Example 1 .
  • An inhibitor of GDF15 signalling may exhibit dose-dependent inhibition of GDF15- induced GFRAL signalling activity in the ERK phosphorylation assay, e.g., with human and/or non-human (e.g., mouse, rat or cynomolgus) GFRAL.
  • the ERK phosphorylation assay may be performed with the inhibitor at a range of concentrations, to produce a dose-response curve from which an IC50 value may be calculated.
  • an inhibitor according to the present invention may be identified through its dose-dependent inhibition in such an enzymatic assay.
  • Potency of the inhibitor may be quantified as IC50.
  • anti-GFRAL antibody may have an IC50 of 15 nM or less in such an assay. Potency of binder polypeptides such as anti-GFRAL antibodies may be compared for reference against one or more anti-GFRAL antibodies described herein.
  • an antibody comprising the VH and VL domains of QUEL-0101 , QUEL-0201 or QUEL-0301 may be used as a reference antibody.
  • the reference antibody may be provided as an IgG.
  • An inhibitor of GDF15 signalling may be one which has an IC50 within 50% or within 10% of the IC50 of QUEL-0101 , QUEL-0201 or QUEL-0301 IgG, or it may have an IC50 which is lower than the IC50 of said reference antibody.
  • IC50 within 50% or within 10% of the IC50 of QUEL-0101 , QUEL-0201 or QUEL-0301 IgG, or it may have an IC50 which is lower than the IC50 of said reference antibody.
  • within x% of it is meant that the IC50 of the test binder polypeptide is no more than x% greater than and no more than x% less than the IC50 of the reference antibody.
  • An anti-GFRAL antibody may have an IC50 of 10 nM or less in an ERK phosphorylation assay, optionally 5 nM or less.
  • binder polypeptides including antibodies
  • antibodies may be generated using laboratory animals such as mice, including transgenic mice (eg, the Kymouse®, Velocimouse®, Omnimouse® , Xenomouse®, HuMab Mouse® or MeMo Mouse®), rats (e.g., the Omnirat®), camelids, sharks, rabbits, chickens or other non-human animals immunised with GFRAL or its encoding nucleic acid, followed optionally by humanisation of the constant regions and/or variable regions to produce human or humanised antibodies.
  • display technologies can be used, such as yeast, phage or ribosome display, as will be apparent to the skilled person.
  • Standard affinity maturation e.g., using a display technology
  • a display technology can be performed in a further step after isolation of an antibody lead from a transgenic animal, phage display library or other library.
  • suitable technologies are described in US20120093818 (Amgen, Inc), which is incorporated by reference herein in its entirety, eg, the methods set out in paragraphs [0309] to [0346]
  • variants of a binder which include optimising a polypeptide sequence for large-scale manufacturing, facilitating purification, enhancing stability or improving suitability for inclusion in a desired pharmaceutical formulation.
  • Protein engineering work can be performed at one or more target residues in the antibody sequence, e.g., to substituting one amino acid with an alternative amino acid (optionally, generating variants containing all naturally occurring amino acids at this position, with the possible exception of Cys and Met), and monitoring the impact on function and expression to determine the best substitution.
  • An antibody may comprise a set of H and/or L CDRs of any of the disclosed antibodies with one or more amino acid mutations within the disclosed set of H and/or L CDRs.
  • the mutation may be an amino acid substitution, deletion or insertion.
  • An antibody may comprise the set of HCDRs, LCDRs or a set of 6 (H and L) CDRs shown for any QUEL antibody herein or may comprise that set of CDRs with one or two conservative substitutions.
  • One or more amino acid mutations may optionally be made in framework regions of an antibody VH or VL domain disclosed herein.
  • one or more residues that differ from the corresponding human germline segment sequence may be reverted to germline.
  • Human germline gene segment sequences corresponding to VH and VL domains of example anti- GFRAL antibodies are indicated in Table G.
  • An antibody may comprise a VH domain that has at least 60, 70, 80, 85, 90, 95, 98 or 99 % amino acid sequence identity with a VH domain of any of the antibodies shown in the appended sequence listing, and/or comprising a VL domain that has at least 60, 70, 80, 85, 90, 95, 98 or 99 % amino acid sequence identity with a VL domain of any of those antibodies.
  • Algorithms that can be used to calculate % identity of two amino acid sequences include e.g. BLAST, FASTA, or the Smith-Waterman algorithm, e.g. employing default parameters.
  • Particular variants may include one or more amino acid sequence alterations (addition, deletion, substitution and/or insertion of an amino acid residue).
  • Alterations may be made in one or more framework regions and/or one or more CDRs. Variants are optionally provided by CDR mutagenesis.
  • the alterations normally do not result in loss of function, so an antibody comprising a thus-altered amino acid sequence may retain an ability to bind human GFRAL and/or mouse GFRAL. It may retain the same quantitative binding ability as an antibody in which the alteration is not made, e.g. as measured in an assay described herein.
  • the antibody comprising a thus-altered amino acid sequence may have an improved ability to bind and/or inhibit human and/or mouse GFRAL.
  • Alteration may comprise replacing one or more amino acid residue with a non-naturally occurring or non-standard amino acid, modifying one or more amino acid residue into a non- naturally occurring or non-standard form, or inserting one or more non- naturally occurring or non-standard amino acid into the sequence. Examples of numbers and locations of alterations in sequences of the invention are described elsewhere herein.
  • Naturally occurring amino acids include the 20 "standard" L-amino acids identified as G, A, V, L, I, M, P, F, W, S, T, N, Q, Y, C, K, R, FI, D, E by their standard single-letter codes.
  • Non-standard amino acids include any other residue that may be incorporated into a polypeptide backbone or result from modification of an existing amino acid residue. Non-standard amino acids may be naturally occurring or non- naturally occurring.
  • variant refers to a peptide or nucleic acid that differs from a parent polypeptide or nucleic acid by one or more amino acid or nucleic acid deletions, substitutions or additions, yet retains one or more specific functions or biological activities of the parent molecule.
  • Amino acid substitutions include alterations in which an amino acid is replaced with a different naturally-occurring amino acid residue. Such substitutions may be classified as “conservative", in which case an amino acid residue contained in a polypeptide is replaced with another naturally occurring amino acid of similar character either in relation to polarity, side chain functionality or size. Such conservative substitutions are well known in the art.
  • substitutions encompassed by the present invention may also be "non-conservative", in which an amino acid residue which is present in a peptide is substituted with an amino acid having different properties, such as naturally-occurring amino acid from a different group (e.g., substituting a charged or hydrophobic amino; acid with alanine), or alternatively, in which a naturally-occurring amino acid is substituted with a non- conventional amino acid.
  • amino acid substitutions are conservative.
  • polynucleotide or polypeptide refers to a polynucleotide or polypeptide that can vary in primary, secondary, or tertiary structure, as compared to a reference polynucleotide or polypeptide, respectively (e.g., as compared to a wild- type polynucleotide or polypeptide).
  • “Modified variants” can include conservative or non conservative amino acid changes, as described below. Polynucleotide changes can result in amino acid substitutions, additions, deletions, fusions and truncations in the polypeptide encoded by the reference sequence.
  • Some aspects use include insertion variants, deletion variants or substituted variants with substitutions of amino acids, including insertions and substitutions of amino acids and other molecules) that do not normally occur in the peptide sequence that is the basis of the variant, for example but not limited to insertion of ornithine which do not normally occur in human proteins.
  • conservative substitution when describing a polypeptide, refers to a change in the amino acid composition of the polypeptide that does not substantially alter the polypeptide's activity. For example, a conservative substitution refers to substituting an amino acid residue for a different amino acid residue that has similar chemical properties (e.g., acidic, basic, positively or negatively charged, polar or nonpolar, etc.).
  • Conservative amino acid substitutions include replacement of a leucine with an isoleucine or valine, an aspartate with a glutamate, or a threonine with a serine.
  • Conservative substitution tables providing functionally similar amino acids are well known in the art.
  • the following six groups each contain amino acids that are conservative substitutions for one another: 1) Alanine (A), Serine (S), Threonine (T); 2) Aspartic acid (D), Glutamic acid (E); 3) Asparagine (N), Glutamine (Q); 4) Arginine (R), Lysine (K); 5) Isoleucine (I), Leucine (L), Methionine (M), Valine (V); and 6) Phenylalanine (F), Tyrosine (Y), Tryptophan (W). (See also Creighton, Proteins, W. FI.
  • substitutions suitable for amino acids on the exterior of a protein or peptide for example, but not limited to, the following substitutions can be used: substitution of Y with F, T with S or K, P with A, E with D or Q, N with D or G, R with K, G with N or A, T with S or K, D with N or E, I with L or V, F with Y, S with T or A, R with K, G with N or A, K with R, A with S, K or P.
  • non-conservative amino acid substitutions are also encompassed within the term of variants.
  • the invention includes methods of producing antibodies containing VH and/or VL domain variants of the antibody VH and/or VL domains shown in Table A.
  • Such antibodies may be produced by a method comprising
  • the VH domain may be the VH domain of QUEL-0201 .
  • Desired characteristics include binding to human and/or non-human GFRAL. Antibodies with comparable or higher affinity for human and/or mouse GFRAL relative to the parent antibody may be identified. Other desired characteristics include inhibition of GDF15 signalling assays described herein, e.g., ERK phosphorylation assay. Identifying an antibody with a desired characteristic may comprise identifying an antibody with a functional attribute described herein, such as its affinity, cross-reactivity, specificity, or neutralising potency, any of which may be determined in assays as described herein.
  • the VL domain may be a VL domain of any of QUEL-0101 , QUEL-0201 or QUEL-0301 , or may be a variant provided by way of addition, deletion, substitution or insertion of one or more amino acids in the amino acid sequence of a parent VL domain, wherein the parent VL domain is the VL domain of any of QUEL-0101 , QUEL-0201 or QUEL-0301 or a VL domain comprising the light chain complementarity determining regions of any of those antibodies.
  • the VL domain may be the VL domain of the same antibody as the VH domain. It may be the VL domain of QUEL-0201 .
  • Methods of generating variant antibodies may optionally comprise producing copies of the antibody or VH/VL domain combination. Methods may further comprise expressing the resultant antibody. It is possible to produce nucleotide sequences corresponding to a desired antibody VH and/or VL domain, optionally in one or more expression vectors. Suitable methods of expression, including recombinant expression in host cells, are set out in detail herein.
  • Isolated nucleic acid may be provided, encoding antibodies according to the present invention.
  • Nucleic acid may be DNA and/or RNA. Genomic DNA, cDNA, mRNA or other RNA, of synthetic origin, or any combination thereof can encode an antibody.
  • the present invention provides constructs in the form of plasmids, vectors, transcription or expression cassettes which comprise at least one polynucleotide as above.
  • Exemplary nucleotide sequences are included in the sequence listing. Reference to a nucleotide sequence as set out herein encompasses a DNA molecule with the specified sequence, and encompasses a RNA molecule with the specified sequence in which U is substituted for T, unless context requires otherwise.
  • the present invention also provides a recombinant host cell that comprises one or more nucleic acids encoding the antibody.
  • Methods of producing the encoded antibody may comprise expression from the nucleic acid, e.g., by culturing recombinant host cells containing the nucleic acid.
  • the antibody may thus be obtained, and may be isolated and/or purified using any suitable technique, then used as appropriate.
  • a method of production may comprise formulating the product into a composition including at least one additional component, such as a pharmaceutically acceptable excipient.
  • Suitable host cells include bacteria, mammalian cells, plant cells, filamentous fungi, yeast and baculovirus systems and transgenic plants and animals.
  • Vectors may contain appropriate regulatory sequences, including promoter sequences, terminator sequences, polyadenylation sequences, enhancer sequences, marker genes and other sequences as appropriate.
  • Nucleic acid encoding an antibody can be introduced into a host cell.
  • Nucleic acid of the invention may be integrated into the genome (e.g. chromosome) of the host cell. Integration may be promoted by inclusion of sequences that promote recombination with the genome, in accordance with standard techniques.
  • Nucleic acid can be introduced to eukaryotic cells by various methods, including calcium phosphate transfection, DEAE-Dextran, electroporation, liposome-mediated transfection and transduction using retrovirus or other virus, e.g. vaccinia or, for insect cells, baculovirus.
  • Introducing nucleic acid in the host cell may use a viral or a plasmid based system.
  • the plasmid system may be maintained episomally or may be incorporated into the host cell or into an artificial chromosome. Incorporation may be either by random or targeted integration of one or more copies at single or multiple loci.
  • suitable techniques include calcium chloride transformation, electroporation and transfection using bacteriophage.
  • the introduction may be followed by expressing the nucleic acid, e.g., by culturing host cells under conditions for expression of the gene, then optionally isolating or purifying the binder polypeptide, e.g., antibody.
  • compositions comprising inhibitors of GDF15 signalling, e.g., anti-GFRAL antibodies, according to the present invention.
  • Compositions are also provided comprising nucleic acid encoding inhibitors of GDF15 signalling that are binder polypeptides, e.g., antibodies.
  • Such compositions may be provided for use in treatment of the human or animal body by therapy, including in any of the example medical treatments described herein.
  • the compositions may further comprise, in addition to the active ingredient (inhibitor or encoding nucleic acid), one or more pharmaceutically acceptable excipients.
  • Binder polypeptides according to the present invention, and their encoding nucleic acid molecules, will usually be provided in isolated form.
  • VFI and/or VL domains, and nucleic acids may be provided purified from their natural environment or their production environment.
  • Isolated binder polypepides and isolated nucleic acid will be free or substantially free of material with which they are naturally associated, such as other polypeptides or nucleic acids with which they are found in vivo, or the environment in which they are prepared (e.g., cell culture) when such preparation is by recombinant DNA technology in vitro.
  • an isolated binder polypeptide or nucleic acid (1) is free of at least some other proteins with which it would normally be found, (2) is essentially free of other proteins from the same source, e.g., from the same species, (3) is expressed by a cell from a different species, (4) has been separated from at least about 50 percent of polynucleotides, lipids, carbohydrates, or other materials with which it is associated in nature, (5) is operably associated (by covalent or noncovalent interaction) with a polypeptide with which it is not associated in nature, or (6) does not occur in nature.
  • Binder polypeptides or their encoding nucleic acids may be formulated with diluents or adjuvants and still for practical purposes be isolated - for example they may be mixed with carriers if used to coat microtitre plates for use in immunoassays, and may be mixed with pharmaceutically acceptable carriers or diluents when used in therapy. As described elsewhere herein, other active ingredients may also be included in therapeutic preparations.
  • the binder polypeptide may be glycosylated, either naturally in vivo or by systems of heterologous eukaryotic cells such as CHO cells, or it may be (for example if produced by expression in a prokaryotic cell) unglycosylated.
  • the invention encompasses antibodies having a modified glycosylation pattern.
  • an isolated product constitutes at least about 5%, at least about 10%, at least about 25%, or at least about 50% of a given sample.
  • a binder polypeptide may be substantially free from proteins or polypeptides or other contaminants that are found in its natural or production environment that would interfere with its therapeutic, diagnostic, prophylactic, research or other use.
  • the invention provides therapeutic compositions comprising the binder polypeptides described herein.
  • Therapeutic compositions comprising nucleic acid encoding such binder polypeptides are also provided. Encoding nucleic acids are described in more detail elsewhere herein and include DNA and RNA, e.g., mRNA.
  • use of nucleic acid encoding the binder polypeptide, and/or of cells containing such nucleic acid may be used as alternatives (or in addition) to compositions comprising the binder polypeptide itself.
  • Cells containing nucleic acid encoding the binder polypeptide, optionally wherein the nucleic acid is stably integrated into the genome thus represent medicaments for therapeutic use in a patient.
  • Nucleic acid encoding the binder polypeptide may be introduced into human cells derived from the intended patient and modified ex vivo. Administration of cells containing the encoding nucleic acid to the patient provides a reservoir of cells capable of expressing the binder polypeptide, which may provide therapeutic benefit over a longer term compared with administration of isolated nucleic acid or the isolated binder polypeptide. Nucleic acid may also be administered directly to the patient for gene therapy. Thus, nucleic acid encoding the binder polypeptide may be provided for use in gene therapy, comprising introducing the encoding nucleic acid into cells of the patient in vivo, so that the nucleic acid is expressed in the patient’s cells and provides a therapeutic effect, examples of which are disclosed herein.
  • compositions may contain suitable carriers, excipients, and other agents that are incorporated into formulations to provide improved transfer, delivery, tolerance, and the like.
  • suitable carriers excipients, and other agents that are incorporated into formulations to provide improved transfer, delivery, tolerance, and the like.
  • a multitude of appropriate formulations can be found in the formulary known to all pharmaceutical chemists: Remington's Pharmaceutical Sciences, Mack Publishing Company, Easton, Pa.
  • formulations include, for example, powders, pastes, ointments, jellies, waxes, oils, lipids, lipid (cationic or anionic) containing vesicles (such as LIPOFECTINTTM), DNA conjugates, anhydrous absorption pastes, oil-in-water and water-in-oil emulsions, emulsions carbowax (polyethylene glycols of various molecular weights), semi-solid gels, and semi-solid mixtures containing carbowax.
  • vesicles such as LIPOFECTINTTM
  • Binder polypeptides, or their encoding nucleic acids may be formulated for the desired route of administration to a patient, e.g., in liquid (optionally aqueous solution) for injection.
  • the composition may optionally be formulated for intravenous or subcutaneous injection.
  • compositions of the invention include, but are not limited to, intradermal, intramuscular, intraperitoneal, intravenous, subcutaneous, intranasal, epidural, and oral routes.
  • the composition may be administered by any convenient route, for example by infusion or bolus injection, by absorption through epithelial or mucocutaneous linings (e.g., oral mucosa, rectal and intestinal mucosa, etc.) and may be administered together with other biologically active agents. Administration can be systemic or local.
  • the antigen-binding molecules are preferably administered by subcutaneous injection. Administration may be self-administration by a patient, e.g., self-injection.
  • the pharmaceutical composition can be also delivered in a vesicle, in particular a liposome (see Langer (1990) Science 249:1527-1533 ; Treat et al. (1989) in Liposomes in the Therapy of Infectious Disease and Cancer, Lopez Berestein and Fidler (eds.), Liss, New York, pp. 353-365 ; Lopez-Berestein, ibid., pp. 317-327 ; see generally ibid.).
  • a liposome see Langer (1990) Science 249:1527-1533 ; Treat et al. (1989) in Liposomes in the Therapy of Infectious Disease and Cancer, Lopez Berestein and Fidler (eds.), Liss, New York, pp. 353-365 ; Lopez-Berestein, ibid., pp. 317-327 ; see generally ibid.).
  • the pharmaceutical composition can be delivered in a controlled release system.
  • a pump may be used (see Langer, supra; Sefton (1987) CRC Crit. Ref. Biomed. Eng. 14:201).
  • polymeric materials can be used; see, Medical Applications of Controlled Release, Langer and Wise (eds.), CRC Pres., Boca Raton, Fla. (1974).
  • a controlled release system can be placed in proximity of the composition's target, thus requiring only a fraction of the systemic dose (see, e.g., Goodson, in Medical Applications of Controlled Release, supra, vol. 2, pp. 115-138, 1984).
  • the injectable preparations may include dosage forms for intravenous, subcutaneous, intracutaneous and intramuscular injections, drip infusions, etc. These injectable preparations may be prepared by methods publicly known. For example, the injectable preparations may be prepared, e.g., by dissolving, suspending or emulsifying the antibody or its salt described above in a sterile aqueous medium or an oily medium conventionally used for injections.
  • aqueous medium for injections there are, for example, physiological saline, an isotonic solution containing glucose and other auxiliary agents, etc., which may be used in combination with an appropriate solubilizing agent such as an alcohol (e.g., ethanol), a polyalcohol (e.g., propylene glycol, polyethylene glycol), a nonionic surfactant [e.g., polysorbate 80, HCO-50 (polyoxyethylene (50 mol) adduct of hydrogenated castor oil)], etc.
  • an alcohol e.g., ethanol
  • a polyalcohol e.g., propylene glycol, polyethylene glycol
  • a nonionic surfactant e.g., polysorbate 80, HCO-50 (polyoxyethylene (50 mol) adduct of hydrogenated castor oil
  • a pharmaceutical composition of the present invention can be delivered subcutaneously or intravenously with a standard needle and syringe. It is envisaged that treatment will not be restricted to use in the clinic. Therefore, subcutaneous injection using a needle-free device is also advantageous.
  • a pen delivery device readily has applications in delivering a pharmaceutical composition of the present invention. Such a pen delivery device can be reusable or disposable.
  • a reusable pen delivery device generally utilizes a replaceable cartridge that contains a pharmaceutical composition. Once all of the pharmaceutical composition within the cartridge has been administered and the cartridge is empty, the empty cartridge can readily be discarded and replaced with a new cartridge that contains the pharmaceutical composition. The pen delivery device can then be reused. In a disposable pen delivery device, there is no replaceable cartridge. Rather, the disposable pen delivery device comes prefilled with the pharmaceutical composition held in a reservoir within the device. Once the reservoir is emptied of the pharmaceutical composition, the entire device is discarded. Numerous reusable pen and autoinjector delivery devices have applications in the subcutaneous delivery of a pharmaceutical composition of the present invention.
  • Examples include, but certainly are not limited to AUTOPENTM (Owen Mumford, Inc., Woodstock, UK), DISETRONICTM pen (Disetronic Medical Systems, Burghdorf, Switzerland), HUMALOG MIX 75/25TM pen, HUMALOGTM pen, HUMALIN 70/30TM pen (Eli Lilly and Co., Indianapolis, Ind.), NOVOPENTM!, II and III (Novo Nordisk, Copenhagen, Denmark), NOVOPEN JUNIORTM (Novo Nordisk, Copenhagen, Denmark), BDTM pen (Becton Dickinson, Franklin Lakes, N.J.), OPTIPENTTM, OPTIPEN PROTM, OPTIPEN STARLETTM, and OPTICLIKTTM (Sanofi-Aventis, Frankfurt, Germany), to name only a few.
  • Examples of disposable pen delivery devices having applications in subcutaneous delivery of a pharmaceutical composition of the present invention include, but certainly are not limited to the SOLOSTARTM pen (Sanofi-Aventis), the FLEXPENTM (Novo Nordisk), and the KWIKPENTM (Eli Lilly).
  • the pharmaceutical compositions for oral or parenteral use described above are prepared into dosage forms in a unit dose suited to fit a dose of the active ingredients.
  • dosage forms in a unit dose include, for example, tablets, pills, capsules, injections (ampoules), suppositories, etc.
  • the amount of the aforesaid antibody contained is generally about 5 to about 500 mg per dosage form in a unit dose; especially in the form of injection, the aforesaid antibody may be contained in about 5 to about 100 mg and in about 10 to about 250 mg for the other dosage forms.
  • the binder polypeptide, nucleic acid, or composition comprising it may be contained in a medical container such as a phial, syringe, IV container or an injection device.
  • a medical container such as a phial, syringe, IV container or an injection device.
  • the binder polypeptide, nucleic acid or composition is in vitro, and may be in a sterile container.
  • a kit comprising the binder polypeptide, packaging and instructions for use in a therapeutic method as described herein.
  • compositions comprising a binder polypeptide or nucleic acid of the invention and one or more pharmaceutically acceptable excipients, examples of which are listed above.
  • “Pharmaceutically acceptable” refers to approved or approvable by a regulatory agency of the USA Federal or a state government or listed in the U.S. Pharmacopeia or other generally recognized pharmacopeia for use in animals, including humans.
  • a pharmaceutically acceptable carrier, excipient, or adjuvant can be administered to a patient, together with a binder polypeptide, e.g., any antibody or polypeptide molecule described herein, and does not destroy the pharmacological activity thereof and is nontoxic when administered in doses sufficient to deliver a therapeutic amount of the agent.
  • the binder polypeptide will be the sole active ingredient in a composition according to the present invention.
  • a composition may consist of the antibody or it may consist of the binder polypeptide with one or more pharmaceutically acceptable excipients.
  • compositions according to the present invention optionally include one or more additional active ingredients.
  • Other therapeutic agents that it may be desirable to administer with binder polypeptides or nucleic acids according to the present invention include other therapeutic agents for cancer, examples of which are described herein. Any such agent or combination of agents may be administered in combination with, or provided in compositions with binder polypeptides or nucleic acids according to the present invention, whether as a combined or separate preparation.
  • the binder polypeptide or nucleic acid according to the present invention may be administered separately and sequentially, or concurrently and optionally as a combined preparation, with another therapeutic agent or agents such as those mentioned herein.
  • compositions can be administered separately or simultaneously. Separate administration refers to the two compositions being administered at different times, e.g. at least 10, 20, 30, or 10-60 minutes apart, or 1 , 2, 3, 4, 5, 6, 7, 8, 9, 10, 12 hours apart. One can also administer compositions at 24 hours apart, or even longer apart. Alternatively, two or more compositions can be administered simultaneously, e.g. less than 10 or less than 5 minutes apart. Compositions administered simultaneously can, in some aspects, be administered as a mixture, with or without similar or different time release mechanism for each of the components.
  • Binder polypeptides and their encoding nucleic acids, can be used as therapeutic agents. Patients herein are generally mammals, typically humans. A binder polypeptide or nucleic acid may be administered to a mammal, e.g., by any route of administration mentioned herein. In a preferred embodiment, a binder polypeptide is administered by subcutaneous injection.
  • Administration is normally in a "therapeutically effective amount", this being an amount that produces the desired effect for which it is administered, sufficient to show benefit to a patient.
  • the exact amount will depend on the purpose of the treatment, and will be ascertainable by one skilled in the art using known techniques (see, for example, Lloyd (1999) The Art, Science and Technology of Pharmaceutical Compounding). Prescription of treatment, e.g. decisions on dosage etc, is within the responsibility of general practitioners and other medical doctors and may depend on the severity of the symptoms and/or progression of a disease being treated.
  • a therapeutically effective amount or suitable dose of binder polypeptide or nucleic acid can be determined by comparing its in vitro activity and in vivo activity in an animal model. Methods for extrapolation of effective dosages in mice and other test animals to humans are known.
  • one or more doses may be administered.
  • a single dose may be effective to achieve a long-term benefit.
  • the method may comprise administering a single dose of the binder polypeptide, its encoding nucleic acid, or the composition.
  • multiple doses may be administered, usually sequentially and separated by a period of days, weeks or months.
  • administration may be every 2 weeks, every 3 weeks or every 4 weeks.
  • the binder polypeptide may be administered to a patient once a month, or less frequently, e.g., every two months or every three months.
  • the terms “treat,” “treatment,” “treating,” or “amelioration” refer to therapeutic treatments, wherein the object is to reverse, alleviate, ameliorate, inhibit, slow down or stop the progression or severity of a condition associated with a disease or disorder.
  • the term “treating” includes reducing or alleviating at least one adverse effect or symptom of a condition, disease or disorder.
  • Treatment is generally “effective” if one or more symptoms or clinical markers are reduced.
  • treatment is “effective” if the progression of a disease is reduced or halted. That is, “treatment” includes not just the improvement of symptoms or markers, but also a cessation of, or at least slowing of, progress or worsening of symptoms compared to what would be expected in the absence of treatment.
  • Beneficial or desired clinical results include, but are not limited to, alleviation of one or more symptom(s), diminishment of extent of disease, stabilised (i.e., not worsening) state of disease, delay or slowing of disease progression, amelioration or palliation of the disease state, remission (whether partial or total), and/or decreased mortality, whether detectable or undetectable.
  • treatment also includes providing relief from the symptoms or side-effects of the disease (including palliative treatment). For treatment to be effective a complete cure is not contemplated. The method can in certain aspects include cure as well. In the context of the invention, treatment may be preventative treatment.
  • Long half-life is a desirable feature in the binder polypeptides of the present invention. Extended half-life translates to less frequent administration, with fewer injections being required to maintain a therapeutically effective concentration of the molecule in the bloodstream.
  • the in vivo half life of antigen-binding molecules of the present invention in humans may be 7, 8, 9, 10,
  • the in vivo half life of antigen binding molecules in non-human primates such as cynomolgus monkeys may be 7, 8, 9, 10, 11,
  • Binder polypeptides may be provided for administration at regular intervals of one week, two weeks, three weeks, four weeks, or one month.
  • An antibody that binds human GFRAL comprising an antibody heavy chain variable (VH) domain and an antibody light chain variable (VL) domain, the VFI domain comprising the QUEL-0201 set of heavy chain complementarity determining regions (HCDRs) HCDR1 SEQ ID NO: 13, HCDR2 SEQ ID NO: 14 and HCDR3 SEQ ID NO: 15, and the VL domain comprising the QUEL-0201 set of light chain complementarity determining regions (LCDRs) LCDR1 SEQ ID NO: 18, LCDR2 SEQ ID NO: 19 and LCDR3 SEQ ID NO: 20.
  • VH antibody heavy chain variable
  • VL antibody light chain variable
  • An antibody that binds human GFRAL comprising a VH domain having at least 90 % amino acid sequence identity to the QUEL-0201 VH domain SEQ ID NO: 12 and a VL domain having at least 90 % amino acid sequence identity to the QUEL-0201 VL domain SEQ ID NO: 17.
  • An antibody that binds human GFRAL comprising a VFI domain encoded by a nucleotide sequence produced by recombination of gene segments IGHV1-3 (e.g., IGHV1-3 * 01) and IGHJ6 (e.g., IGHJ6 * 02), and a VL domain encoded by a nucleotide sequence produced by recombination of gene segments IGLV1-40 (e.g., IGLV1 -40 * 01) and IGLJ3 (e.g., IGLJ3 * 02).
  • VFI domain is encoded by a nucleotide sequence produced by recombination of gene segments IGHV1 -3 (e.g., IGHV1 -3 * 01 ), IGHD5- 18 (e.g., IGHD5-18 * 01) and IGHJ6 (e.g., IGHJ6 * 02).
  • An antibody that competes for binding human GFRAL with a QUEL-0201 IgG comprising QUEL-0201 VH domain SEQ ID NO: 12 and QUEL-0201 VL domain SEQ ID NO: 17.
  • An antibody according to any of clauses 1 to 11 comprising a VH domain having at least 95 % amino acid sequence identity to the QUEL-0201 VH domain SEQ ID NO: 12 and a VL domain having at least 95 % amino acid sequence identity to the QUEL-0201 VL domain SEQ ID NO: 17. 13. An antibody according to any preceding clause, comprising a VH domain having at least 98 % amino acid sequence identity to the QUEL-0201 VH domain SEQ ID NO: 12 and a VL domain having at least 98 % amino acid sequence identity to the QUEL-0201 VL domain SEQ ID NO: 17.
  • An antibody according to clause 13 comprising the QUEL-0201 VH domain SEQ ID NO: 12, optionally with one or two amino acid alterations and the QUEL-0201 VL domain SEQ ID NO: 17, optionally with one or two amino acid alterations.
  • An antibody according to clause 14 comprising the QUEL-0201 VH domain SEQ ID NO: 12 comprising the QUEL-0201 set of HCDRs HCDR1 SEQ ID NO: 13, HCDR2 SEQ ID NO: 14 and HCDR3 SEQ ID NO: 15, optionally with one or two amino acid alterations in the VH domain framework, and the QUEL-0201 VL domain SEQ ID NO: 17 comprising the QUEL-0201 set of LCDRs LCDR1 SEQ ID NO: 18, LCDR2 SEQ ID NO: 19 and LCDR3 SEQ ID NO: 20, optionally with one or two amino acid alterations in the VL domain framework.
  • An antibody that binds human GFRAL comprising an antibody heavy chain variable (VH) domain and an antibody light chain variable (VL) domain, the VH domain comprising a set of heavy chain complementarity determining regions (HCDRs), wherein HCDR1 is SEQ ID NO: 23 or SEQ ID NO: 133, HCDR2 is SEQ ID NO: 24, SEQ ID NO: 124, SEQ ID NO: 134 or SEQ ID NO: 142, and HCDR3 is SEQ ID NO: 25, SEQ ID NO: 125, SEQ ID NO: 135 or SEQ ID NO: 143, and the VL domain comprising a set of light chain complementarity determining regions (LCDRs), wherein LCDR1 is SEQ ID NO: 28, SEQ ID NO: 128 or SEQ ID NO: 138, LCDR2 is SEQ ID NO: 29 or SEQ ID NO: 129 and LCDR3 is SEQ ID NO: 30, SEQ ID NO: 130, SEQ ID NO: 139 or SEQ ID NO: 146
  • An antibody that binds human GFRAL comprising an antibody heavy chain variable (VH) domain and an antibody light chain variable (VL) domain, the VH domain comprising the QUEL-0301 set of heavy chain complementarity determining regions (HCDRs) HCDR1 SEQ ID NO: 23, HCDR2 SEQ ID NO: 24 and HCDR3 SEQ ID NO: 25, and the VL domain comprising the QUEL-0301 set of light chain complementarity determining regions (LCDRs) LCDR1 SEQ ID NO: 28, LCDR2 SEQ ID NO: 29 and LCDR3 SEQ ID NO: 30.
  • VH antibody heavy chain variable
  • VL antibody light chain variable
  • An antibody that binds human GFRAL comprising a VH domain and a VL domain, the VH domain comprising the QUEL-0302 set of HCDRs HCDR1 SEQ ID NO: 23,
  • HCDR2 SEQ ID NO: 124 and HCDR3 SEQ ID NO: 125 and the VL domain comprising the QUEL-0302 set of LCDRs LCDR1 SEQ ID NO: 128, LCDR2 SEQ ID NO: 129 and LCDR3 SEQ ID NO: 130.
  • An antibody that binds human GFRAL comprising a VH domain and a VL domain, the VH domain comprising the QUEL-0303 set of HCDRs HCDR1 SEQ ID NO: 133,
  • An antibody that binds human GFRAL comprising a VH domain and a VL domain, the VH domain comprising the QUEL-0304 set of HCDRs HCDR1 SEQ ID NO: 133,
  • HCDR2 SEQ ID NO: 142 and HCDR3 SEQ ID NO: 143 and the VL domain comprising the QUEL-0304 set of LCDRs LCDR1 SEQ ID NO: 138, LCDR2 SEQ ID NO: 29 and LCDR3 SEQ ID NO: 146.
  • An antibody that binds human GFRAL comprising a VH domain having at least 90 % amino acid sequence identity to the QUEL-0301 VH domain SEQ ID NO: 22 and a VL domain having at least 90 % amino acid sequence identity to the QUEL-0301 VL domain SEQ ID NO: 27.
  • An antibody that binds human GFRAL comprising a VH domain encoded by a nucleotide sequence produced by recombination of gene segments IGHV3-7 (e.g., IGHV3-7 * 01) and IGHJ4 (e.g., IGHJ4 * 02), and a VL domain encoded by a nucleotide sequence produced by recombination of gene segments IGLV1-44 (e.g., IGLV1 -44 * 01) and IGLJ3 (e.g., IGLJ3 * 02).
  • VH domain is encoded by a nucleotide sequence produced by recombination of gene segments IGHV3-7 (e.g., IGHV3-7 * 01), IGHD1-7 (e.g., IGHD1-7 * 01) and IGHJ4 (e.g., IGHJ4 * 02).
  • An antibody that binds human GFRAL comprising a VH domain encoded by a nucleotide sequence produced by recombination of gene segments IGHV3-7 (e.g., IGHV3-7 * 01) and IGHJ4 (e.g., IGHJ4 * 02), and a VL domain encoded by a nucleotide sequence produced by recombination of gene segments IGLV1-47 (e.g., IGLV1 -47 * 01) and IGLJ3 (e.g., IGLJ3 * 02).
  • VH domain is encoded by a nucleotide sequence produced by recombination of gene segments IGHV3-7 (e.g., IGHV3-7 * 01), IGHD1- 20 (e.g., IGHD1 -20 * 01) and IGHJ4 (e.g., IGHJ4 * 02).
  • IGHV3-7 e.g., IGHV3-7 * 01
  • IGHD1- 20 e.g., IGHD1 -20 * 01
  • IGHJ4 e.g., IGHJ4 * 02
  • An antibody that binds human GFRAL comprising an antibody heavy chain variable (VH) domain and an antibody light chain variable (VL) domain, the VH domain comprising a set of HCDRs HCDR1 , HCDR2 and HCDR3, wherein HCDR1 is SEQ ID NO: 3, SEQ ID NO: 99, SEQ ID NO: 107 or SEQ ID NO: 118,
  • HCDR2 is SEQ ID NO: 4
  • HCDR3 is SEQ ID NO: 5
  • SEQ ID NO: 101 SEQ ID NO: 113 or SEQ ID NO: 119
  • the VL domain comprising a set of LCDRs LCDR1 , LCDR2 and LCDR3, wherein LCDR1 is SEQ ID NO: 8,
  • LCDR2 is SEQ ID NO: 9 and
  • LCDR3 is SEQ ID NO: 10 or SEQ ID NO: 104.
  • An antibody that binds human GFRAL comprising an antibody heavy chain variable (VH) domain and an antibody light chain variable (VL) domain, the VH domain comprising the QUEL-0101 set of heavy chain complementarity determining regions (HCDRs) HCDR1 SEQ ID NO: 3, HCDR2 SEQ ID NO: 4 and HCDR3 SEQ ID NO: 5, and the VL domain comprising the QUEL-0101 set of light chain complementarity determining regions (LCDRs) LCDR1 SEQ ID NO: 8, LCDR2 SEQ ID NO: 9 and LCDR3 SEQ ID NO: 10.
  • VH antibody heavy chain variable
  • VL antibody light chain variable
  • An antibody that binds human GFRAL comprising a VH domain and a VL domain, the VH domain comprising the QUEL-0102 set of HCDRs HCDR1 SEQ ID NO: 99,
  • HCDR2 SEQ ID NO: 100 and HCDR3 SEQ ID NO: 101 and the VL domain comprising the QUEL-0102 set of LCDRs LCDR1 SEQ ID NO: 8, LCDR2 SEQ ID NO: 9 and LCDR3 SEQ ID NO: 104.
  • An antibody that binds human GFRAL comprising a VH domain and a VL domain, the VH domain comprising the QUEL-0103 set of HCDRs HCDR1 SEQ ID NO: 107,
  • HCDR2 SEQ ID NO: 108 and HCDR3 SEQ ID NO: 5 and the VL domain comprising the QUEL-0103 set of LCDRs LCDR1 SEQ ID NO: 8, LCDR2 SEQ ID NO: 9 and LCDR3 SEQ ID NO: 104.
  • An antibody that binds human GFRAL comprising a VH domain and a VL domain, the VH domain comprising the QUEL-0104 set of HCDRs HCDR1 SEQ ID NO: 107,
  • HCDR2 SEQ ID NO: 108 and HCDR3 SEQ ID NO: 113 and the VL domain comprising the QUEL-0104 set of LCDRs LCDR1 SEQ ID NO: 8, LCDR2 SEQ ID NO: 9 and LCDR3 SEQ ID NO: 104.
  • An antibody that binds human GFRAL comprising a VH domain and a VL domain, the VH domain comprising the QUEL-0105 set of HCDRs HCDR1 SEQ ID NO: 118,
  • HCDR2 SEQ ID NO: 108 and HCDR3 SEQ ID NO: 119 and the VL domain comprising the QUEL-0105 set of LCDRs LCDR1 SEQ ID NO: 8, LCDR2 SEQ ID NO: 9 and LCDR3 SEQ ID NO: 104.
  • An antibody that binds human GFRAL comprising a VH domain having at least 90 % amino acid sequence identity to the QUEL-0101 VH domain SEQ ID NO: 2 and a VL domain having at least 90 % amino acid sequence identity to the QUEL-0101 VL domain SEQ ID NO: 7.
  • An antibody that binds human GFRAL comprising a VH domain encoded by a nucleotide sequence produced by recombination of gene segments IGHV3-30 (e.g., IGHV3-30 * 18) and IGHJ6 (e.g., IGHJ6 * 02), and a VL domain encoded by a nucleotide sequence produced by recombination of gene segments IGKV1-27 (e.g., IGKV1 -27 * 01) and IGKJ4 (e.g., IGKJ4 * 01). 52.
  • VH domain is encoded by a nucleotide sequence produced by recombination of gene segments IGHV3-30 * 18 (e.g., IGHV3-30), IGHD3-10 (e.g., IGHD3-10 * 01) and IGHJ6 (e.g., IGHJ6 * 02).
  • a host cell in vitro comprising nucleic acid as defined in clause 75.
  • composition comprising an antibody according to any of clauses 1 to 74 or nucleic acid according to clause 75, formulated with a pharmaceutically acceptable excipient.
  • composition according to clause 77 formulated for intravenous or subcutaneous injection.
  • a method of treating a medical condition associated with the GDF15-GFRAL pathway in a patient comprising administering a composition according to clause 77 or clause 78 to the patient.
  • Example 1 In vitro ERK phosphorylation assay for inhibition of GFRAL sionallino
  • FIEK293 cells were engineered to express DNA encoding human GFRAL SEQ ID NO:
  • Fluman GDF15 (recombinant human GDF15 protein with 6-His tag (R&D Systems, 957-GD)) was added to 96-well cell culture containing the following FIEK293 cell lines:
  • FIEK293 cells expressing human GFRAL only FIEK293 cells expressing human RET only FIEK293 cells co-expressing human GFRAL and RET The cells were incubated with GDF154 nM, then medium was removed and cell samples were lysed. Cell lysates were transferred into 384 well plates and an HTRF assay was performed to measure the phosphorylation of ERK gene product, representing the downstream signal of the GDF15-GFRAL-RET pathway. When RET is activated by GDF15-GFRAL tetramer, it triggers phosphorylation of ERK, which is detectable by FITRF.
  • anti-GFRAL antagonistic antibody mAb Q was included for comparison.
  • QUEL-0101 , QUEL-0201 and QUEL-0301 showed a good range of affinity to human GFRAL as determined by SPR.
  • Binding of antibodies to human GFRAL was measured by SPR using His-tagged human GFRAL as analyte, and test antibody coupled to the surface of a CM4 biosensor chip via immobilised anti-human Fc antibody.
  • Running buffer was HBS-P+ buffer pH 7.4 (GE BR100671) with 1 mM CaCh.
  • Antibodies were captured for 60 seconds at 1 pg/ml ( ⁇ 80 - 140 RU captured, depending on the binder).
  • Analyte recombinant soluble GFRAL protein, R&D catalogue no.
  • QUEL-0301 had the highest affinity for human GFRAL at approximately 1 picomolar (pM), stronger than the reference antibody mAb Q which had a measured affinity of 4.3 pM.
  • the ⁇ 4-fold difference between these antibodies indicates a comparable affinity (2-fold difference is within the experimental error of the Biacore apparatus).
  • the indicated KD value is merely an estimate due to the very slow dissociation of antibody
  • QUEL-0301 with a kd approaching the measurement limit of the apparatus.
  • QUEL-0201 bound human GFRAL with an affinity of approximately 84 pM.
  • QUEL-0101 bound human GFRAL with an affinity of 140 pM.
  • the QUEL antibodies have very good kinetic characeristics. Their kinetic constants are in the range that one would aspire to in a potent therapeutic antibody against a receptor.
  • the Koff and Kon values are very informative here, showing that the antibodies bind very fast to GFRAL, and stay attached for a long time.
  • Affinity for mouse GFRAL was determined by SPR by the same method as in Example 2 using a mouse GFRAL-Fc fusion (R&D catalogue no. 9844-GR-050) as analyte.
  • Figure 6 Affinity for mouse GFRAL was determined by SPR by the same method as in Example 2 using a mouse GFRAL-Fc fusion (R&D catalogue no. 9844-GR-050) as analyte.
  • KD and other kinetic constants for binding to mouse GFRAL were calculated and results are shown in the table below.
  • QUEL-0201 had the highest affinity for mouse GFRAL at approximately 64.8 pM
  • QUEL-0101 had an affinity of approximately 1.2 nanomolar (nM)
  • QUEL-0301 had relatively low affinity at just under 5 nM.
  • QUEL-0201 Comparing the measured affinities of these antibodies for human and mouse GFRAL, QUEL-0201 has greatest cross-reactivity, having an affinity difference within 1.5-fold for human and mouse GFRAL (human ⁇ 84 pM; mouse ⁇ 65 pM). QUEL-0101 also had good cross reactivity, within 10-fold, although the absolute affinities were lower (human -0.14 nM; mouse -1 .2 nM). QUEL-0301 was also cross-reactive but showed relatively weak binding to mouse GFRAL in contrast to its high affinity for human GFRAL (human -1 pM; mouse -5 nM).
  • Epitope mapping assays were performed for selected antagonistic mAbs to investigate possible mechanisms of inhibiting GDF15-GFRAL-RET signalling.
  • pairs of antibodies are tested for their ability to simultaneously bind antigen.
  • a first antibody is coupled via its Fc region to a solid support and the GFRAL antigen is added in solution, allowing formation of an antibody-antigen complex.
  • a second antibody is then added in solution. If binding of the second antibody is detected, this indicates that the first and second antibody bind different epitopes. If binding of the second antibody is not detected, this indicates that the first and second antibody compete for binding to the same epitope.
  • the same principle may be used to assess whether an antibody and a native ligand compete for binding to antigen.
  • a test antibody is coupled to a solid support and the GFRAL antigen is added in solution, allowing formation of an antibody-GFRAL complex.
  • GDF15 is then added in solution. If binding of GDF15 is detected, this indicates that the antibody does not inhibit formation of a GDF15-GFRAL complex.
  • QUEL-0101 , QUEL-0201 , QUEL-0301 and reference antibody mAb Q appear to inhibit binding of GDF15 to GFRAL, since a binding signal was observed following addition of GDF15 to the antibody-GFRAL complex.
  • the observation that none of QUEL-0101 , QUEL-0201 and QUEL-0301 compete with GDF15 for binding to GFRAL indicates that these antibodies inhibit GFRAL signalling via a different mechanism, such as inhibiting association of RET with GDF15-GFRAL. Formation of the GDF15-GFRAL-RET active signalling complex is thus inhibited.
  • Each of QUEL-0101 , QUEL-0201 and QUEL-0301 appear to bind a different epitope of GFRAL compared with mAb Q, since a binding signal was observed following addition of mAb Q to the antibody-GFRAL complex.
  • the tandem assay is an alternative to the sandwich assay described in (a) above.
  • it is the antigen rather than the antibody which is surface bound.
  • Fluman GFRAL with a 6His tag was captured on an anti-His antibody immobilised on the surface of a biosensor chip, and the first antibody was injected over the antigen followed by the second antibody.
  • QUEL- 0201 recognises a different epitope compared to QUEL-0101 and QUEL-0301 which share one epitope bin. None of the QUEL antibodies competed with mAb Q in this assay (confirming the results from the sandwich assay), which thus places mAb Q in a third epitope bin.
  • QUEL-0101 We isolated antibodies from immunised mice which have sequences that appear to share the same or parallel evolutionary lineage from germline as QUEL-0101 , on the basis of their high sequence homology with the QUEL-0101 VFI and VL domains. These are designated QUEL-0102, QUEL-0103, QUEL-0104 and QUEL-0105. Each of these clones displayed binding to human GFRAL.
  • GDF15 contributes to radiation-induced senescence through the ROS-mediated p16 pathway in human endothelial cells.
  • TGF-b Superfamily Cytokine MIC-1/GDF15 Is a Physiological Appetite and Body Weight Regulator’. Edited by Christopher Morrison. PLoS ONE 8 (2): e55174. doi 10.1371/journal. pone.0055174
  • Table A shows variable domain sequences of antibodies described in this specification. All QUEL VH domains, QUEL VL domains, QUEL CDRs, QUEL heavy chains and QUEL light chains, antibodies comprising them, as well as their encoding nucleic acids, represent embodiments of the present invention.
  • Table C shows sequences of antibody heavy and light chain constant regions and nucleic acids encoding them. These are examples f constant regions that may be used in antibodies of the present invention.
  • Human germline V, D and J gene segments generate VH domains of antibodies through recombination
  • human germline V and J geneegments generate the VL domains of antibodies through recombination.
  • Nucleotide and/or amino acid sequences of antibody variable domainsan be compared against human germline gene segments to identify the closest matching gene segments.
  • Table G1 shows the germline gene segments identified for the QUEL antibodies.
  • Table G2 shows variable domains with germline v and j framework regions corresponding to QUEL-0101 , QUEL-0201 and QUEL-0301.
  • Antibodies of the present invention optionally comprise framework regions with germline residues as shown here.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Immunology (AREA)
  • Organic Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Biophysics (AREA)
  • Biochemistry (AREA)
  • Genetics & Genomics (AREA)
  • Medicinal Chemistry (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Peptides Or Proteins (AREA)

Abstract

L'invention concerne des anticorps qui se lient à la protéine de type alpha du récepteur du facteur neurotrophique dérivé de cellules gliales (GFRAL) et qui inhibent l'activité de celle-ci. L'invention concerne également des procédés thérapeutiques utilisant des anticorps anti-GFRAL pour inhiber l'interaction GDF15-GFRAL et traiter des états tels que la cachexie cancéreuse.
PCT/EP2022/058556 2021-03-31 2022-03-31 Anticorps dirigés contre gfral WO2022207785A1 (fr)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
GBGB2104553.9A GB202104553D0 (en) 2021-03-31 2021-03-31 Antibodies to GFRAL
GB2104553.9 2021-03-31
GBGB2107330.9A GB202107330D0 (en) 2021-05-21 2021-05-21 Antibodies to GFRAL
GB2107330.9 2021-05-21

Publications (1)

Publication Number Publication Date
WO2022207785A1 true WO2022207785A1 (fr) 2022-10-06

Family

ID=81454648

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2022/058556 WO2022207785A1 (fr) 2021-03-31 2022-03-31 Anticorps dirigés contre gfral

Country Status (1)

Country Link
WO (1) WO2022207785A1 (fr)

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5869046A (en) 1995-04-14 1999-02-09 Genentech, Inc. Altered polypeptides with increased half-life
US6121022A (en) 1995-04-14 2000-09-19 Genentech, Inc. Altered polypeptides with increased half-life
WO2002060919A2 (fr) 2000-12-12 2002-08-08 Medimmune, Inc. Molecules a demi-vies longues, compositions et utilisations de celles-ci
US20120093818A1 (en) 2007-08-23 2012-04-19 Simon Mark Jackson Antigen binding proteins to proprotein convertase subtilisin kexin type 9 (pcsk9)
WO2017172260A1 (fr) * 2016-03-31 2017-10-05 Ngm Bioparmaceuticals, Inc. Protéines de liaison et leurs procédés d'utilisation

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5869046A (en) 1995-04-14 1999-02-09 Genentech, Inc. Altered polypeptides with increased half-life
US6121022A (en) 1995-04-14 2000-09-19 Genentech, Inc. Altered polypeptides with increased half-life
WO2002060919A2 (fr) 2000-12-12 2002-08-08 Medimmune, Inc. Molecules a demi-vies longues, compositions et utilisations de celles-ci
US20120093818A1 (en) 2007-08-23 2012-04-19 Simon Mark Jackson Antigen binding proteins to proprotein convertase subtilisin kexin type 9 (pcsk9)
WO2017172260A1 (fr) * 2016-03-31 2017-10-05 Ngm Bioparmaceuticals, Inc. Protéines de liaison et leurs procédés d'utilisation

Non-Patent Citations (50)

* Cited by examiner, † Cited by third party
Title
"NCBI", Database accession no. 995316.2
ABULIZI, PALIDANERUJA LOGANATHANDUO ZHAOTINA MELEYIXIN ZHANGTERRY ZWIEPKEXIANG LIUXIUFEN ZHENG: "Growth Differentiation Factor-15 Deficiency Augments Inflammatory Response and Exacerbates Septic Heart and Renal Injury Induced by Lipopolysaccharide", SCIENTIFIC REPORTS, vol. 7, no. 1, 2017, pages 1037, XP055845343, DOI: 10.1038/s41598-017-00902-5
ANKER, MARKUS S.RICHARD HOLCOMBMAURIZIO MUSCARITOLISTEPHAN VON HAEHLINGWILHELM HAVERKAMPAMINAH JATOIJOHN E. MORLEY ET AL.: "Orphan Disease Status of Cancer Cachexia in the USA and in the European Union: A Systematic Review", JOURNAL OF CACHEXIA, SARCOPENIA AND MUSCLE, vol. 10, no. 1, 2019, pages 22 - 34
BARACOS, VICKIE E.LISA MARTINMURRAY KORCDENIS C. GUTTRIDGEKENNETH C. H.FEARON: "Cancer-Associated Cachexia", NATURE REVIEWS DISEASE PRIMERS, vol. 4, no. 1, 2018, pages 1 - 18
BO, YACONG, MINGJIE YAO, LING ZHANG, WOLDE BEKALO, WEIQUAN LU, AND QUANJUN LU.: "Preoperative Nutritional Risk Index to Predict Postoperative Survival Time in Primary Liver Cancer Patients", ASIA PACIFIC JOURNAL OF CLINICAL NUTRITION, vol. 24, no. 4, 2015, pages 591 - 97
BORNER, TITOMYRTHA ARNOLDJOHAN RUUDSAMUEL N. BREITWOLFGANG LANGHANSTHOMAS A. LUTZANDERS BLOMQVISTTHOMAS RIEDIGER: "Anorexia-cachexia Syndrome in Hepatoma Tumour-bearing Rats Requires the Area Postrema but Not Vagal Afferents and Is Paralleled by Increased MIC-1/GDF15", JOURNAL OF CACHEXIA, SARCOPENIA AND MUSCLE, vol. 8, no. 3, 2017, pages 417 - 27
BOURGEOIS, BRIANNABO FANNEIL JOHANNSENMARIA CRISTINA GONZALEZBENNETT K. NGMARKUS J. SOMMERJOHN A. SHEPHERDSTEVEN B. HEYMSFIELD: "Improved Strength Prediction Combining Clinically Available Measures of Skeletal Muscle Mass and Quality", JOURNAL OF CACHEXIA, SARCOPENIA AND MUSCLE, vol. 10, no. 1, 2019, pages 84 - 94
BREIT, S.N.BROWN, D.A.TSAI, V.W: "The GDF15-GFRAL Pathway in Health and Metabolic Disease: Friend or Foe?", ANNU REV PHYSIOL, 2020
BREIT, SAMUEL N.JUAN J. CARREROVICKY WANG-WEI TSAINASREEN YAGOUTIFAMWEI LUOTAMARA KUFFNERASNE R. BAUSKIN ET AL.: "Macrophage Inhibitory Cytokine-1 (MIC-1/GDF15) and Mortality in End-Stage Renal Disease", NEPHROLOGY DIALYSIS TRANSPLANTATION, vol. 27, no. 1, 2012, pages 70 - 75
BROWN, DAVID A.JOHN MOOREHEIKO JOHNENTOM J. SMEETSASNE R. BAUSKINTAMARA KUFFNERHELEN WEEDON ET AL.: "Serum Macrophage Inhibitory Cytokine 1 in Rheumatoid Arthritis: A Potential Marker of Erosive Joint Destruction", ARTHRITIS & RHEUMATISM, vol. 56, no. 3, 2007, pages 753 - 64, XP055173045, DOI: 10.1002/art.22410
DORDO ET AL., J. MOL BIOL, vol. 217, 1999, pages 721 - 739
EMMERSON, PAUL JFENG WANGYONG DUQIAN LIURICHARD T PICKARDMALGORZATA D GONCIARZTAMER COSKUN ET AL.: "The Metabolic Effects of GDF15 Are Mediated by the Orphan Receptor GFRAL", NATURE MEDICINE, vol. 23, no. 10, 2017, pages 1215 - 19, XP055427499, DOI: 10.1038/nm.4393
FEARON, KENNETH, FLORIAN STRASSER, STEFAN D ANKER, INGVAR BOSAEUS, EDUARDO BRUERA, ROBIN L FAINSINGER, AMINAH JATOI, THE LANCET ONCOLOGY, vol. 12, no. 5, 2011, pages 489 - 95
FEJZO, MARLENA S., OLGA V. SAZONOVA, J. FAH SATHIRAPONGSASUTI, INGILEIF B. HALLGRFMSDOTTIR, VLADIMIR VACIC, KIMBER W. MACGIBBON, F: "Placenta and Appetite Genes GDF15 and IGFBP7 Are Associated with Hyperemesis Gravidarum", NATURE COMMUNICATIONS, vol. 9, no. 1, 2018
GOODSON, MEDICAL APPLICATIONS OF CONTROLLED RELEASE, vol. 2, 1984, pages 115 - 138
HESSELINK, MATTHIJS K. C.VERA SCHRAUWEN-HINDERLINGPATRICK SCHRAUWEN: "Skeletal Muscle Mitochondria as a Target to Prevent or Treat Type 2 Diabetes Mellitus", NATURE REVIEWS ENDOCRINOLOGY, vol. 12, no. 11, 2016, pages 633 - 45
HSIAO, EDWARD C.LEONIDAS G. KONIARISTERESA ZIMMERS-KONIARISSUZANNE M. SEBALDTHANH V. HUYNHSE-JIN LEE: "Characterization of Growth-Differentiation Factor 15, a Transforming Growth Factor β Superfamily Member Induced Following Liver Injury", MOLECULAR AND CELLULAR BIOLOGY, vol. 20, no. 10, 2000, pages 3742 - 3751, XP002525556, DOI: 10.1128/MCB.20.10.3742-3751.2000
HSU ET AL., ERRATUM. NATURE, vol. 551, 2017, pages 398
HSU, JER-YUANSUZANNE CRAWLEYMICHAEL CHENDINA AAYUPOVA, DARRIN ALINDHOUT, JARED HIGBEEALAN KUTACH ET AL.: "Non-Homeostatic Body Weight Regulation through a Brainstem-Restricted Receptor for GDF15", NATURE, vol. 550, no. 7675, 2017, pages 255 - 59
JOHNEN, HEIKOSHU LINTAMARA KUFFNERDAVID A BROWNVICKY WANG-WEI TSAIASNE R BAUSKINLIYUN WU ET AL.: "Tumor-Induced Anorexia and Weight Loss Are Mediated by the TGF-β Superfamily Cytokine MIC-1", NATURE MEDICINE, vol. 13, no. 11, 2007, pages 1333 - 40, XP002607045, DOI: 10.1038/NM1677
LANGER, SCIENCE, vol. 249, 1990, pages 1527 - 1533
LERNER, LORENAJULIE TAOQING LIURICHARD NICOLETTIBIN FENGBRIAN KRIEGERELIZABETH MAZSA ET AL.: "MAP3K11/GDF15 Axis Is a Critical Driver of Cancer Cachexia", JOURNAL OF CACHEXIA, SARCOPENIA AND MUSCLE, vol. 7, no. 4, 2016, pages 467 - 82, XP055870965
LERNER, LORENATERESA G HAYESNIANJUN TAOBRIAN KRIEGERBIN FENGZHENHUA WURICHARD NICOLETTIM ISABEL CHIUJENO GYURISJOSE M GARCIA: "Plasma Growth Differentiation Factor 15 Is Associated with Weight Loss and Mortality in Cancer Patients", JOURNAL OF CACHEXIA, SARCOPENIA AND MUSCLE, vol. 6, no. 4, 2015, pages 317 - 24
LI, ZHIHUABINGWEI WANGXUEFEI WUSHI-YUAN CHENGLUMINITA PARAOANJIAWEI ZHOU: "Identification, Expression and Functional Characterization of the GRAL Gene", JOURNAL OF NEUROCHEMISTRY, vol. 95, no. 2, 2005, pages 361 - 76, XP055426849, DOI: 10.1111/j.1471-4159.2005.03372.x
LLOYD, THE ART, SCIENCE AND TECHNOLOGY OF PHARMACEUTICAL COMPOUNDING, 1999
LOCKHART, S.M.SAUDEK, V.O'RAHILLY, S: "GDF15: A Hormone Conveying Somatic Distress to the Brain", ENDOCR REV, 2020, pages 41
MACIA, LAURENCEVICKY WANG-WEI TSAIAMY D. NGUYENHEIKO JOHNENTAMARA KUFFNERYAN-CHUAN SHISHU LIN ET AL.: "Macrophage Inhibitory Cytokine 1 (MIC-1/GDF15) Decreases Food Intake, Body Weight and Improves Glucose Tolerance in Mice on Normal & Obesogenic Diets", PLOS ONE, vol. 7, no. 4, 2012, pages e34868, XP002687296, DOI: 10.1371/JOURNAL.PONE.0034868
MORITAKE, TAKASHIHIDETOSHI FUJITAMITSURU YANAGISAWAMIYAKO NAKAWATARIKAORI IMADOMEETSUKO NAKAMURAMAYUMI IWAKAWATAKASHI IMAI: "Strain-Dependent Damage in Mouse Lung After Carbon Ion Irradiation", INTERNATIONAL JOURNAL OF RADIATION ONCOLOGY*BIOLOGY*PHYSICS, vol. 84, no. 1, 2012, pages e95 - 102
MULLICAN, SHANNON EXIEFAN LIN-SCHMIDTCHEN-NI CHINJOSE A CHAVEZJENNIFER L FURMANANTHONY A ARMSTRONGSTEPHEN C BECK ET AL.: "GFRAL Is the Receptor for GDF15 and the Ligand Promotes Weight Loss in Mice and Nonhuman Primates", NATURE MEDICINE, vol. 23, no. 10, 2017, pages 1150 - 57
NICOLINI, ANDREAPAOLA FERRARIMARIA CHIARA MASONIMILENA FINISTEFANIA PAGANIOTTAVIO GIAMPIETROANGELO CARPI: "Malnutrition, Anorexia and Cachexia in Cancer Patients: A Mini-Review on Pathogenesis and Treatment", BIOMEDICINE & PHARMACOTHERAPY, vol. 67, no. 8, 2013, pages 807 - 17
OKAZAKI, RYUJIYUSEOK MOONTOSHIYUKI NORIMURATHOMAS ELING: "Ionizing Radiation Enhances the Expression of the Nonsteroidal Anti-Inflammatory Drug-Activated Gene (NAG1) by Increasing the Expression of TP53 in Human Colon Cancer Cells", RADIATION RESEARCH, vol. 165, no. 2, 2006, pages 125 - 30
PARK, H.KIM, C.H.JEONG, J.H.PARK, M.KIM, K.S: "GDF15 contributes to radiation-induced senescence through the ROS-mediated p16 pathway in human endothelial cells", ONCOTARGET, vol. 7, 2016, pages 9634 - 9644
PAUSCH, THOMASWERNER HARTWIGULF HINZTHOMAS SWOLANABOGOTA D. BUNDYTHILO HACKERTLARS GRENACHERMARKUS W. BUCHLERJENS WERNER: "Cachexia but Not Obesity Worsens the Postoperative Outcome after Pancreatoduodenectomy in Pancreatic Cancer", SURGERY, TOPICS IN PANCREATIC SURGERY: A FESTSCHRIFT HONORING ANDREW L. WARSHAW, vol. 152, no. 3, 2012, pages 81 - 88
POWELL ET AL.: "Compendium of excipients for parenteral formulations", J PHARM SCI TECHNOL, vol. 52, 1998, pages 238 - 311, XP009119027
ROCHA, LLANNA MARQUES GOMES DAALINE MARCADENTIGALTIERI OTAVIO CUNHA DE MEDEIROSRICARDO ANDRADE BEZERRAJULIANA FLORINDA DE MENDONGA: "Is Cachexia Associated with Chemotherapy Toxicities in Gastrointestinal Cancer Patients? A Prospective Study", JOURNAL OF CACHEXIA, SARCOPENIA AND MUSCLE, vol. 10, no. 2, 2019, pages 445 - 54
RUPARELIA, NEILJOSHUA T. CHAIEDWARD A. FISHERROBIN P. CHOUDHURY: "Inflammatory Processes in Cardiovascular Disease: A Route to Targeted Therapies", NATURE REVIEWS CARDIOLOGY, vol. 14, no. 3, 2017, pages 133 - 44
S. FRENCHB. ROBSON, J. MOL. EVOL., vol. 19, 1983, pages 171
SENESSE, PIERREERIC ASSENATSTEPHANE SCHNEIDERCYRUS CHARGARINICOLAS MAGNEDAVID AZRIAXAVIER HEBUTERNE: "Nutritional Support during Oncologic Treatment of Patients with Gastrointestinal Cancer: Who Could Benefit?", CANCER TREATMENT REVIEWS, vol. 34, no. 6, 2008, pages 568 - 75, XP025346728, DOI: 10.1016/j.ctrv.2008.03.003
SHIMADA, K.MITCHISON, T.J: "Unsupervised identification of disease states from high-dimensional physiological and histopathological profiles", MOL SYST BIOL, vol. 15, 2019, pages e8636
STROHL, BIODRUGS, vol. 29, 2015, pages 215 - 239
SURIBEN, ROWENAMICHAEL CHENJARED HIGBEEJULIE OEFFINGERRICHARD VENTURABETTY LIKALYANI MONDAL ET AL.: "Antibody-Mediated Inhibition of GDF15-GFRAL Activity Reverses Cancer Cachexia in Mice", NATURE MEDICINE, vol. 26, no. 8, 2020, pages 1264 - 70, XP037215670, DOI: 10.1038/s41591-020-0945-x
TAYLOR ET AL., J. THEOR. BIOL., vol. 119, 1986, pages 205 - 218
TITO BORNEREVAN D. SHAULSONMISGANA Y. GHIDEWONBART C. DE JONGHE: "GDF15 Induces Anorexia through Nausea and Emesis", CELL METAB, 2020
TREAT ET AL.: "Liposomes in the Therapy of Infectious Disease and Cancer", 1989, LISS, pages: 353 - 365
TSAI, VICKY W. W.YASMIN HUSAINIRAKESH MANANDHARK. K. MICHELLE LEE-NGHONG PING ZHANGKATE HARRIOTTLELE JIANG ET AL.: "Anorexia/Cachexia of Chronic Diseases: A Role for the TGF-β Family Cytokine MIC-1/GDF15", JOURNAL OF CACHEXIA, SARCOPENIA AND MUSCLE, vol. 3, no. 4, 2012, pages 239 - 43, XP055105895, DOI: 10.1007/s13539-012-0082-6
TSAI, VICKY WANG-WEILAURENCE MACIAHEIKO JOHNENTAMARA KUFFNERRAKESH MANADHARSEBASTIAN BECK JORGENSENKA KI MICHELLE LEE-NG ET AL.: "TGF-b Superfamily Cytokine MIC-1/GDF15 Is a Physiological Appetite and Body Weight Regulator", PLOS ONE, vol. 8, no. 2, 2013, pages e55174, XP055491912, DOI: 10.1371/journal.pone.0055174
XIONG, YUMEI, KENNETH WALKER, XIAOSHAN MIN, CLARENCE HALE, THANHVIEN TRAN, RENEE KOMOROWSKI, JERRY YANG, SCIENCE TRANSLATIONAL MEDICINE, vol. 9, no. 412, 2017
YANG, LINDACHIH-CHUAN CHANGZHE SUNDENNIS MADSENHAISUN ZHUSOREN B PADKJAERXIAOAI WU ET AL.: "GFRAL Is the Receptor for GDF15 and Is Required for the Anti-Obesity Effects of the Ligand", NATURE MEDICINE, vol. 23, no. 10, 2017, pages 1158 - 66
ZHENG, H.WU, Y.GUO, T.LIU, F.XU, Y.CAI, S: "Hypoxia Induces Growth Differentiation Factor 15 to Promote the Metastasis of Colorectal Cancer via PERK-elF2alpha Signaling", BIOMED RES INT, 2020, pages 5958272
ZHOU, TINGBANGYAN WANGHUIQUAN LIUKAIXIANG YANGSUDIP THAPAHAOWEN ZHANGLU LISHIYING YU: "Development and Validation of a Clinically Applicable Score to Classify Cachexia Stages in Advanced Cancer Patients", JOURNAL OF CACHEXIA, SARCOPENIA AND MUSCLE, vol. 9, no. 2, 2018, pages 306 - 14

Similar Documents

Publication Publication Date Title
US20210101969A1 (en) High Affinity Human Antibodies to Human Angiopoietin-2
JP6654165B2 (ja) 抗tie2抗体およびその使用
JP6272970B2 (ja) ヒトtnf様リガンド1a(tl1a)に対するヒト抗体
JP5896993B2 (ja) ヒトgdf8に対する抗体
AU2015316522B2 (en) Anti-met antibodies and compositions
BR112020017676A2 (pt) Anticorpo antagonizante cd73
TW201402610A (zh) 抗-egfr抗體及其用途
CA2849508A1 (fr) Anticorps anti-erbb3 et leurs utilisations
BR112020023746A2 (pt) anticorpo, composição farmacêutica, método para tratar câncer, ácido nucleico isolado, vetor, célula hospedeira, processo para a produção de um anticorpo e reagente de diagnóstico
JP2022527373A (ja) Retを結合するヒト抗体およびその使用方法
JP2024016204A (ja) 抗trkbモノクローナル抗体および使用の方法
EP4142793A1 (fr) Anticorps spécifiques à abcb5 et leurs utilisations
US20240182585A1 (en) Therapeutic inhibitors of gdf15 signalling
WO2022207785A1 (fr) Anticorps dirigés contre gfral
CN117120477A (zh) 针对密蛋白18a2和cd3的双特异性抗体及其应用
WO2023143444A1 (fr) Anticorps anti-dkk1, composition pharmaceutique et utilisation associées
BR112014018651B1 (pt) Anticorpos anti-asic1, e composição farmacêutica

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: 22720329

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: 22720329

Country of ref document: EP

Kind code of ref document: A1