GB2524553A - Monoclonal antibodies to growth and differentiation factor 15 (GDF-15), and use thereof for treating cancer cachexia - Google Patents

Monoclonal antibodies to growth and differentiation factor 15 (GDF-15), and use thereof for treating cancer cachexia Download PDF

Info

Publication number
GB2524553A
GB2524553A GB1405477.9A GB201405477A GB2524553A GB 2524553 A GB2524553 A GB 2524553A GB 201405477 A GB201405477 A GB 201405477A GB 2524553 A GB2524553 A GB 2524553A
Authority
GB
United Kingdom
Prior art keywords
antibody
amino acid
antigen
acid sequence
ser
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Granted
Application number
GB1405477.9A
Other versions
GB2524553C (en
GB201405477D0 (en
GB2524553B (en
Inventor
Jorg Wischhusen
Markus Junker
Tina Schafer
Dirk Puhringer
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Julius Maximilians Universitaet Wuerzburg
Original Assignee
Julius Maximilians Universitaet Wuerzburg
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Julius Maximilians Universitaet Wuerzburg filed Critical Julius Maximilians Universitaet Wuerzburg
Priority to GB1405477.9A priority Critical patent/GB2524553C/en
Publication of GB201405477D0 publication Critical patent/GB201405477D0/en
Priority to ES15712629T priority patent/ES2763859T3/en
Priority to AU2015238264A priority patent/AU2015238264B2/en
Priority to PL15712629T priority patent/PL3122775T3/en
Priority to PT192053213T priority patent/PT3653644T/en
Priority to ES19205321T priority patent/ES2965626T3/en
Priority to NZ724649A priority patent/NZ724649A/en
Priority to RS20191652A priority patent/RS59700B1/en
Priority to CN201580027932.8A priority patent/CN106536553B/en
Priority to PL19205321.3T priority patent/PL3653644T3/en
Priority to DK15712629.3T priority patent/DK3122775T3/en
Priority to HUE15712629A priority patent/HUE047398T2/en
Priority to DK19205321.3T priority patent/DK3653644T3/en
Priority to BR122023027559-4A priority patent/BR122023027559A2/en
Priority to US15/128,604 priority patent/US20170204174A1/en
Priority to EP19205321.3A priority patent/EP3653644B1/en
Priority to FIEP19205321.3T priority patent/FI3653644T3/en
Priority to HUE19205321A priority patent/HUE064573T2/en
Priority to HRP20231514TT priority patent/HRP20231514T1/en
Priority to JP2016559286A priority patent/JP6865585B2/en
Priority to LTEP19205321.3T priority patent/LT3653644T/en
Priority to PCT/EP2015/056654 priority patent/WO2015144855A1/en
Priority to BR112016022013A priority patent/BR112016022013A2/en
Priority to EP15712629.3A priority patent/EP3122775B1/en
Priority to PT157126293T priority patent/PT3122775T/en
Priority to KR1020167029710A priority patent/KR102520978B1/en
Priority to LTEP15712629.3T priority patent/LT3122775T/en
Priority to CA2943694A priority patent/CA2943694A1/en
Priority to SI201531059T priority patent/SI3122775T1/en
Priority to EP23203704.4A priority patent/EP4316596A3/en
Publication of GB2524553A publication Critical patent/GB2524553A/en
Priority to IL248038A priority patent/IL248038B/en
Application granted granted Critical
Publication of GB2524553B publication Critical patent/GB2524553B/en
Publication of GB2524553C publication Critical patent/GB2524553C/en
Priority to US15/912,358 priority patent/US10604565B2/en
Priority to CY20201100008T priority patent/CY1122456T1/en
Priority to HRP20200034TT priority patent/HRP20200034T1/en
Priority to US16/794,401 priority patent/US11634482B2/en
Priority to US16/794,406 priority patent/US11760795B2/en
Priority to IL276405A priority patent/IL276405B/en
Priority to AU2020260440A priority patent/AU2020260440B2/en
Priority to JP2021064572A priority patent/JP2021119138A/en
Priority to JP2023062338A priority patent/JP2023098966A/en
Priority to US18/364,173 priority patent/US20230382987A1/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

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
    • C07K16/22Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against growth factors ; against growth regulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/39533Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals
    • A61K39/3955Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals against proteinaceous materials, e.g. enzymes, hormones, lymphokines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/24Immunoglobulins specific features characterized by taxonomic origin containing regions, domains or residues from different species, e.g. chimeric, humanized or veneered
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/30Immunoglobulins 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
    • C07K2317/73Inducing cell death, e.g. apoptosis, necrosis or inhibition of cell proliferation
    • 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

Abstract

A monoclonal antibody capable of binding to human GDF-15 (MIC-1/NAG-1), or an antigen-binding portion thereof, comprises a heavy chain variable domain CDR3 region at least 90% identical to the sequence ARSSYGAMDY, and a light chain variable domain CDR3 region at least 85% identical to the sequence QQYNNFPYT, may be used for treating cancer cachexia in a mammal. The antibody may bind to a conformational or discontinuous epitope on human GDF-15, comprising residues 40-55 and 94-114. The antibodies of the invention include murine antibodies, chimeric antibodies and humanized antibodies, and preferably treat the cancer itself as well as cancer cachexia.

Description

Monoclonal Antibodies to Growth and Differentiation Factor 15 (GDF-l5), and uses thereof for treating cancer cachexia
FIELD OF THE INVENTION
The present invention relates to monoclonal ant±-human-GDF-l5 antibodies, pharmaceutical compositions, kits, methods and uses and the cell lines capable of producing the monoclonal antibodies described herein. The present invention further relates to antibodies to human GDF-15 capable of inhibiting cancer growth, treating cancer-induced weight loss and cancer cachexia.
BACKGROUND
To date, many cancers are still areas of unmet medical needs, and accordingly, means to more effectively treat cancer, and to treat cancer in a broader range of cancers are needed.
In addition to the suffering caused by the cancer itself, many patients suffer from cancer cachexia, a medical condition triggered by the cancer that typically involves weight loss and loss of skeletal muscle mass. Cancer cachexia accounts f or more than 20 percent of all cancer-related deaths (Murphy KT and Lynch GB: Update on emerging drugs for cancer cachexia. Expert Opin Emerg Drugs. 2009 Dec;14(4):6l9-32.).
Thus, in order to improve the treatment and prognosis of cancers which lead to cancer cachexia, treatment regimens that target both of these medical conditions are needed. To date, most of the emerging drugs for treatments of cancer cachexia are drugs that target cachexia but not the cancer itself (see Murphy KT and Lynch GB: Update on emerging drugs for cancer cachexia. Expert Opin Emerg Drugs. 2009 Dec;l4(4):619.-32.). Only very few drugs are effective against both the cancer and cancer cachexia, and therefore, complex treatment regimens that combine anti-cancer drugs and anti-cancer cachexia drugs are oftentimes needed. Accordingly, there is still an unmet medical need for drugs that can be used to effectively treat both cancer and cancer cachexia in a broad range of cancers.
Many types of cancer are known to express growth factors, including factors such as VEGF, PDGF, TGF-and GDF-l5.
GDF-lS, growth and differentiation factor-is, is a divergent member of the TGF-J3 superfamily. It is a protein which is intracellularly expressed as a precursor, subsequently processed and eventually becomes secreted from the cell into the environment. Both the active, fully processed (mature) form and the precursor of GDP-15 can be found outside cells.
The precursor covalently binds via its COON-terminal amino acid sequence to the extraceliular matrix (Bauskin ?R et a!., Cancer Research 2005) and thus resides on the exterior of a cell. The active, fully processed (mature) form of GDF-l5 is soluble and is found in blood sera. Thus, the processed form of GDF-i5 may potentially act on any target cell within the body that is connected to the blood circulation, provided that the potential target cell expresses a receptor for the soluble GDF-15 ligand.
During pregnancy, GDF-15 is found under physiological conditions in the placenta. However, many malignant cancers (especially aggressive brain cancers, melanoma, lung cancer, gastrointestinal tumors, colon cancer, pancreatic cancer, prostate cancer and breast cancer (Mimeault N and Batra SR.
J. Cell Physiol 2010)) exhibit increased GDF-15 levels in the tumor as well as in blood serum. Likewise, correlations have been described between high GDF-l5 expression and chemoresistance (Huang CY et al.., din. Cancer Res. 2009) and between high GDF-15 expression and poor prognosis, respectively (Brown DA et al., din. Cancer Res. 2009).
GDF-l5 is expressed in gliomas of different WHO grades as assessed by immunohistochemistry (Roth et al., Cliii. Cancer Res. 2010). Further, Roth et al. stably expressed short hairpin RNA-expressing DNA constructs targeting endogenous GDF-15 or control constructs in SMAGGO glioma cells. When using these pre-established stable cell lines, they observed that tumor formation in mice bearing GDF-l5 knockdown SMAS6O cells was delayed compared to mice bearing control constructs.
Patent application PCT/EP2013/070127 relates to monoclonal anti-GDF-l5 antibodies, in particular to an antibody produced by the hybridoma cell line B1-23 deposited with the Deutsche Sammlung für Mikroorganismen tmd Zellkultureri GmbH (DSMZ) under the accession No. OSM ACC3142 under the Budapest treaty. PCT/EP20l3/070127 also relates to other uses of the anti-GDF-l5 antibodies.
Patent applications WO 2005/099746 and WO 2009/021293 relate to an anti-human-GDF-15 antibody (Mab2G) capable of antagonizing effects of human GDF-lS on tumor-induced weight loss in viva in mice; In these documents, immunologically compromised mice were administered with human tumor cells (prostate carcinoma cells DU145) transfected with plasmids overexpressing human GDF-l5. Tumor cells carrying plasmids lacking a GDF-l5 sequence served as a negative control. Those mice expressing xenograft GOP-is exhibited a tumor-induced weight loss (clinical term: cachexia) and anorexia. A single untraperitoneal administration of 1 mg of Mab2G from NO 2005/099746 resulted in a complete reversal of tumor-induced weight loss. WO 2005/099746 and WO 2009/021293 do not disclose effects of an anti-human-GOF-iS antibody on tumor growth.. Moreover, these documents are silent as to whether anti-human-GDF-l5 antibodies could lead to an increase in body weight of the treated mice compared to their body weight before the onset of cachexia.
Similarly, Johnen H et al. (Nature Medicine, 2007) reported effects of an anti-human-GDF-15 monoclonal antibody on cancer-induced anorexia and weight loss but did not observe any effects of the anti-human-GDF-l5 antibody on the size of the tumor formed by the cancer, even when the antibody was administered at a high dosage of 1 mg, and thus the antibody did not inhibit growth of the cancer.
Accordingly, to date, there was still a need in the art for means to effectively treat cancer cachexia and additionally treat cancer, and for means to treat cancer cachexia and additionally cancer in a broader range of cancers.
It is therefore an object of the invention to obtain means that can be used to effectively treat cancer cachexia, and to also effectively treat cancer, and means that can be used to treat cancer cachexia, and to also effectively treat cancer in a broader range of cancers.
In an effort to find means to achieve these objects, the present inventors have surprisingly found that a monoclonal antibody to human CDF-15 can be used to treat cancer cachexia and to also treat cancer of human xenograft tumors in mice.
Additionally, an antibody to human GDF-15 in accordance with the present invention has an equilibrium dissociation constant of about 790 pM for recombinant GDF-15 even without additional affinity maturation, which is a higher affinity compared to most known therapeutic antibodies.
Thus, the antibody to human GDF-l5 in accordance with the present invention has superior properties compared to antibodies known from the art, and is particularly useful for inhibiting cancer cachexia, and additionally useful for treating cancer. Accordingly, the present invention was completed.
BRIEF DESCRIPTION OF THE INVENTION
The present invention solves the above-mentioned objects by using the monoclonal antibodies, pharmaceutical compositions, kits, uses and the cell lines capable of producing the monoclonal antibodies described herein.
In particular, the present inventors surprisingly show that monoclonal antibodies to human GDF-l5 and antigen binding portions thereof according to the invention are capable of inhibiting cancer cachexia and/or cancer growth. This was unexpected because those monoclonal antibodies to GDF-15 that were previously known from the art (WO 2005/099746, WO 2009/021293 and Jobnen H et al., Nature Medicine, 2007) were only known to cause a reversal of cancer-induced weight loss (i.e. a reversal of a secondary symptom induced by the GDF-lS expressed by the cancer), but were shown to fail at inhibiting growth of the cancer.
By showing that the monoclonal antibodies to human GDF-lS according to the invention can be used to treat cancer-induced weight loss and/or cancer cachexia and treat cancer, the present inventors also surprisingly show that human GDF-protein can be targeted by the antibodies of the invention in a way that both cancer growth is inhibited and cancer-induced weight loss and cancer cachexia is treated. It is expected that the same mechanisms of cancer growth inhibition and treatment of cancer-induced weight loss and cancer cachexia are applicable to a large number of cancers that overexpress human GDF-lS including the cancers listed below.
Thus, the present invention relates to a monoclonal antibody capable of binding to human GDF-15, or an antigen-binding portion thereof, wherein the heavy chain variable domain comprises a CR3 region comprising the amino acid sequence of SEQ ID NO: S or an amino acid sequence at least 90% identical thereto, and wherein the light chain variable domain comprises a CDR3 region comprising the amino acid sequence of SEQ ID NO: 7 or an amino acid sequence at least 85% identical thereto, for use in a method for treating cancer cachexia in a mammal. The method comprises administering the antibody or antigen-binding portion thereof to said mammal. Additionally, the present invention relates to a corresponding method for treatment.
Further, the invention also relates to a monoclorial antibody capable of binding to human GDF-lS, or an antigen-binding portion thereof, wherein the binding is binding to a conformational or discontinuous epitope on human GDF-lS comprised by the amino acid sequences of SEQ ID No: 25 and SEQ ID No: 26, for use in a method for treating cancer cachexia in a mammal. The method comprises administering the antibody or antigen-binding portion thereof to said mammal.
Additionally, the present invention relates to a corresponding method for treatment.
The invention also relates to a pharmaceutical composition comprising the antibody or antigen-binding portion thereof according to the invention.
The invention also relates to an antibody or antigen-binding portion thereof according to the invention for use in medicine.
Additionally, the invention relates to a kit comprising the pharmaceutical composition according to the invention.
The invention also relates to an expression vector comprising a nucleotide sequence encoding the antibody or antigen-binding portion thereof according to the invention.
Further, the invention relates to a cell line capable of producing an antibody or antigen-binding portion thereof according to the invention.
Thus, by providing monoclonal antibodies to human GOF-iS, the present invention provides means for the treatment of cancer cachexia and a cancer growth inhibitor that meets the above-defined needs in the art.
BRIEF DESCRIPTION OF THE DRAWINGS
Figure 1: NKG2D Expression on NK Cells after Treatment with or without GDF-15. The cell surface expression of NKG2D was determined on NK cells after treatment with the indicated cytokines in the presence or absence of the anti-GDF-l5 antibody utAh Bl-23. The figure displays specific fluorescence intensities determined by flow cytometry, quantified relative to an unspecific control antibody.
Figure 2: Akt Phosphorylation in the Ovarian Carcinoma Cell Line SIC-OV-3. In order to quantify the Western Blot for the ovarian carcinoma cell line SK-OV-3, the ratio of phosphorylated Akt to the total amount of Akt was calculated and normalized to the untreated control.
Figure 3: JNK1/2 Phosphorylation in Immune Cells. In order to quantify the Western Blot, the ratio of phosphorylated 1JNK1/2 to the total amount of JNI< was calculated and normalized to the untreated control.
Figure 4: An anti-tumor effect of murine Bl-23 in vivo. Balb/c/nu nude mice were used in a xenograft setting with the melanoma cell line tJACC-257. The tumor size of the animal cohort treated with El-23 (open squares) was significantly decreased, compared to the PBS control group (filled solid a circles). Significance was defined as p.cO.O5 as assessed by WilcoxonTh log-rank test.
Figure 5: Treatment of cancer cachexia with anti-GDF-lS antibodies. The figure shows a comparison of the mean body weight of all treated Balb/cnu/nU nude mice, which were inoculated with UACC-257 cells. The changes of the body weight are depicted in percent as compared to the starting body weight on day 0, for a period of 38 days.
Figure 6: Coomassie stain of antibodies used in the study No. 140123.
DETAILED DESCRIPTION OF THE INVENTION
Definitions Unless otherwise defined below, the terms used in the present invention shall be understood in accordance with their common meaning known to the person skilled in the art.
The term "antibody" as used herein refers to any functional antibody that is capable of specific binding to the antigen of interest, as generally outlined in chapter 7 of Paul, W.E.
(Ed.).: Fundamental Immunology 2nd Ed. Raven Press, Ltd., New York 1989, which is incorporated herein by ref erence. Without particular limitation, the term "antibody" encompasses antibodies from any appropriate source species, including chicken and mammalian such as mouse, goat, non-human primate and human. Preferably, the antibody is a humanized antibody.
The antibody is preferably a monoclonal antibody which can be prepared by methods well-known in the art. The term "antibody" encompasses an IgG-l, -2, -3, or -4, IgE, IgA, 1gM, or IgD isotype antibody. The term "antibody" encompasses monomeric antibodies (such as IgD, 1gB, IgG) or oligomeric antibodies (such as IgA or 1gM). The term "antibody" also encompasses -without particular limitations -isolated antibodies and modified antibodies such as genetically engineered antibodies, e.g. chimeric antibodies.
The nomenclature of the domains of antibodies follows the terms as known in the art. Each monomer of an antibody comprises two heavy chains and two light chains, as generally known in the art. Of these, each heavy and light chain comprises a variable domain (termed V11 for the heavy chain and VL for the light chain) which is important for antigen binding. These heavy and light chain variable domains comprise (in an N-terminal to C-terminal order) the regions FR1, CR1, FR2, CDR2, FR3, CDR3, and FR4 (FR, framework region; CDR, complementarity determining region which is also known as hypervariable region). The identification and assignment of the above-mentioned antibody regions within the antibody sequence is generally in accordance with Kabat et al. (Sequences of proteins of immunological interest, U.S. Dept. of Health and Human Services, Public Health Service, NatiOnal Institutes of Health, Bethesda, Md. 1983), or Chothia et al. (Conformations of immunoglobulin hypervariable regions. Nature. 1989 Dec 21-28;342(6252):877-83.), or may be performed by using the IMGT/V-QUEST software described in Giudicelli et al. (IMGT/V-QuEST, an integrated software program for immunoglobulin and P cell receptor V-J and V-D-J rearrangement analysis. Nucleic Acids lIes. 2004 Jul l;32(Web Server issue):W435-40.), which is incorporated herein by reference. Preferably, the antibody regions indicated above are identified and assigned by using the IMGT/V-QUEST software.
A "monoclonal antibody" is an antibody from an essentially homogenous population of antibodies, wherein the antibodies are substantially identical in sequence (i.e. identical except for minor fraction of antibodies containing naturally occurring sequence modifications such as amino acid modifications at their N-and C-termini). Unlike polyclonal antibodies which contain a mixture of different antibodies directed to numerous epitopes, monoclonal antibodies are directed to the same epitope and are therefore highly specific. The term "monoclonal antibody" includes (but is not limited to) antibodies which are obtained from a monoclonal cell population derived from a single cell clone, as for instance the antibodies generated by the hybridoma method described in Icôhler and Milstein (Nature, 1975 Aug 7;256(5517):495-7) or Harlow and Lane ("Antibodies: A Laboratory Manual" Cold Spring Harbor Laboratory Press, Cold Spring Harbor, New York 1988). A monoclonal antibody may also be obtained from other suitable methods, including phage display techniques such as those described in Clackson et al. (Nature. 1991 Aug l5;352(6336):624-8) orMarks et al. (J Md Biol. 1991 Dec 5;222(3) :581-97). A monoclonal antibody may be an antibody that has been optimized for antigen-binding properties such as decreased Kd values, optimized association and dissociation kinetics by methods known in the art. For instance, lCd values may be optimized by display methods including phage display, resulting in affinity-matured monoclonal antibodies. The term "monoclonal antibody" is not limited to antibody sequences from particular species of origin or from one single species of origin. Thus, the meaning of the term "monoclonal antibody" encompasses chimeric monoclonal antibodies such as humanized monoclonal antibodies.
"Humanized antibodies" are antibodies which contain human sequences and a minor portion of non-human sequences which confer binding specificity to an antigen of interest (e.g. human GDF-15). Typically, humanized antibodies are generated by replacing hypervariable region sequences from a human acceptor antibody by hypervariable region sequences from a non-human donor antibody (e.g. a mouse, rabbit, rat donor antibody) that binds to an antigen of interest (e.g. human GDF-15) . In some cases, framework region sequences of the acceptor antibody may also be replaced by the corresponding sequences of the donor antibody. In addition to the sequences derived from the donor and acceptor antibodies, a "humanized antibody" may either contain other (additional or substitute) residues or sequences or not, Such other residues or sequences may serve to further improve antibody properties such as binding properties (e.g. to decrease lcd values) and/or immunogenic properties (e.g. to decrease antigenicity in humans). Non-limiting examples for methods to generate humanized antibodies are known in the art, e.g. from Riecbmann et al. (Nature. 1988 Mar 24;332(6162):323-7) or Jones et al. (Nature. 1986 May 29-Jun 4;321(6069) :522-5) The term "human antibody" relates to an antibody containing human variable and constant domain sequences. This definition encompasses antibodies having human sequences bearing single amino acid substitutions or modifications which may serve to further improve antibody properties such as binding properties (e.g. to decrease lcd values) and/or immunogenic properties (e.g. to decrease antigenicity in humans). The term "human antibody" excludes humanized antibodies where a portion of non-human sequences confers binding specificity to an antigen of interest.
An "antigen-binding portion" of an antibody as used herein refers to a portion of an antibody that retains the capability of the antibody to specifically bind to the antigen (e.g. GDF-l5), i.e. the "antigen-binding portion" is capable of competing with the antibody for specific binding to the antigen. The "antigen-binding portion" may contain one or more fragments of the antibody. Without particular limitation, it can be produced by any suitable method known in the art, including recombinant DNA methods and preparation by chemical or enzymatic fragmentation of antibodies.
Antigen-binding portions may be Fab fragments, F(ab' fragments, F(ab' )2 fragments, single chain antibodies (scFv), single-domain antibodies, diabodies or any other portion(s) of the antibody that allow(s) to retain binding to the antigen.
An "antibody" (e.g. a monoclonal antibody) or an "antigen-binding portion" may have been derivatized or be linked to a different molecule. For example, molecules that may be linked to the antibody are other proteins (e.g. other antibodies), a molecular label (e.g. a fluorescent, luminescent, colored or radioactive molecule), a pharmaceutical and/or a toxic agent.
The antibody or antigen-binding portion may be linked directly (e.g. in form of a fusion between two proteins), or via a linker molecule (e.g. any suitable type of chemical linker known in the art).
As used herein, the terms "binding" or "bind" refer to specific binding to the antigen of interest (e.g. human GDF- 15). Preferably, the lcd value is less than 100 nN. more preferably less than 50 nM, still more preferably less than nN, still more preferably less than S nM and most preferably less than 2 nIl.
The term "epitope" as used herein refers to a small portion of an antigen that forms the binding site for an antibody.
In the context of the present invention, binding or competitive binding of antibodies or their antigen-binding portions to the antigen of interest (e.g. human GDF-15) is measured by using surface plasmon resonance measurements as a reference standard assay, as described below.
The terms "Ku" or "Kr value" relate to the equilibrium dissociation constant as known in the art. In the context of the present invention, these tens relate to the equilibrium dissociation constant of an antibody with respect to a particular antigen of interest (e.g. human GDF-15) The equilibrium dissociation constant is a measure of the propensity of a complex (e.g. an antigen-antibody complex) to reversibly dissociate into its components (e.g. the antigen and the antibody). For the antibodies according to the invention, lCp values (such as those for the antigen human GDF-1S) are generally determined by using surface plasmon resonance measurements as described below.
The term "cancer growth" as used herein relates to any measureable growth of the cancer. For cancers forming solid tumors, "cancer growth" relates to a measurable increase in tumor volume over time. If the cancer has formed only a single tumor, "cancer growth" relates only to the increase in volume of the single tumor. If the cancer has formed multiple tumors such as metastases, "cancer growth" relates to the increase in volume of all measurable tumors. For solid tumors, the tumor volume can be measured by any method known in the art, including magnetic resonance imaging and computed tomography (CT scan).
For leukemias which are characterized by the presence of cancerous cells of the blood system in blood, "cancer growth" relates to a measurable increase in the number of cancer cells per blood volume. In order to carry out such measurements, cancer cells can be identified from blood samples by using any method known in the art, including cell morphology measurements, or staining of tumor cell marker proteins such as tumor marker cell surface proteins, e.g. by staining with specific antibodies, and the cancer cells can be counted.
Terms such as "inhibiting cancer growth" as used herein refer to a measurable inhibition of cancer growth in patient treated with the antibody. Preferably, the inhibition is statistically significant. Inhibition of cancer growth may be assessed by comparing cancer growth in a group of patients treated in accordance with the present invention to a control group of untreated patients, or by comparing a group of patients that receive a standard cancer treatment of the art plus a treatment according to the invention with a control group of patients that only receive a standard cancer treatment of the art. Such studies for assessing the inhibition of cancer growth are designed in accordance with accepted standards for clinical studies, e.g. double-blinded, randomized studies with sufficient statistical power. The term "inhibiting cancer growth" includes an inhibition of cancer growth where the cancer growth is inhibited partially (i.e. where the cancer growth in the patient is delayed compared to the control group of patients), an inhibition *where the cancer growth is inhibited completely (i.e. where the cancer growth in the patient is stopped), and an inhibition where cancer growth is reversed (i.e. the cancer shrinks).
An "isolated antibody" as used herein is an antibody that has been identified and separated from the majority of components (by weight) of its source environment, e.g. from the components of a hybridoma cell culture or a different cell culture that was used for its production (e.g. producer cells such as CEO cells that recombinantly express the antibody).
The separation is performed such that it sufficiently removes components that may otherwise interfere with the suitability of the antibody for the desired applications (e.g. with a therapeutic use of the anti-human GDF-l5 antibody according to the invention). Methods for preparing isolated antibodies are known in the art and include Protein A chromatography, anion exchange chromatography, cation exchange chromatography, virus retentive filtration arid ultrafiltration. Preferably, the isolated antibody preparation is at least 70 % pure (w/w), more preferably at least 80 % pure (w/w), still more preferably at least 90 % pure (w/w), still more preferably at least 95 % pure (w/w), and most preferably at least 99 % pure (w/w), as measured by using the Lowry protein assay.
A "diabody" as used herein is a small bivalent antigen-binding antibody portion which comprises a heavy chain variable domain linked to a light chain variable domain on the same polypeptide chain linked by a peptide linker that is too short to allow pairing between the two domains on the same chain. This results in pairing with the complementary domains of another chain and in the assembly of a dimeric molecule with two antigen binding sites. Diabodies may be bivalent and monospecific (such as diabodies with two antigen binding sites for human GDF-l5), or may be bivalent and bispecific (e.g. diabodies with two antigen binding sites, one being a binding site for human GDF-lS, and the other one being a binding site for a different antigen). A detailed description of diabodies can be found in Fiolliger P et al. ("Diabodies": small bivalent and bispecific antibody fragments." Proc Nati Acad Sci U S A. 1993 Jul 15;90(14) :6444-8.).
A "single-domain antibody" (which is also referred to as "NanobodyTM") as used herein is an antibody fragment consisting of a single monomeric variable antibody domain.
Structures of and methods for producing single-domain antibodies are known from the art, e.g. from Holt U et al. ("Domain antibodies: proteins for therapy." Trends Biotechriol. 2003 Nov;21(11):484-90.), Saerens D et al. ("Single-domain antibodies as building blocks for novel therapeutics." Curr Opin Pharrnacol. 2008 Oct;8(5) :600-8. Epub 2008 Aug 22.), and Arbabi Ghahroudi M et al. ("Selection and identification of single domain antibody fragments from camel heavy-chain antibodies." FEBS Lett. 1997 Sep 15;414(3):52l-6.).
The term "higher" as used herein means that a value (e.g. a GDF-l5 level) in a patient sample is higher than a value in a corresponding control sample or group of control samples.
Preferably, the difference is statistically significant.
The term "elevated GDF-15 levels" as used herein means that the human patient has higher GDF-15 levels in blood serum before administration of the antibody or antigen-binding portion thereof or the pharmaceutical composition according to the invention, when compared to median GDF-l5 levels in blood sera of healthy human control individuals as a reference.
A preferred median reference for 01W-iS level in blood sera of healthy human control individuals is c 0.8 ng/ml. The expected range is between 0.2 ng/ml and 1.2 ng/ml in healthy human controls (Reference: Tanno T et al.: "Growth differentiation factor 15 in erythroid health and disease." Curr Opin Hematol. 2010 May; 17(3) 184-190.).
Preferably, the levels are 1.2-fold higher, more preferably 1.5-fold higher, still more preferably 2-fold higher and most preferably S-fold higher.
The term "prior to administration" as used herein means the period of time immediately before administration of the antibody, fragment thereof or the pharmaceutical composition according to the invention. Preferably, the term "prior to administration" means a period of 30 days immediately before administration; most preferably a period of one week immediately before administration.
The terms "significant", "significantly", etc. as used herein refer to a statistically significant difference between values.
The terms "cancer" and "cancer cell" is used herein in accordance with their common meaning in the art (see for instance Weinberg.R. et al.: The Biology of Cancer. Garland Science: New York 2006. 850p.).
The term "cancer-induced weight loss" is used herein in accordance with its common meaning in the art. Cancer-induced weight loss is frequently seen as an adverse effect in individuals having cancer (see, for instance Pearon K. èt al.: Definition and classification of cancer cachexia: an international consensus. Lancet Oncol. 2011 May; 12(5) :489- 95.; Tisdale MJ.: Mechanisms of cancer cachexia. Physiol Rev.
2009 Apr;89(2h381-410.). The term "cancer-induced weight loss" relates to the body weight loss induced by the cancer.
Additional body weight loss in addition to the cancer-induced weight loss -e.g. body weight loss induced by cancer treatments such as surgery, chemotherapy and radiotherapy -can also occur in individuals having cancer. It is understood that the meaning of the term "cancer-induced weight loss" does not include this additional body weight loss. However, this does not exclude the possibility that the antibodies of the present invention -in addition to their effects on cancer-induced weight loss and on cancer growth -may have beneficial effects against such additional body weight loss, e.g. by reverting or partly reverting such additional weight loss, or by preventing orpartly preventing such additional body weight loss.
Body weight can easily be measured by weighing, and body weight is typically expressed in units of mass such as kg.
The term "cancer cachexia" is used herein in accordance with its common meaning in the art (see, for instance Fearon K. et al.; Definition and classification of cancer cachexia: an international consensus. Lancet Oncol. 2011 May; 12(5):489- 95.; Tisdale MJ.: Mechanisms of cancer cachexia. Physiol Rev.
2009 Apr;89(2h381-410.). The most common symptom of cancer cachexia is cancer-induced weight loss. Thus, according to one definition, cancer cachexia is characterized by an ongoing loss of skeletal muscle mass (with or without loss of fat mass) that cannot be fully reversed by conventional nutrition. In human patients, cancer cachexia can be defined by a weight loss of more than 5% during the past months, or by a body mass index of less than 20 g/m2 and any degree of ongoing weight loss that is higher than 21, or by sarcopenia (i.e. degenerative loss of muscle mass) and an ongoing weight loss that is higher than 2% (see, Fearon K. et al.: Definition and classification of cancer cachexia; an international consensus. Lancet Oncol. 2011 May; 12(5):489- 95.). A further symptom of cancer cachexia can be a depletion of adipose tissue.
With respect to cancer cachexia, a "treatment" according to the present invention may be a treatment for preventing and/or a treatment for inhibiting or reverting cancer cachexia. Typically, a treatment for preventing cancer cachexia is a treatment that is given prophylactically at a stage of the cancer disease where no cancer cachexia has yet occurred. A treatment for inhibiting cancer cachexia is typically a treatment that is given at a stage of the cancer disease where some cancer cachexia has occurred, in order to inhibit a further progression of the cancer cachexia. A treatment for reverting and cancer cachexia is typically a treatment that is started at a stage of the cancer where some cancer cachexia has occurred, and which reverts the cancer cachexia. The effect of the treatment can be a partial effect, i.e. a partial prevention, a partial inhibition or a partial reversion of cancer cachexia, or a complete effect, i.e. a complete prevention, a complete inhibition or a complete reversion of cancer cachexia. Preferably, according to the present invention, the effect of the treatment is a complete prevention, a complete inhibition or a complete reversion of cancer cachexia. More preferably, the effect of the treatment according to the present invention is a complete prevention or a complete reversion of cancer cachexia.
As used herein, the term "complete Cly)" in connection with a treatment of cancer cachexia according to the invention means that in case of a treatment for preventing, no cancer cachexia occurs in the treated individual during and/or following the treatment. In case of a treatment for inhibiting cancer cachexia, the term "complete(ly)" means that no further progression of the cancer cachexia occurs in the treated individual during and/or following the treatment.
In case of a treatment for reverting cancer cachexia, the term "complete(ly)" means that during or following the treatment, the cancer cachexia is completely reverted such that no cancer cachexia is present in. the treated individual.
With respect to these effects of the treatment according to the invention, the term "no cancer cachexia" means that by using standard methods for measurements and for diagnosis known in the art, no cancer cachexia is detectable. Likewise, the term "no further progression of the cancer cachexia" means that by using standard methods for measurements and for diagnosis known in the art, no further progression of cancer cachexia is detectable. The methods known in the art and referred to herein are for instance described in Fearon ICC.: Cancer cachexia: developing multimodal therapy for a multidimensional problem. Eur J Cancer. 2008 May;44(8):1124- 32; Fearon K. et al.: Definition and classification of cancer cachexiat an international consensus. Lancet Oncol. 2011 May; 12(5):489-95.; or Tisdale MJ.: Mechanisms of cancer cachexia.
Physiol Rev. 2009 Apr;89(2) :381-410.
In addition to completely preventing or completely reverting cancer cachexia, the treatment methods and products for use in these methods according to the invention may increase the body weight of the treated mammal compared to its body weight before the onset of cancer cachexia. As used herein, the term "before the onset of cancer cachexia" means a point in time during the course of the cancer disease, after which cancer cachexia becomes measurable by the methods known in the art such as the methods referred to above.
Preferably, the above-defined effects of the cancer cachexia treatment according to the invention are statistically significant when assessed against a suitable control group whereas individual patients who are treated would not show significant cachexia.
In accordance with the present invention, each occurrence of the term "comprising" may optionally be substituted with the term "consisting of".
Methods and Techniques Generally, unless otherwise defined herein, the methods used in the present invention (e.g. cloning methods or methods relating to antibodies) are performed in accordance with procedures known in the art, e.g. the procedures described in Sambrook et al. ("Molecular Cloning: A Laboratory Manual.", 2nd Ed., Cold Spring Harbor Laboratory Press, Cold Spring Harbor, New York 1989), Ausubel et al. ("Current Protocols in Molecular Biology." Greene Publishing Associates and Wiley Interscience; New York 1992), and Harlow and Lane ("Antibodies: A Laboratory Manual" Cold Spring Harbor Laboratory Press, Cold Spring Harbor, New York 1988), all of which are incorporated herein by reference.
Molecular weight is measured by methods known in the art such as mass spectrometry. It is expressed in Dalton (Da) or Kilodalton (kDa).
Binding of inonoclonal anti-human-GDF-lS antibodies according to the invention is generally assessed by employing surface plasmon resonance measurements using a Biorad ProteOn XPR36 system and Biorad GLC sensor chips as described for murine anti-human GDF-15 mAb-Bl-23 in Example 1.
Sequence Alignments of sequences according to the invention are performed by using the BLAST algorithm (see Altschul et al. (1990) "Basic local alignment search tool." Journal of Molecular Biology 215. p. 403-410.; Altschul et al. : (1997) Gapped BLAST and PSI-BLAST: a new generation of protein database search programs. Nucleic Acids Res. 25:3389-3402.).
Preferably, the following parameters are used: Max target sequences 10; Word size 3; BLOSUM 62 matrix; gap costs: existence 11, extension 1; conditional compositional score matrix adjustment. Thus, when used in connection with sequences, terms such as "identity" or "identical" refer to the identity value obtained by using the BLAST algorithm.
Monoclonal antibodies according to the invention can be produced by any method known in the art, including but not limited to the methods referred to in Siegel DL ("Recombinant monoclonal antibody technology." Transfus din Biol. 2002 Jan;9(1h15-22.). In a preferred embodiment, an antibody according to the invention is produced by the hybridoma cell line Bl-23 deposited with the Deutsche Sammlung für Mikroorganismen und Zellkulturen GmbH (DSMZ) under the accession No. DSM ACC3142 under the Budapest treaty. The deposit was filed on September 29, 2011.
Cell proliferation can be measured by suitable methods known in the art, including (but not limited to) visual microscopy, metabolic assays such as those which measure mitochondrial redox potential (e.g. MTT (3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide) assay; Resazurin staining which is also known as Alamar Blue® assay), staining of known endogenous proliferation biomarkers (e.g. Ki-67), and methods measuring cellular DNA synthesis (e.g. BrdU and [3H]-Thymidine incorporation assays).
Immunosuppression can be measured by suitable methods known in the art, including (but not limited to) immune cell proliferation, cytokine secretion, intracellular cytokine staining by flow cytometry, cytokine measurement by qRT-PCR, redirected target cell lysis, further cytotoxicity or degranulation assays, downregulation of activating immune cell receptors (like NKG2D), upregulation of inhibitory immune cell receptors, immunological synapse formation, immune cell infiltration. For the term immunosuppression to apply, an effect shall be measurable in at least one of these or in any other suitable assay. The lack of effect in a specific test does not imply a general absence of immunosuppres s ion.
Human GDF-15 levels can be measured by any method known in the art, including measurements of GDF-15 mRNA levels by methods including (but not limited to) quantitative real-time PCR (qRT-PCR) for human GDF-15 mRNA using primers specific to human GDF-15, mRNA in situ hybridization with probes specific to human GDF-15, rnRNA deep sequencing methods; and including measurements of GDF-15 protein levels by methods including (but not limited to) mass spectrometry for proteins or peptides derived from human GDF-15, Western Blotting using antibodies specific to human GDF-15, flow cytometry using antibodies specific to human GDF-lS, strip tests using antibodies specific to human GDF-lS, or immunocytochemistry using antibodies specific to human GDF-l5. For such methods using antibodies specific to human GDF-l5, the anti-human GDF-15 antibodies of the present invention are preferred, and the antibody of the invention produced by the hybridoma cell line 31-23 deposited with the Deutsche Sammlung für Mikroorganismen und Zellkulturen GmbH (DSMZ) under the accession No. DSM ACC3142 is most preferred.
Embodiments of the Invention As described above, the inventors show that human GDF-15 protein can be targeted by an antibody in accordance with the invention in a way that cancer cachexia and cancer-induced weight loss can be treated and that also cancer growth is inhibited.
When taking into account the present invention, it becomes clear that the anti-GDF-lS antibodies known from WO 2005/099746, WO 2009/021293 and Johnen H et al., Nature Medicine, 2007 only inhibit one of the effects of htman GDF- (i.e. cancer-induced weight loss), but fail to inhibit other effects of human GDF-15 such as those related to cancer growth. In view of the present invention, one possible explanation for this failure is that the antibodies known from the above documents may only interfere with transport of human GDF-lS across the blood-brain barrier (by forming a large complex that cannot be transported across the blood-brain barrier) but are incapable of binding human GDF-l5 in a way that renders it generally unable to interact with its receptor (e.g. a receptor residing on cells outside the brain). Furthermore, and different from the antibodies of the present invention, the anti-GDF-l5 antibodies known from WO 2005/099746, Wa 2009/021293 and Jobnen H et al., Nature Medicine, 2007 did not lead to a detectable increase in the body weight of the mammals compared to its body weight before he onset of cancer cachexia.
Accordingly, the effects of the antibodies for use according to the invention are unexpected in view of the art.
The following properties of the antibodies of the present invention are expected to contribute to their capability of inhibiting the effects of human GDF-l5 more completely, including the treatment of cachexia and the inhibition of cancer growth: Broad Binding Specificity to Forms of Human GDF-lS The antibodies of the present invention are capable of binding to mature recombinant human GDF-lS (represented by SEQ ID No: 8) and are therefore capable of binding to active, fully processed (mature) human GDF-l5.
Additionally, by performing staining experiments with the murine mAb-Bl-23 antibody according to the invention on human cells, the inventors show that the mAb-Bl-23 antibody according to the invention is capable of binding to the human GDF-lS precursor on human cells.
Thus, it is expected that binding and effects of the antibodies according to the present invention, in particular the inhibition of cancer growth, are not generally limited to effects on a particular form of human GDF-l5.
As to the effects of human GDF-lS on cancer cachexia, these effects may be caused a subset of forms human GDF-lS, for instance to soluble forms human GDF-l5, which are capable of passing the blood-brain barrier. As exemplified in the Examples of the present invention, all of the tested anti-GDF-l5 antibodies according to the invention can be used to treat cancer-induced cachexia. Thus; the antibodies according to the present invention can interfere with the forms of human GDF-lS which are responsible for cancer cachexia.
High Binding Affinity The antibodies and antigen binding portions thereof according to the invention have high binding affinity, as demonstrated by the tnAb-Bl-23 antibody according to the invention which has an equilibrium dissociation constant of about 79OpM for recombinant human GDF-15. Notably, such affinity values are superior to most of the existing therapeutic antibodies, e.g. to the therapeutic antibody Rituximab which has an equilibrium dissociation constant of about 8 nM.
High binding affinity will ensure that the antibody to human GDF-lS according to the invention stably binds to human GDF- 15, such that effects of human GDF-l5 including effects on cancer growth are effectively inhibited. Likewise, stable binding of the antibodies according to the invention is expected to ensure that forms of human GDF-15 which cause cancer cachexia cannot carry out their pathological function.
This may for instance be due to an antibody-dependent sequestration of these forms of human GDF-lS from their possible site of action in the brain. Such binding and sequestration may take place at the site of the cancer, or the antibodies according to the invention may interfere with the transport of human-GDF-15 across the blood brain-barrier.
Binding to a Discontinuous or Conformational Epitope The antibodies and antigen binding portions thereof according to the invention bind to a discontinuous or conformational epitope, as demonstrated below for amurine mAb-Bl-23 antibody according to the invention.
Binding of antibodies and antigen binding portions thereof according to the invention to a discontinuous or conformational GDF-lS epitope may help to keep human GDF-lS in a specific conformation. This conformation-specificity may be advantageous to keep GDF-15 in a form that cannot be released from the tumor, or that cannot cross the blood brain-barrier and cause cancer cachexia at a possible site of action in the brain. Additionally, such binding to a discontinuous or conformational GDF-l5 epitope may contribute to the effective inhibition of effects of human GDF-l5 including effects on cancer growth, e.g. by keeping GDF-15 in a conformation that cannot functionally interact with its receptor.
Thus, the invention relates to the following embodiments: A) Antibodies, Vectors and Cell Lines Concretely, a monoclonal antibody capable of binding to human GDF-15, or an antigen-binding portion thereof is provided, wherein the heavy chain variable domain comprises a CDR3 region comprising the amino acid sequence of SEQ ID NO: S or an amino acid sequence at least 90% identical thereto, and wherein the light chain variable domain comprises a CDR3 region comprising the amino acid sequence of SEQ ID NO: 7 or an amino acid sequence at least 85% identical thereto.
Alternatively, a monoclonal antibody capable of binding to human GDF-l5, or an aütigen-binding portion thereof is provided, wherein the heavy chain variable domain comprises a CDR3 region comprising the amino acid sequence of SEQ ID NO: S or an amino acid sequence that differs by not more than one amino acid from the amino acid sequence of SEQ ID NO: 5, and wherein the light chain variable domain comprises a CR3 region comprising the amino acid sequence of SEQ ID NO: 7 or an amino acid sequence or an amino acid sequence that differs by not more than one amino acid from the amino acid sequence of SEQ ID NO: 7.
In a second embodiment in accordance with the above embodiments, the heavy chain variable domain of the monoclonal antibody or antigen-binding portion thereof comprises a CDR3 region comprising the amino acid sequence of SEQ ID NO: 5, or the light chain variable domain comprises a CDR3 region comprising the amino acid sequence of SEQ ID NO: 7.
In a third embodiment in accordance with the above embodiments, the heavy chain variable domain of the monoclonal antibody or antigen-binding portion thereof comprises a CR3 region comprising the amino acid sequence of SEQ ID NO: 5, and the light chain variable domain comprises a CDR3 region comprising the amino acid sequence of SEQ ID NO: 7.
In a fourth embodiment in accordance with the above embodiments, the heavy chain variable domain of the monoclonal antibody or antigen-binding portion thereof comprises a CDRJL region comprising the aminO acid sequence of SEQ ID NO: 3 and a CDR2 region comprising the amino acid sequence of SEQ ID NO: 4, and the light chain variable domain of the monoclonal antibody or antigen-binding portion thereof comprises a CDIII region comprising the amino acid sequence of SEQ ID NO: 6 and a CDR2 region comprising the amino acid sequence ser-ala-ser.
In a fifth embodiment in accordance with the above embodiments, the constant domain of the heavy chain of the monoclonal antibody or antigen-binding portion thereof comprises the amino acid sequence of SEQ ID No: 29, or an amino acid sequence at least 85%, preferably at least 90%, more preferably at least 95% identical thereto, and the constant domain of the light chain of the monoclonal antibody or antigen-binding portion thereof comprises the amino acid sequence of SEQ ID No: 32, or an amino acid sequence at least 85%, preferably at least 90%, more preferably at least 95% identical thereto. In a preferred aspect of this embodiment, the constant domain of the heavy chain comprises the amino acid sequence of SEQ ID No: 29, or an amino acid sequence at* least 98%, preferably at least 99% identical thereto, and the constant domain of the light chain comprises the amino acid sequence of SEQ ID No: 32, or an amino acid sequence at least 98%, preferably at least 99% identical thereto. In a more preferred aspect of this embodiment, the constant domain of the heavy chain comprises the amino acid sequence of SEQ ID No: 29, and the constant domain of the light chain comprises the amino acid sequence of SEQ ID No: 32.
In a sixth embodiment in accordance with the above embodiments, the antibody is a humanized antibody, and all of the variable domains of the antibody are humanized variable domains.
In a further embodiment in accordance with the above embodiments, the heavy chain variable domain of the monoclonal antibody or antigen-binding portion thereof comprises the amino acid sequence of SEQ ID No: 28, or an amino acid sequence at least 901, preferably at least 95%, more preferably at least 98%, still more preferably at least 99% identical thereto, and the light chain variable domain of the monoclonal antibody or antigen-binding portion thereof comprises the amino acid sequence of SEQ ID No; 31, or an amino acid sequence at least 90%, preferably at least 95%, more preferably at least 98%, still more preferably at least 99% identical thereto. In the most preferred aspect of this embodiment, the heavy chain variable domain comprises the amino acid sequence of SEQ ID No: 28, and the light chain variable domain comprises the amino acid sequence of SEQ ID No: 31.
In a further preferred embodiment in accordance with the above embodiments, the heavy chain of the monoclonal antibody or antigen-binding portion thereof comprises the amino acid sequence of SEQ ID No: 27, and the light chain of the monoclonal antibody or antigen-binding portion thereof comprises the amino acid sequence of SEQ ID No: 30.
In. a another preferred embodiment in accordance with the above embodiments, the heavy chain variable domain of the monoclonal antibody or antigen-binding portion thereof comprises the amino acid sequence of SEQ ID No: 34, or an amino acid sequence at least 75%, more preferably at least 801, more preferably at least 851, more preferably at least 90%, more preferably at least 95%, more preferably at least 98%, still more preferably at least 99% identical thereto, and the light chain variable domain of the monoclonal antibody or antigen-binding portion thereof comprises the amino acid sequence of SEQ ID No: 37, or an amino acid sequence at least 80%, more preferably at least 85%, more preferably at least 90%, more preferably at least 95%, more preferably at least 98%, still more preferably at least 99% identical thereto. In the most preferred aspect of this embodiment in accordance with the above embodiments, the heavy chain variable domain comprises the amino acid sequence of SEQ ID No: 34, and the light chain variable domain comprises the amino acid sequence of SEQ ID No: 37.
In still another embodiment in accordance with the above first to third embodiment, the heavy chain variable domain comprises a region comprising an FR1, a CDR1, an FR2, a CDR2 and an FR3 region and comprising the amino acid sequence of SEQ ID NO: 1 or a sequence 85%, 90%-, 91%-, 92%, 93%, 941, 95%, 96%, 971, 98% or 99% identical thereto, and the light chain variable domain comprises a region comprising an FR1, a CDR1, an FR2, a CDR2 and an FR3 region and comprising the amino acid sequence of SEQ ID NO: 2 or a sequence 85%, 901, 91%, 921, 93%, 94%, 95%, 96%, 97%, 981 or 99% identical thereto.
In a preferred embodiment in accordance with the above first to third embodiment, the heavy chain variable domain comprises a region comprising an FR1, a CDR1, an FR2, a CDR2 and an FR3 region and comprising the amino acid sequence of SEQ ID NO: 1 or a sequence 951 identical thereto, and the light chain variable domain comprises a region comprising an FR.l, a CDR1, an FR2, a CDR2 and an FR3 region and comprising the amino acid sequence of SEQ ID NO: 2 or a sequence 951 identical thereto.
In a more preferred embodiment in accordance with the above first to third embodiment, the heavy chain variable domain comprises a region comprising an FR1, a CDR1, an FR2, a CR2 and an PR3 region and comprising the amino acid sequence of SEQ ID NO: 1 or a sequence 98% identical thereto, and the light chain variable domain comprises a region comprising an FR1, a CDR1, an FR2, a CDR2 and an FR3 region and comprising the amino acid sequence of SEQ ID NO; 2 or a sequence 98% identical thereto.
In a still more preferred embodiment in accordance with the above first to third embodiment, the heavy chain variable domain comprises a region comprising an FR1, a CDR1, an FR2, a CDR2 and an FR3 region and comprising the amino acid sequence of SEQ ID NO: 1, and the light chain variable domain comprises a region comprising an FR1, a CR1, an FR2, a CDR2 and an FR3 region and comprising the amino acid sequence of SEQ ID NO: 2.
Further, a monoclonal antibody capable of binding to human GDF-lS, or an antigen-binding portion thereof is provided, wherein the heavy chain variable domain comprises a CDR1 region comprising the amino acid sequence of SEQ ID NO: 3 and a CDR2 region comprising the amino acid sequence of SEQ ID NO: 4, and wherein the light chain variable domain comprises a CDRJ. region comprising the amino acid sequence of SEQ ID NO: 6 and a CDR2 region comprising the amino acid sequence of SEQ ID NO: 7. In a preferred aspect of this embodiment, the antibody may have CDR3 sequences as defined in any of the embodiments of the invention described above.
In another embodiment, the a monoclonal antibody capable of binding to human GDF-lS, or an antigen-binding portion thereof is provided, wherein the antibody or antigen-binding portion thereof is capable of inhibiting cancer growth in a mammal, preferably a human patient.
In another embodiment in accordance with the above embodiment, an antigen-binding portion capable of binding to human GDF-lS is provided, wherein the antigen-binding portion is a single-domain antibody (also referred to as "Nanobody"). In one aspect of this embodiment, the single-domain antibody comprises the CDR1, CDR2, and CDR3 amino acid sequences of SEQ ID NO; 3, SEQ ID NO: 4, and SEQ ID NO: 5, respectively. In another aspect of this embodiment, the single-domain antibody comprises the CDR1, CDR2, and CDR3 amino acid sequences of SEQ ID NO: 6, SEQ ID NO: 7, and SEQ ID NO; 7, respectively. In a preferred aspect of this embodiment, the single-domain antibody is a humanized antibody.
Preferably, the antibodies in accordance with the invention capable of binding to human GDF-lS or the antigen-binding portions thereof have an equilibrium dissociation constant for human GDF-l5 that is equal to or less than 100 nM, less than 20 nN, preferably less than 10 nN, more preferably less than 5 nN arid most preferably between 0.1 flY! and 2 riM.
In *another embodiment, the antibody capable of binding to human GDF-15 or the antigen-binding portion thereof binds to the same human GDF-lS epitope as the antibody to human GDF-15 obtainable from the cell line Bl-23 deposited with the Deutsche Sammlung für Mikroorganismen und Zellkulturen GrnbH (DMSZ) under the accession No. DSM ACC3142. As described herein, antibody binding to human GDF-15 in accordance with the present invention is assessed by surface plasmon resonance measurements as a reference standard method, in accordance with the procedures described in Example 1.
Binding to the same epitope on human GDF-15 can be assessed similarly by surface plasmon resonance competitive binding experiments of the antibody to human GDF-15 obtainable from the cell line B1-23 and the antibody that is expected to bind to the same human GDF-l5 epitope as the antibody to human GDF-15 obtainable from the cell line Bl-23.
In a very preferred embodiment, the antibody is the monoclonal antibody capable of binding to human GDF-15 obtainable from the cell line B1-23 deposited with the Deutsche Sammlung für Mikroorganismen und Zellkulturen GmbH (DMSZ) under the accession No. DSM ACC3142 or an antigen-binding portion thereof.
In a preferred embodiment, the antibody capable of binding to human GDF-lS or the antigen-binding portion thereof is a humanized monoclonal antibody or an antigen-binding portion thereof. For any given non-human antibody sequence in accordance with the invention (i.e. a donor antibody sequence), humanized monoclonaJ. anti-human-GDF-1S antibodies of the invention or antigen-binding portions thereof can be generated in accordance with techniques known in the art, as described above.
In a very preferred embodiment, the monoclonal antibody capable of binding to human GDF-15 or antigen-binding portion thereof is a humanized antibody derived from the monoclonal antibody capable of binding to human GDF-15 obtainable from the cell line Bl-23 deposited with the Deutsche Sammiung für Mikroorganismen und Zeilkulturen Grubli (DMSZ) under the accession No. DSM ACC3142, or an antigen-binding portion thereof. In a non-limiting aspect of this embodiment, the heavy chain variable domain of the humanized antibody or antigen-binding portion thereof comprises a CDR3 region comprising the amino acid sequence of SEQ ID NO: 5, and the light chain variable domain of the humanized antibody or antigen-binding portion thereof comprises a CDR3 region comprising the amino acid sequence of SEQ ID NO: 7. In a further non-limiting aspect of this embodiment, the heavy chain variable domain of the humanized antibody or antigen-binding portion thereof comprises or further comprises a CDR1 region comprising the amino acid sequence of SEQ ID NO: 3 and a CDR2 region comprising the amino acid sequence of SEQ ID NO: 4, and the light chain variable domain of the humanized antibody or antigen-binding portion thereof comprises or further comprises a CDR1 region comprising the amino acid sequence of SEQ ID NO: 6 and a CDR2 region comprising the amino acid sequence of SEQ ID NO: 7.
Further, a monoclonal antibody capable of binding to human GDF-15, or an antigen-binding portion thereof is provided, wherein the binding is binding to a conformational or discontinuous epitope on human GDF-l5 comprised by the amino acid sequences of SEQ ID No: 25 and SEQ ID No: 26. In a preferred aspect of this embodiment, the antibody or antigen-binding portion thereof is an antibody or antigen-binding portion thereof as defined in any one of the above embodiments.
In another embodiment in accordance with the above embodiments, the antibody capable of binding to human GDF-lS or the antigen-binding portion thereof is a diabody. In one aspect of this embodiment, the diabody is bivalent and monospecific, with two identical antigen binding sites for human GDF-lS. In a second, alternative aspect of this embodiment, the diabody is bivalent and bispecific, with one antigen binding site being a binding site for human GDF-15, and the other antigen binding site being a binding site f or a different antigen. Non-limiting examples for the different antigen according to this second aspect of this embodiment are 1) cell surface antigens that are co-expressed with GDF-lb at high levels on the same cancer (e.g. at higher levels compared to a control sample of the same patient obtained from a non-cancerous part of the tissue which is the tissue of origin of the cancer), and ii) cell surface antigens on cells of the immune system which are known as useful antigens for the recruitment of cells of the immune system to the tumor.
In still another embodiment in accordance with the above embodiments, the antibody capable of binding to human GDF-15 or the antigen-binding portion thereof is linked to a drug.
In non-limiting aspects of this embodiment, the drug dan be a known anticancer agent and/or an immune-stiniulatory molecule.
Known anticancer agents include alkylating agents such as cisplatin, carboplatin, oxaliplatin, mechlorethamine, cyclophosphamide, chlorambucil, and ifosfamide; anti-metabolites such as azathioprine and mercaptopurine; alkaloids such as ymca alkaloids (e.g. vincristine, vinblastine, vinorelbine, and vindesine), taxanes (e.g. paclita.xel, docetaxel) etoposide and teniposide; topoisomerase inhibitors such as camptothecins (e.g. irinotecan and topotecan); cytotoxic antibiotics such as actinomycin, anthracyclines, doxorubicin, daunorubicin, valrubicin, idarubicin, epirubicin, bleomycin, plicamycin and mitomycin; and radioisotopes. Linking of the antibodies or the antigen-binding portions thereof of the invention to anticancer agents is expected to result in stronger cancer tumor growth inhibition compared to the antibody without the anticancer agent, because the resulting conjugate will accumulate at the site of the tumor due to the presence of GDF-15 in the tumor, leading to the accumulation of the anticancer agent at the site of the tumor and to enhanced effects of the anticancer agent on the tumor.
In a further embodiment in accordance with the above embodiments, the antibody capable of binding to human GDF-15 or the antigen-binding portion thereof is modified by an amino acid tag. Non-limiting examples of such tags include Polyhistidin (His-) tags, FLAG-tag, Hemagglutinin (HA) tag, glycoprotein ID (gD) tag, and c-myc tag. Tags may be used for various purposes. For instance, they may be used to assist purification of the antibody capable of binding to human GDF-or the antigen-binding portion thereof, or they may be used for detection of the antibody or the antigen-binding portion thereof (e.g. when used in diagnostic assays).
Preferably, such tags are present at the C-terminus or N-terminus of the antibody capable of binding to human GDF-15 or the antigen-binding portion thereof.
In a preferred embodiment in accordance with the above embodiments, the antibody capable of binding to human GDF-15 or the antigen-binding portion thereof is capable of inhibiting cancer growth in a mammal, preferably a human patient.
In another preferred embodiment in accordance with the above embodiments, the human GDF-l5 is recombinant human GDF-l5 having the amino acid sequence represented by SEQ ID No: 8.
In still another preferred embodiment in accordance with the above embodiments, the binding of the antibody capable of binding to human GDF-l5 or the antigen-binding portion thereof is a binding to a conformational or discontinuous epitope on human GDF-lS.
Preferably, the monoclonal antibodies used according to the present invention capable of binding to human GDF-l5 or the antigen-binding portions thereof are isolated antibodies.
An expression vector comprising a nucleotide sequence encoding the antibody or antigen-binding portion thereof as defined above is also provided.
Further, a cell line capable of producing an antibody or antigen-binding portion thereof according to the present invention is provided.
In one embodiment, the cell line can be derived from any cell line that is known in that art and suitable for the production of antibodies or antigen-binding portions thereof.
In a preferred embodiment, the cell line is the cell line Bl- 23 deposited with the Deutsche Sammiung für Mikroorganismen und Zelikulturen GntbH (DMSZ) under the accession No. DSM ACC3 142.
In another preferred embodiment, the cell line contains an expression vector according to the invention as defined above.
B) Pharmaceutical Compositions In a further embodiment, a pharmaceutical composition comprising any of the antibodies or antigen-binding portions thereof as defined above is provided in accordance with the invention.
Pharmaceutical compositions in accordance with the present invention are prepared in accordance with known standards for the preparation of pharmaceutical compositions containing antibodies and portions thereof.
For instance, the compositions are prepared in a way that they can be stored and administered appropriately, e.g. by using pharmaceutically acceptable components such as carriers, excipients or stabilizers.
Such pharmaceutically acceptable components are not toxic in the amotnts used when administering the pharmaceutical composition to a patient. The pharmaceutical acceptable components added to the pharmaceutical compositions may depend on the particular intended use of the pharmaceutical compositions and the route of administration.
In general, the pharmaceutically acceptable components used in connection with the present invention are used in accordance with knowledge available in the art, e.g. from Remington's Pharmaceutical Sciences, Ed. AR Gennaro, 20th edition, 2000, Williams & Wilkins, PA, USA.
C) Therapeutic Methods and Products for Use in these Methods The present invention further relates to a method for treating cancer cachexia ma mammal. The method comprises administering an antibody or antigen-binding portion thereof as defined above, or a pharmaceutical composition as defined above to said mammal. Alternatively, the present invention relates to an antibody or antigen-binding portion thereof as defined above, or a pharmaceutical composition as defined above for use in these methods. In a very preferred aspect of these embodiments, the mammal is a human patient.
When taking into account the present invention, it becomes clear that the anti-GDF-lS antibodies known from WO 2005/099746, WO 2009/021293 and Johnen H et al., Nature Medicine, 2007 only inhibit cancer-induced weight loss, but fail to inhibit other effects of human GDF-lS such as those related to cancer growth.
The present invention relates to several surprising advantages compared to the effects observed in the art.
In particular, one main benefit of the invention lies in that the anti-GDF-15 antibodies disclosed herein can be used to more effectively treat cancer-induced weight loss and/or cancer cachexia.
For instance, the treatment with the antibodies according to the invention can completely prevent cancer cachexia (when given prophylactically) or completely reverse cancer cachexia (when given after the onset of cancer cachexia) Moreover, the antibodies according to the invention can even increase the body weight of the treated mammal during a prophylactic treatment for the prevention of cachexia.
Likewise, it is expected that in the course of a therapeutic treatment started after the onset of cancer cachexia, the antibodies according to the invention can not only reverse the loss in body weight, but also increase the body weight of the treated mammal compared to its body weight before the onset of cancer cachexia.
This unexpected effect of the antibodies according to the invention may be beneficial in various clinical situations.
For instance1 administration of many ingredients that are pharmaceutically active against cancer (e.g. various chemotherapeutic drugs) can lead to a loss of body weight of mammals including human patients. Such an additional loss in body weight could be counteracted by the increase in body weight due to the administration of the antibodies according to the invention. Therefore, the uses of the antibodies according to the invention may be particularly advantageous and safe for combination regimens with additional chemotherapeutic drugs. Similarly, the uses of the antibodies according to the invention may be particularly advantageous for mammals such as human patients that already had a low body weight prior to the onset of cancer and/or prior to the onset of cancer cachexia. Patients with a low body weight may for instance be cachectic patients, e.g. patients with a body-mass-index of less than 18 kg/m2.
Moreover, unexpectedly, according to the invention, the antibodies are not only effective for the treatment of cancer cachexia, but also effective for the treatment of cancer.
Thus, the treatment methods and products for use of the antibodies according to the invention are expected to be particularly beneficial for the treatment of cancer patient sub-groups which suffer from cancer-induced weight loss and/or cancer cachexia, respectively.
However, the effects according to the invention are also expected to be advantageous for the treatment of a complete patient group of a cancer referred to herein: y using the antibodies according to the invention that are effective both against the cancer itself and against cancer-induced weight loss and/or cancer cachexia, cancer and cancer cachexia treatments may be simplified by using the same treatment for all cancer patients, irrespective of whether or not they suffer from cancer-induced weight loss and/or cancer cachexia. This is because due to the dual effects of the antibodies against cancer and cancer cachexia, it is expected that these antibodies will obviate the need for additional drugs for the treatment of cancer cachexia.
Likewise, due to the dual effects of the antibodies in accordance with the invention, it may also become unnecessary to diagnose cancer-induced weight loss and/or cancer cachexia. Hence it is expected that the overall costs of therapy and diagnosis will be reduced.
Therefore, in a preferred embodiment of the above methods, or antibodies, antigen-binding portions thereof or pharmaceutical compositions for use in these methods according to the invention, the method for treating cancer cachexia is a method for completely preventing or completely reverting cancer cachexia. In a more preferred embodiment of this method, or the antibodies, antigen-binding portions thereof or pharmaceutical compositions for use in this method, the method for treating cancer cachexia is a method for completely preventing cancer cachexia. In an alternative more preferred embodiment of this method, or the antibodies, antigen-binding portions thereof or pharmaceutical compositions for use in this method, the method for treating cancer cachexia is a method for completely reverting cancer cachexia.
In a preferred embodiment of the above methods, or antibodies, antigen-binding portions thereof or pharmaceutical compositions for use in these methods according to the invention, only mammals suffering from both 1) the cancer1 and ii) cancer cachexia are treated in the method.
In a preferred embodiment of the above methods, or antibodies, antigen-binding portions thereof or pharmaceutical compositions for use in these methods according to the invention, the method increases body weight of the mammal compared to its body weight before the onset of cancer cachexia. Preferably, the increase in body weight of the mammal is at least 1.5%, preferably at least 2.51, more preferably at least 5% compared to its body weight before the onset of cancer cachexia.
In a preferred embodiment of the above methods, or antibodies, antigen-binding portions thereof or pharmaceutical compositions for use in these methods according to the invention, the method is a method for both treating cancer and treating cancer cachexia in the same mammal.
In a preferred embodiment of the above methods, or antibodies, antigen-binding portions thereof or pharmaceutical compositions for use in these methods according to the invention, the antibody has a size of more than 100 kDa, preferably more than 110 kDa, more preferably more than 120 kDa, still more preferably more than 130 kDa, and most preferably more than 140 kDa. Preferably, the antibody is a full-length antibody, more preferably a full-length IgO antibody.
In a further preferred embodiment of the above methods, or antibodies, antigen-binding portions thereof or pharmaceutical compositions for use in these methods according to the invention, the antibody has an Fc portion which is capable of binding to the Fc receptor.
In a preferred embodiment of the above methods, or antibodies, antigen-binding portions thereof or pharmaceutical compositions for use in these methods according to the invention, the cancer cells of the mammal endogenously express GDF-15 and/or the cancer cells of the mammal stimulate endogenous expression of GDF-lS in non-cancerous cells of the mammal.
In a preferred embodiment of the above methods, or antibodies, antigen-binding portions thereof or pharmaceutical compositions for use in these methods according to the invention, the cancer cells of the mammal are characterized in that they endogenously express GDF-15.
In a preferred embodiment of the above methods, or antibodies, antigen-binding portions thereof or pharmaceutical compositions for use in these methods according to the invention, the mammal is human patient.
In. a preferred embodiment of the above methods, or antibodies, antigen-binding portions thereof or pharmaceutical compositions for use in these methods according to the invention, the human GDF-l5 is recombinant human GDF-l5 having the amino acId sequence represented by SEQ ID No: 8.
In a preferred embodiment of the above methods, or antibodies, antigen-binding portions thereof or pharmaceutical compositions for use in these methods, the human patient has elevated GDF-l5 levels in blood serum before administration. In a patient sub-group having elevated GDF-lS levels in blood serum, the treatment methods according to the invention are expected to be particularly effective at inhibiting cancer growth. In the most preferred aspect of this embodiment, GDF-lS levels are GDF-lS protein levels measured using the antibody according to the invention obtainable from the hybridoma cell line 31-23 deposited with the Deutsche Sammlung für Mikroorganismen und Zellkulturen GmbH (DSMZ) under the accession No. DSMACC3142, preferably measured by immunochemistry.
In another embodiment of the above methods, or antibodies, antigen-binding portions thereof or pharmaceutical compositions for use in these methods, the antibody or antigen-binding portion thereof is the sole ingredient pharmaceutically active against cancer used in the method.
In an alternative embodiment of the above methods, or antibodies, antigen-binding portions thereof or pharmaceutical compositions for use in these methods, the antibody or antigen-binding portion thereof is used in combination with one or more further ingredients pharmaceutically active against cancer. In one aspect of this embodiment, the one or more further ingredients pharmaceutically active against cancer is a known anticancer agent and/or an immune-stimulatory molecule as defined above.
Thus, the anticancer agent can for instance be selected from alkylating agents such as cisplatin, carboplatin, oxaliplatin, mechlorethamine, cyclophosphamide, chiorambucil, and ifosfamide; anti-metabolites such as azathioprine and mercaptopurine; alkaloids such as ymca alkaloids (e.g. vincristine, vinblastine, vinorelbine, and vindesine), taxanes (e.g. paclitaxel, docetaxel) etoposide and teniposide; topoisomerase inhibitors such as camptothecins (e.g. irinotecan and topotecan); cytotoxic antibiotics such as actinomycin, anthracyclines, doxonibicin, daunonthicin, valrubicin, idarubicin, epirubicin, bleomycin, plicamycin and mitomycin; and radioisotopes. Due to the increasing effect of the antibodies according to the invention on body weight of the mammals including human patients, these combined uses of the antibodies or antigen-binding portions thereof and the ingredients pharmaceutically active against cancer are expected to be particularly safe, because they may compensate a possible additional weight loss resulting from the administration of the ingredients pharmaceutically active against cancer.
In a preferred embodiment of the above methods, or antibodies, antigen-binding portions thereof or pharmaceutical compositions for use in these methods, the cancer is selected from the group consisting of brain cancers including glioma, cancers of the nervous system, melanoma, Lung cancer, lip and oral cavity cancer, hepatic carcinoma, leukemia, Hodgkin lymphoma, Non-Hodgkin lyrnphoma, bladder cancer, cervix uteri cancer, corpus uteri cancer, testis cancer, thyroid cancer, kidney cancer, gallbladder cancer, multiple myeloma, nasopharynx cancer, larynx cancer, pharynx cancer, oesophagus cancer, gastrointestinal tumors including stomach and colorectal cancer, pancreatic cancer, prostate cancer, ovarian cancer and breast cancer, preferably from the group consisting of melanoma, prostate cancer, breast cancer, brain cancers including glioma, colorectal cancer, stomach cancer, oesophagus cancer and ovarian cancer, and most preferably is melanoma. In one embodiment the cancer is selected from the above group, which further comprises endometrial cancer, such as endometrial carcinoma, breast cancer including subtypes of breast cancer, in particular triple-negative breast cancer and bladder cancer such as urothelial cell carcinoma.
In another preferred embodiment of the above methods, or antibodies, antigen-binding portions thereof or pharmaceutical compositions for use in these methods, the tumor or tumors formed by the cancer have higher human GDF-l5 levels prior to administration compared to a control sample of the same patient obtained from a non-cancerous part of the tissue which is the tissue of origin of the cancer, preferably 1.2-fold higher levels, more preferably 1.5-fold higher levels, still more preferably 2-fold higher levels and most preferably S-fold higher levels. In a patient sub-group having higher GDF-l5 levels in the tumor or tumors formed by the cancer compared to the above control sample, the treatment methods according to the invention are expected to be particularly effective at inhibiting cancer growth.
In a very preferred embodiment of the above methods, or antibodies, antigen-binding portions thereof or pharmaceutical compositions for use in these methods, the method for treating cancer comprises inhibiting cancer growth. In a preferred aspect of this embodiment, cancer growth is stopped. In a more preferred aspect, the cancer shrinks.
In a preferred embodiment of the above methods, or antibodies, antigen-binding portions thereof or pharmaceutical compositions for use in these methods, the method for treating cancer comprises the induction of killing of cancer cells by JTK cells and 08+ T cells in the human patient. Due to their capability of preventing GDF-lS mediated dot-regulation of the known immune surveillance regulator NKG2D, the antibodies or antigen-binding portions thereof according to the invention are expected to restore immune surveillance and induce the killing of cancer cells by NK cells and CDB÷ T cells, in addition to effects of the antibodies or antigen-binding portions thereof that are independent of the immune system.
D) Kits Kits comprising the pharmaceutical compositions as defined above are also provided.
In one embodiment according to the invention, the kits are kits for use in the methods according to the invention as defined above.
In further embodiments, the present invention also provides a diagnostic kit comprising any of the antibodies or antigen-binding portions thereof according to the invention.
In one embodiment, the diagnostic kit may be used to detect whether the tumor or tumors of a cancer patient formed by the cancer have higher human GDF-1S levels compared to a control sample of the same patient obtained from a non-cancerous part of the tissue which is the tissue of origin of the cancer.
In another embodiment, the diagnostic kit may be used to detect whether a human cancer patient has elevated GDF-lS levels in blood serum.
E} Sequences The amino acid sequences referred to in the present application are as follows (in an N-terminal to C-terminal order; represented in the one-letter amino acid code): SEQ ID No: 1 (Region of the Heavy Chain Variable Domain comprising an FRI., a CDR1, an FR2, a CDR2 and an FR3 region from the Polypeptide Sequence of monoclonal anti-human GDF-l5 mAb-B1-23)
QVKLQQSGPGILQSSQTLSLTCSFSGFSLSTSGMCVSWIRQPSGICCLEWLAHIYWDDDKRY
NPTLKSRLTISKDPSRNQVFLKITSVDTADTATYYC
SEQ ID No: 2 (Region of the Light Chain Variable Domain comprising an FR]., a CDR1, an FR2, a CDR2 and an FR3 region from the Polypeptide Sequence of monoclonal anti-human GDF-15 mAb-Bl-23) D IVLTQSPKFMSTS VGDRVS VTCKASQNVGThVAWFLQKPGQS PKALIYSASYRYSGVPDR
FTGSGSGTDFTLTISNVQSEDLABYFC
SEQ ID No: 3 (Heavy Chain CDR1 Region Peptide Sequence of monoclonal anti-human GDF-1S mAb-B1-23):
GFSLSTSGMG
SEQ ID No: 4 (Heavy Chain CDR2 Region Peptide Sequence of monoclonal anti-human GDF-15 mAb-B1-23):
IYWDDDK
SEQ ID No: S (Heavy Chain CDR3 Region Peptide Sequence of monoclonal anti-human GDF-1S m.Ab-B1-23):
ARSSYGAMDY
SEQ ID No: 6 (Light Chain CDR1 Region Peptide Sequence of monoclonal anti-human GDF-1S mlth-B1-23):
QNVGTN
Light Chain CDR2 Region Peptide Sequence of monoclonal anti-human GDF-15 mAb-B1-23
SAS
SEQ ID No: 7 (Light Chain CDR3 Region Peptide Sequence of monoclonal anti-human GDF-1S mAb-B1-23):
QQYMIFPYT
SEQ ID No: 8 (recombinant mature human GDF-15 protein): GSPJRNGDHCPLGPGRCCRLHTVRASLEDLGWADWVLSPREVQVTMCIGACPSQFRAA1NHA
QIKTSLHRLKPDTVPAPCCVPASYNPMVLIQKTDTGVSLQTYDDLLAXDCHCI
SEQ ID No: 9 (huntan GDF-15 precursor protein):
MPGQELRTVNGSQMLLVLLVLSWLPHGGALSLAEASRASFPGPSELHSEDSRFRELRKRYE
DLLTRLRANQSWEDSNTDLVPAPAVRILTPEVRLGSGGHLHLRI SRAALPEGLPEASRLHR
ALFRLSPTASRSWDVTRPLRRQLSI1ARPQAPALHLRLSPPPSQSDQLLAESSSARPQLELH
LRPQAARGRRRARARIIGDHCPLGPGRCCRLHTVRASLEDLGWADWVLS PREVQVTMCIGAC
PSQFAPNMHAQIKTSLHRLKPDTVPAPCCVPASPMVLIQKTDTGVSLQTYDDLLAXDC
HCI
SEQ ID No: 10 (human GDF-1S precursor protein + N-terminal and C-terminal GSGS linker): GSGSGSGMPGQELRTVNGSQMLLVLLVLSWLPHGGALSLA.EASRASP'PGPSELHSEDSRFR
ELRKRYEDLLTRLRANQSWEDSNTDLVPAPAVRI LTPEVRLGSGGHLHLRISRAALPEGIP
EASRLI-IRALFRLSPTASRSWDVTRPLRRQLSLARPQAPALHLRLSPPPSQSDQLLAESSSA
RPQLELHLRPQAARGRRRARARNGDHCPLGPQRCCRLHTVRASLEDLGWADWVLS PREVQV
TMCIGACPSQFRAANMHAQIKTSLHRLKPDTVPAPCCVPASYNPMVLIQKTDTGVSLQTYD
DLIJAXDCECIGSGSGSG
SEQ ID No: 11 (Flag peptide): DYKDDDDKGG SEQ ID No; 12 (HA peptide): YPYDVPDYAG SEQ ID No: 13 (peptide derived from human GDF-15):
ELHLRPQAARGRR
SEQ ID No: 14 (peptide derived from human GDF-15):
LHLRPQAARGRRR
SEQ ID No: 15 (peptide derived from human GDF-15):
HLJRPQAARGRRRA
SEQ ID No: 16 (peptide derived from human GDF-15):
LRPQAARGRRRAR
SEQ ID No: 17 (peptide derived from human GDF-15):
RPQAARGRRRABA
SEQ ID No: 18 (peptide derived from human GDF-15):
PQAARGRRRARAR
SEQ ID No: 19 (peptide derived from human GDF-15): QAARGRRRARAP}1 SEQ ID No: 20 (peptide derived from human GDF-15):
MHAQIKTSLHRLIC
SEQ ID No: 25 (GDF-15 peptide comprising part of the CDF-l5 Epitope that binds to B1-23):
EVQVTMCIGACPSQFR
SEQ ID No: 26 (GOF-iS peptide comprising part of the GDF-15 Epitope that binds to B1-23): TDTGVSLQTYDDLLAXDCI4CI The nucleic acid sequences referred to in the present application are as follows (in a 5' to 3' order; represented in accordance with the standard nucleic acid code): SEQ ID No: 21 (DNA nucleotide sequence encoding the amino acid sequence defined in SEQ ID No: 1):
CAAGTGAAGCTGCAGCAGTCAGGCCCTGGGATATTGCAGTCCTCCCAGACCCTCAGTCTGA
CTTGTTCTTTCTCTGGGTTTTCACTGAGTACTTCTGGTATGGGTGTGAGCTGGATTCGTCA
GCCTTCA.GGAAAGGGTCTGGAGTGGCTGGCACACATTTACTGGGATGATGACAAGCGTAT
AACCCAACCCTGAAGAGCCGGCTCACAATCTCCAAGGATCCCTCCAGAAACCAGGTATTCC
TC21⁄2AGATCACCAGTGTGGACACTGCAGATACTGCCAATACTACTGT SEQ ID No: 22 (DNA nucleotide sequence encoding the amino acid sequence defined in SEQ ID No: 2):
GACATTGTGCTCACCCAGTCTCCAAAATTCATGTCCACATCAGTAGGAGACAGGGTCAZCG
TCACCTGCAAGGCCAGTCAGAATGTGGGTACTAATGTGGCCTGGTTTCTACAGAAACCAZG
GCAATCTCCTAAAGCACTTATTTACTCGGCATCCTACCGGTACAGTGGAGTCCCTGATCGC
TTCACAGGCAGTGGATCTGGGACAGATTTCACTCTCACCATCAGCAACGTGCAGTCTGAAG
ACTTGGCAGAGTATTTCTGT
SEQ ID No: 23 (DNA nucleotide sequence encoding the amino acid sequence defined in SEQ ID No: 5):
GCTCGAAGTTCCTACGGGGCAATGGACTAC
SEQ ID No: 24 (DNA nucleotide sequence encoding the amino acid sequence defined in SEQ ID No: 7):
CAGCAATATAACAACTTTCCGTACACG
SEQ ID No: 27 (amino acid sequence of the heavy chain of the H1L5 humanized 31-23 anti-GDF-15 antibody):
QITLKESGPTLVKPTQTLTLTCTFSGFSLSTSGMGVSWIRQPPGKGLEWLAHIYWDDDKRY
NPTLKSRLT I TKDPSKNQVVLTMTNMDPVDTATYYCARS SYG.14DYWGQGTLVWSSASTK
GPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTVSWNSGALTSGVHTFPAVLQSSGLYSL
SSVVTVPSSSLGTQTYI CNVN}3KPSNTKVDKKVEPICSCDKTHTCPPCPAPELLGGPSVFLF
PPKPICTLMISRTPEVTCVWDVSHEDPEVKFNWYVDCVEVHNAXTKPREEQYNSTYRVVS
VLTVLHQDWLNGKEYKCKVSNKALPAPIEKTI SKAKGQPREFQVYTLPPSREEMTIUQVSL
TCLVKGFYFSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCS
VMHEAL}HflQKSLSLS PGK SEQ ID No: 28 (amino acid sequence of the heavy chain variable domain of the H1LS humanized 31-23 anti-GDF-15 antibody):
QITLKESGPTLVKPTQTLTLTCTFSGFSLSTSGMGVSWIRQPPGKGLEWLANIYWDDDKRY
NPTLKSRLTITKDPSIC'JQVVLTMTNIvIDPVDTATYYCARS SYGANDYWGQGTLVTVSS SEQ ID No: 29 (amino acid sequence of the heavy chain constant domain of the H1LS humanized 31-23 anti-GDF-15 antibody):
ASTKGFSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTVSWNSGALTSGVHTFPAVLQSSG
LYSLSSVVTVPSSSLGTQTYIcNVNIIKPSNTKVDKKVEPICSCDKTHTCPPCPAPELLGGPS VFLFPPKPI(DTLMISRTPEVTCVVVDVSHEDPEVKFNWYSIDGVEVHMAKTKPREEQYNSTY RVVSVLTVLHQDWLNGKEYXCKVSNKALPAP IEI(TISKAKGQPREPQVYTLPPSREEMTfl QVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVOKSRWQQG1'IV
FSCSVMHEALHNHYTQKSLSLSPGK
SEQ ID No: 30 (amino acid sequence of the light chain of the H1LS humanized B1-23 anti-GDF-15 antibody):
DIVLTQSPSFLSASVGDRVT ITCICASQNVGTNVAWFQQKPGKSPKALIYSASYRYSGVPDR
FTGSGSGTEFTLTISSLQPEDFAAYFCQQYNNFPYTFGGGTKLEIKRAPSVFIFPPSDEQL
ICSGTASVVCLLNNFYPREAKVQWKVDNALQSGNSQESVTEQDSKDSTYSIJSSTLTLSKADY
EKHKVYACEVTHQGLSS PVTKSFNRGEC
SEQ ID No: 31 (amino acid sequence of the light chain variable domain of the H1L5 humanized B1-23 anti-GDF-l5 antibody):
DIVLTQSPSFLSASVGDRVTITCKASQNVGTNVAWFQQKPGKSPKALIYSASYRYSGVPDR
FTGSGSGTEFTLTISSLQPEDFAPLYFCQQ?NFPYTFGGGTKLEIKR SEQ ID No: 32 (amino acid sequence of the light chain constant domain of the H1L5 humanized E1-23 anti-GDF-1S antibody):
APSVFI FPPSDEQLKSGTASVVCLLNNFYPRRAKVQWICVDNALQSGNSQESVTEQDSKDST
YSLSSTLTLS}CADYEKHKVYACEVTHQGLSS PVTKSFNRGEC SEQ ID No: 33 (amino acid sequence of the heavy chain of the chimeric 81-23 anti-GDF-15 antibody):
QVKLQQSGPGILQSSQTLSLTCSFSGFSLSTSGMGVSWIRQPSGKGLEWLAHIYWDDDKRY
NPTLKSRLTI SKDPSRNQVFLKITSVDTADTATYYCARSSYGAMDYWGQGTSVTVSSASTK
GPSVFPLAPSSKSTSGGTAIthGCLVKDYFPEPVTVSWNSGALTSGVHTFPAVLQSSGLYSL SSVVTVPSSSLGTQTYI NVNHKPSNTKVDKKVEPKSCDKTHTCPPCPA?ELLGGPSVFLF
PPKPKDTLMI SRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAXTKPREEQYNS TYRVVS
VLTVIJHQDWLNGKEYKCKVSNXALPAPIEKTI SKAKGQPREPQVYTLPPSREEMT1C1QVS L, TCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVF'SCS
VMHEALHNHYTQKSLSLSPGK
SEQ ID No: 34 (amino acid sequence of the heavy chain variable domain of the chimeric 81-23 anti-GDF-15 antibody):
QVKLQQSGPGILQSSQTLSLTCSFSGFSLSTSGMGVSWIRQPSGKGLEWLAHIYWDDDKRY
NP'TLKSRLTI SKDPSRNQVFLKITSVDTADTATYYCARSSYGM4DYWGQGTSV'IVSS SEQ ID No; 35 (amino acid sequence of the heavy chain constant domain of the chimeric 81-23 anti-GDF-15 antibody); ASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPV'I'VSWNSGALTSGVHTFPAVLQSSG
LYSLS SVVTVPS SSLGTQTYI NVIKPSNTKVDKKVEPKS CDKTHTCPPCPAPELLGGPS
VFI1FPPKPKDTLMI SRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYNS TY
RVVSVLTVLHQDWLNGKEYXCKVSNKALPAPIEKTISKAXGQPREPQVYTLPPSREEMTKN
QVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKL.TVDKSRWQQGNV
FSCSVMHEALHNHYTQKSLSLSPGK
SEQ ID No: 36 (amino acid sequence of the light chain of the chimeric 81-23 anti-CDF-1S antibody):
DIVLTQSPKFMSTSVGDRVSVTCKASQNVGTNVAWFLQKPGQSPKALIYSASYRYSGVPDR
FTGSGSGTDFTLTISNVQSEDLAEYFCQQYNNFPYTFGGGTKLEIKRTVAAPSVFIFPPSD
EQLJKSGTASVVCLLNNFYPREAXVQWKVDNALQSGNSQESVTEQDSKDSTYSLSSTLTLSK
ADYEKHICVYACEVTRQGLSSPVTKSFTGEC
SEQ ID No; 37 (amino acid sequence of the light chain variable domain of the chimeric B1-23 antiaGDF15 antibody):
DIVLTQSPKFMSTSVGDRVSVTCKPSSQNVGTNVAWFLQKPGQSPKALIYSASYRYSGVPDR
FTGSGSGTDFTLTISNVQSEDLAEYFCQQYIqNFPYTFGGGTKLEIKRTVA SEQ ID No: 38 (amino acid sequence of the light chain constant domain of the chimeric 81-23 anti-GDF-15 antibody);
APSVFIFPPSDEQLKSGTASVVCLLNNFYPREAKVQWKVDNALQSGNSQESVTEQDSKDST
YSLSSTLTLSKADYFICHKVYACEVTHQGLSSPVT1CSFNRGEC
F) Examples
The present invention is illustrated by the following non-
limiting Examples;
Example 1: Generation and characterization of the uturine GDF-Jntibody 31-23, and generation of chimeric and humanized antibodies The antibody 31-23 was generated in a GDF-15 knock out mouse.
Recombinant human GDF-l5 (SEQ ID No: 8) was used as the immunogen.
The hybridomá cell line 31-23 producing mAb-B1-23 was deposited with the Deutsche Sammlung für Mikroorganismen und Zeilkulturen GmbH (DMSZ) under the accession No. DSM ACC3142, in accordance with the Budapest Treaty.
By means of a commercially available test strip system, 31-23 was identified as an IgG2a (kappa chain) isotype. Using surface plasmon resonance measurements, the dissociation constant (Kd) was determined as follows: Binding of the monoclonal anti-human-GDF-15 antibody anti-human GDF-15 mlth-B1-23 according to the invention was measured by employing surface plasinon resonance measurements using a Biorad ProteOn XPR3G system and Biorad GLC sensor chips For preparing the biosensors recombInant mature human GDF-15 protein was immobilized on flow cells 1 and 2. On one flow cell recombinant GDF-15 derived from Baculvirus-transfected insect cells (HighFive insect cells) and on the other recombinant protein derived from expression in E. coli was used. The GLC sensor chip was activated using Sulfo-NHS (N- Hydroxysulfosuccinimide) and EDC (l-Ethyl-3-[3-dimethylaminopropyl] carbodiimide hydrochloride) (Biorad Protean Amine Coupling Kit) according to the manufacturer' S recommendation, the sensor surface was subsequently loaded with the proteins up to a density of about SOORU (iRu = lpg nrnr2). The non-reacted coupling groups were then quenched by perfusion with lM ethanolamine pH 8.5 and the biosensor was equilibrated by perfusing the chip with running buffer (1OM HEPES, 150mM NaC1, 3.4mM EDTA, 0.005% Tween-20, pH 7.4, referred to as HBSl50). As controls two flow cells were used, one empty with no protein coupled and one coupled with an non-physiological protein partner (human Interleukin-5), which was immobilized using the same coupling chemistry and the same coupling density. For interaction measurements anti-human GDF-lS mAb-B1-23 was dissolved in HBS1SO and used in six different concentrations as analyte (concentration: 0.4, 0.8, 3, 12, 49 und 98 nM). The analyte was perfused over the biosensor using the one-shot kinetics setup to avoid intermittent regeneration, all measurements were performed at 25°C and using a flow rate of lOOpl min1. For processing the bulk face effect and unspecific binding to the sensor matrix was removed by subtracting the SPR data of the empty flow cell (flow cell 3) from all other SPR data. The resulting sensogram was analyzed using the software ProteOn Manager version 3.0. For analysis of the binding kinetics a 1:1 Langmuir-type interaction was assumed. For the association rate constant a value of 5.4÷O.06xl05 M1s1 (kon) and for the dissociation rate constant a value of 4.3i-O.03x10-4 s- (k0ff) could be determined (values are for the interaction of anti-human GDF-15 mAb-Bl-23 with GDF-l5 derived from insect cell expression). The equilibrium dissociation constant was calculated using the equation RD = koff/kon to yield a value of about 79OpM. Affinity values for the interaction of GDF-15 derived from E. coli expression and the anti-human GDF-lS mAb-Bl-23 differ by less than a factor of 2, rate constants for GDF-l5 derived from insect cells and E. coli deviate by about 45's and are thus within the accuracy of SPR measurements and likely do not reflect a real difference in affinity. Under the conditions used the anti-human GDF-15 mAb-Bl-23 shows no binding to human interleukin-5 and thus confirms the specificity of the interaction data and the anti-human GDF-l5 mAb-Bl-23.
The amino acid sequence of recombinant human GDF-lS (as expressed in Baculovirus-transfected insect cells) is:
GSARNGDHCP LGPGRCCRLH TVRASLEDLG WADWVLSPRE VQVTMCIGAC
PSQFRAANMH AQIKTSLHRL KPDTVPAPCC VPASYNPMVL IQKTDTGVSL.
QTYDDLLIAKD CHCI
(SEQ ID No: 8) Thus, using surface plasmon resonance measurements, the dissociation constant (lcd) of 79OpM was determined. As a comparison: the therapeutically used antibody Rituximab has a significantly lower affinity (lcd = 8 nM).
From the murine anti-human GDF-lS mAb-Bl-23, a chimeric anti-human GDF-15 mAb-Bl-23 antibody was generated by replacing constant domains of the murine antibody with the constant domains of a human IgGl antibody (trastuzumab backbone). The amino acid sequence of the heavy chain of this chimeric antibody is shown in SEQ ID No: 33, and the amino acid sequence of the light chain of this chimeric antibody is shown in SEQ ID Nc: 36.
From the chimeric anti-human GDF-l5 mAb-B1-23, a humanized anti-human GDF-lS mAb-B1-23 antibody was developed by humanizing the variable domains of the chimeric antibody, i.e. by replacing the framework regions of the chimeric antibody with human sequences. The amino acid sequence of the heavy chain of this humanized antibody is shown in SEQ ID No: 27, and the amino acid sequence of the light chain of this humanized antibody is shown in SEQ ID No: 30. This antibody is referred to as FIlLS anti-GDF-15 antibody, or humanized Bl- 23-H1L5 antibody, or FIlLS antibody.
In order to generate the above-mentioned chimeric anti-human GDF-15 mLb-B1-23 antibody and the humanized Bl-23-H1L5 antibody as indicated above, the cDNAs encoding the antibody sequences were optimized, and the genes were synthesized. The gene sequences were then cloned into a cloning/expression vector system. From these vectors, plasmid DNA with low endotoxin levels was synthesized.
The plasmid DNA was then transiently transfected into Cl-JO cells, followed by an analysis and quantification of antibody expression using a protein A biosensor. The cDNA of candidate cultures for antibody expression was sequenced. the obtained monoclonal antibodies were analyzed (see Examples 7 to 9).
Example 2: Antagonization of GDF-lS Mediated Effects with niAB Bi -23 a) The NKG2D (Natural Killer Group 2D) receptor, which is expressed on NK cells and CDB+ T cells, is known to play an important role in the immune surveillance against tumors Transformed as well as viral infected cells express ligands, which bind to the NKG2D receptor, thereby activating the cytotoxic effector functions of the described immune cells.
In that way transformed cells can be detected and eliminated by the immune system. After treatment of immune cells with either recombinant human GDF-l5 or tumor cell secreted GDF-1S in vitro for 72 hours, the expression level of NKG2D on the cell surface of lymphocytes was downregtilated (Figure 1).
After 72 hours incubation the immune cells were stained with the following FACS-antibodies: anti CD3, anti 056, anti-NKG2D. using this antibody combination, the experiment focused on MC cells and their NKG2D surface expression. The low NKG2D level on immune cells led to an impaired tumor/target cell lysis. The GDF-15 mediated downregulation of NKG2D was prevented by mAb B1-23.
It is therefore concluded that human GDF-lS downregulates expression of NKG2D on the cell surface of lymphocytes and thereby downregulates immune surveillance against tumors. By binding to human GDF-l5, the antibodies of the present invention are capable of preventing GDF-lS mediated downregulation of NKG2D and should be capable of restoring immune surveillance and inducing the killing of cancer cells by NK cells and CD8+ T cells.
b) The treatment of the ovarian cancer cell line SK-OV-3 with recombinant GDF-lS led to the phosphorylation of ART. ART is a molecule, which is part of the P13K-pathway and contributes to the activation and proliferation of cells. In this experiment SK-OV-3 cells were treated with 10 ng/ml recombinant GDF-l5 for 10 mm at 37°C, 5% C02. 5 minutes preincubation of 2 g mAb-Bl-23 with 10 ng/ml GDF-15 at 37°C blocked the GOF-iS mediated AKT-phosphcrylation (Figure 2).
This showed the neutralizing effect of mAb-Bl-23.
c) Treatment of immune cells with recombinant GDF-15 led to the phosphorylation of JNK, a kinase, which is activated either by cytokines or by stress. Antagonization of 10 ng/mJ.
GIDF-15 with 2 ig mA.b-B1-23 (5 minute preincubation at 37°) blocked the GDF-l5 mediated JNK1/2.-phosphorylation (Figure 3) Example 3: Inhibition of Cancer Cell Proliferation Using mAb B1-23 Data generated with B1-23 showed an antiproliferative effect of the antibody on cancer cells in vitro. The strongest antiproliferative effect was observed using the prostate cancer cell line LnCap, which produces lots of GDF-15. A metabolic assay (Alamar Blue assay) showed a decrease of proliferation of 30% after 72hrs when nthb-B1-23 was present, compared with the control group, where the antibody was not applied. Since cytotoxic effects of the antibody have been excluded in different assays, this effect proves a significantly decreased cell division rate after blockade of GDF-l5.
Example 4: nAb B1-23 inhibits Growth of tumors in vivo The following in vivo study was carried out: To assess an anti-tumor effect of B1-23 in vivo, Balb/cnu/nU nude mice were used in a xenograft setting with the melanoma cell line tJACC-257. The mice were treated either with the antibody 31-23 or with PBS. Each treatment cohort contained Balb/cnlu/h1u nude mice.
Prior to injection, the tJACC-257 melanoma cells were grown in complete medium, excluding any contamination. The cells were harvested when 70-80% confluence was reached in the cell culture flask. Cells were then washed with PBS and counted.
1x107 viable cells were suspended in PBS.
The first injection/treatment was administered in 6 week old Balb/cnu/u nude mice. The inoculation area of the mice was cleaned with ethanol. The T.JACC 257 cells were mixed and drawn into a syringe without a needle, in order to avoid negative pressure on the tumor cells. The cell suspension containing lxlo7 cells in PBS was injected subcutaneously (s.c.) into the lower flank of the mice.
The intraperitoneal (i.p.) injection of either Bl-23 (25mg/kg body weight) or the same volume of PBS started immediately after the tumor cell inoculation (defined as day 1) and was administered twice a week. The tumors were grown for 48 days.
The tumor diameters were measured with a caliper and the tumor volume in mm3 was calculated by the formula: Volume = (width)2 x length/2 The results which were obtained from the study are shown in Figure 4.
As demonstrated in the Figure, the tumor size of the animal cohort treated with B1-23 was significantly decreased, compared to the PBS control group.
Example 5: ml1⁄2b Bl-23 recognizes a conformational or a discontinuous epitope of human GDF-l5 Epitope Mapping: Monoclonal mouse antibody GDF-lS against l3mer linear peptides derived from GDF-lS Antigen: GDF-lS:
GSGSGSGMPGQELRTVNGSQMLLVLLVLSWLPHGGALSLAEASRASFPGPSELHSEDSRFR
ELRKRYEDLLTRLRANQSWEDSNTDLVPAPAVRILTPEVRLGSGGHLHLRISRAALPEGLP
EASRLHRALFRLSPTASRSWDVTRPIJRRQLSLARPQAPALHLRLSPPPSQSDQLLAESSSA
RPQLELHLRPQAARGRRRARARNGDHCPLGPGRCCRLHTVRASLEDLGWADNVLSPREVQV
TMCI GACPSQFRAANMHAQIKTS LHRLKPDTVPAPCCVPASYNPMVL IQKTDTGVSLQTYD
DLLAXDCHCIGSGSGSG (322 amino acids with linker) (SEQ ID No: 10) The protein sequence was translated into l3mer peptides with a shift of one amino acid. The C-and N-termini were elongated by a neutral GSGS linker to avoid truncated peptides (bold letters).
Control Peptides: Flag: DYKDDDDKGG (SEQ ID No:l3), 78 spots; HA: YPYDVPDYAG (SEQ ID No:14), 78 spots (each array copy) Peptide Chip Identifier: 000264_Cl (10/90, Ala2Asp linker) Staining Conditions: Standard buffer: PBS, pH 7.4 + 0.05% Tween 20 Blocking buffer: Rockland blocking buffer MB-070 Incubation buffer: Standard buffer with 10% Rockland blocking buffer MB-070 Primary sample: Monoclonal mouse antibody GDF-15 (1 pg/pl): Staining in incubation buffer for 16 h at 4°C at a dilution of l;lOO and slight shaking at 500 rpm Secondary antibody: Goat anti-mouse IgG (H+L) IRDyeG8O, staining in incubation buffer with a dilution of 1:5000 for miii at room temperature (RT) Control antibodies: Monoclona]. anti-HA (12CAS)-LL-Atto 680 (1:1000), monoclonal anti-FLAG(M2)--Fluo?robes7s2 (1:1000); staining in incubation buffer for 1 h at RT Scanner: Odyssey Imaging System, LI-COR Biosciences Settings: offset: 1mm; resolution: 21 pm; intensity green/red: 7/7 Results: After 30 mm pre-swelling in standard buffer and 30 mm in blocking buffer, the peptide array with 10, 12 and lsmer B7H3-derived linear peptides was incubated with secondary goat anti-mouse IgG (H+L) IRDyeG8O antibody only at a dilution of 1:5000 for lh at room temperature to analyze background interactions of the secondary antibody. The PEPperCHIP® was washed 2x1 mm with standard buffer, rinsed with dist. water and dried in a stream of air. Read-out was done with Odyssey Imaging System at a resolution of 21 pm and green/red intensities of 7/7: We observed a weak interaction of arginine-rich peptides (ELHLRPQAARGRR (SEQ ID No:l5), LHLRPQAARGRRR (SEQ ID No:16), HLRPQAARGRRRA (SEQ ID No:17), LRPQAARGRRRAR (SEQ ID 14o:18), RPQAARGRRRARA (SEQ ID No:19), PQAARGRRRARAR (SEQ ID No:20) and QAARGRRRARARN (SEQ ID No:21)) that are known as frequent binders, and with the basic peptide MHAQIKTSLHRLI( (SEQ ID No:22) due to ionic interactions with the charged antibody dye.
After pre-swelling for 10 mm in standard buffer, the peptide microarray was incubated overnight at 4 °C with monoclonal mouse antibody GDF-15 at a dilution of 1:100. Repeated washing in standard buffer (2xl mm) was followed by incubation for 30 mm with the secondary antibody at a dilution of 1:5000 at room temperature. After 2xlO sec.
washing in standard buffer and short rinsing with diet.
water, the PEPperCIjIP® was dried in a stream of air. Read-out was done with Odyssey Imaging System at a resolution of 21 pm and green/red intensities of 7/7 before and after staining of control peptides by anti-HA and anti-FLAG(M2) antibodies.
It was shown that none of the linear l3mer peptides derived from.GDF-lS interacted with monoclonal mouse antibody GDF-l5 even at overregulated intensities. Staining of Flag and HA control peptides that frame the array, however, gave rise to good and homogeneous spot intensities.
Summary:
The Epitope Mapping of monoclonal mouse GDF-lS antibody against GDF-15 did not reveal any linear epitope with the l3mer peptides derived from the antigen. According to this finding it is very likely that monoclonal mouse antibody GDF-recognizes a conformational or a discontinuous epitope with low affinity of partial epitopes. Due to the obvious absence of any GDF-15 signal above the background staining of the secondary antibody only, quantification of spot intensities with PepSlide® Analyzer and subsequent peptide annotation were omitted.
Example 6: Structural identification of peptide ligand epitopes by mass spectrometric epitope excision and epitope extraction The epitope of recombinant human GDF-15 which binds to the antibody Bl-23 was identified by means of the epitope excision method and epitope extraction method (Suckau et al. Proc Natl Acad Sci U S A. 1990 December; 87(24): 9848-9852.; R.Stefanescu ét al., Eur.J.Mass Spectrom. 13, 69-75 (2007)).
For preparation of the antibody column, the antibody Bl-23 was added to mIS-activated 6-aminohexanoic acid coupled sepharose. The sepharose-coupled antibody B1-23 was then loaded into a 0,8 ml microcolwnn and washed with blocking and washing buffers.
Epitope extraction experiment: Recombinant human GDF-15 was digested with trypsin for 2h at 37°C (in solution), resulting in different peptides, according to the trypsin cleavage sites in the protein. After complete digestion, the peptides were loaded on the affinity column containing the immobilized antibody B1-23. tJribound as well as potentially bound peptides of GDF-l5 were used for mass spectrornetry analysis. An identification of peptides by means of mass spectrometry was not possible. This was a further indicator that the binding region of GDF-l5 in the immune complex Bl-23 comprises a discontinuous or conformational epitope. In case of a continuous linear epitope, the digested peptides should bind its interaction partner, unless there was a trypsin cleavage site in the epitope peptide. A discontinuous or conformational epitope could be confirmed by the epitope excision method described in the following part.
Epitope excision experiment: The immobilized antibody Bl-23 on the affinity column was then incubated with recombinant GDF-15 for 2h. The formed immune complex on the affinity column was then incubated with trypsin for 2h at 37°C. The cleavage resulted in different peptides derived from the recombinant GDF-15. The immobilized antibody itself is proteolytically stable. The resulting peptides of the digested GDF-15 protein, which were shielded by the antibody and thus protected from proteolytic cleavage, were eluted under acidic conditions (TEA, ph2), collected and identified by mass spectrometry.
The epitope excision method using Ms/Ms identification resulted in the following peptides: Peptide Position in sequence Mass Ion/Charge EVQVTMCIGACPSQFR 40-55 1769.91 590.50(3+) (SEQ ID No: 25) TDTGVSLQTYDDLLAXDCHCI 94-114 2310,96 771:33(3+) (SEQ ID No: 26) The part of human GDF-lS, which binds the antibody Bl-23, comprises a discontinuous or conformational epitope. Mass spectrometry identified 2 peptides in the GDF-lS protein, which are responsible for the formation of the immune complex. These peptides are restricted to the positions 40- (EVQVTMCIGACPSQFR) and 94-114 (TDTGVSLQTYDDLLAKDCHCI) in the GDF-15 amino acid sequence. Thus, these two peptides comprise an epitope of the G]DF-15 protein that binds to the antibody Bl-23.
Example 7: Treatment of cancer-induced weight loss with anti-GDF-15 antibodies. In the underlying animal study No. 140123, Ralb/c nu/nu mice per treatment group were subcutaneously inoculated with 10x106 tJACC-257 cells per animal in a 1:1 volume ratio with matrigel (100 z1 cells + 100 ptl matrigel).
The animals were treated on the same day with the respective antibodies, as indicated below: Study groups 1-6 Amounts of substances (10 animals per group) (for 45 days) 1. Dacarbazine* (reference, Lot. No.: C120522C) 80 mg 2. PBS (SIGMA, Lot. No.: RNBDO341) 30 ml 3. B1-23 anti-GDF-15 antibody (murine, Lot. No.: 515980) 75 mg 4. Chimeric B1-23 anti-GDF-1S antibody (chimeric; Lot. :PROOS7) 75 mg 5. H1L5 anti-GEF-iS antibody (humanized B1-23, Lot. :PR3176) 75 mg 6. B12 Isotype control antibody (Isotype antibody, Lot. No.: 1D3195) 75 mg *Detidemac 500 mg (exp. : 03/2015) The dacarbazine group (group 1) served as a reference group /positive control for tumor growth arrest (cytostatic drug for the treatment of malignant melanoma in humans).
The PBS group (group 2) served as a growth control/vehicle control group, because all used substances of the other groups were administered in PBS.
The group of the murine Bl-23 lead candidate antibody (group 3) served as reference group for a comparison with the chimeric B1-23 antibody and with the humanized B1-23 H1LS (groups 4 and 5).
Group 4 is the group of the chimerized Bl-23 lead candidate antibody, which contains murine variable domains and constant domains of a human IgGl antibody (trastuzumab backbone).
Group S is the group of the H1LS humanized 81-23 lead candidate antibody, which contains humanized frameworks within the murine variable regions and constant domains of a human. IgGl antibody (trastuzumab backbone).
Group 6 is the group of the B12 isotype antibody. For thIs isotype control group, the antibody 312 (Lot. No.: 1D3195) was produced by the company Evitria AG. 812 binds to an HIV antigen and should therefore neither bind to antigens in nude mice nor to antigens of the human tumor. 312 was selected as a highly suitable isotype control, because the immunoglobulin backbone of 812 also consists of the human IgGl antibody trastuzumab and is therefore almost identical to the chimeric 81-23 and the RiLE humanized 81-23 antibodies, except for their variable regions.
The study was carried out in a double-blinded manner for the treatment with the antibodies and for the treatment with PBS.
In groups 1, 2 and 6 which did not receive anti-GDF-1S antibodies, more than 10% body weight loss was observed (i.e. weight loss to a relative body weight of less than 90% compared to day 0). In contrast, in the groups which had received treatment with the anti-GDF-15 antibodies 31-23, chimeric B1-23 and humanized Bl-23-H1L5, respectively, an increase in body weight was observed (Figure 5).
Thus, surprisingly, treatment with all of the tested anti- GDF-l5 antibodies antibodies completely prevents cancer-induced weight loss in mice. This effect was significant for all of the groups treated with anti-GDF-lS antibodies (two-way MIOVA; pcO.UE).
It is also noteworthy that the mice of the groups that did not receive treatment with anti-GDF-lS antibodies exhibited a weight loss of more than 10%. In humans, a weight loss of as little as 5% over a period of 6 months is considered as being indicative of cancer cachexia (Fearon K. et al.: Definition and classification of cancer cachexia: an international consensus. Lancet Oncol. 2011 May; 12(5h489-95J. Given the larger weight loss of the mice observed in the present study which even exceeded 10%, it is expected that the mice in the study, which did not receive treatment with anti-GDF-15 antibodies, not only exhibited weight loss but also exhibited cancer cachexia. This effect is completely prevented by the anti-GDF-15 antibodies tested. It is therefore expected that the anti-GDF-15 antibodies in accordance with the invention are capable of both treating cancer-induced weight loss and treating cancer cachexia.
Notably, the extent of weight loss did not correlate with the respective tumor size (r2=106). If the prevention of weight loss were only a secondary effect resulting from the inhibition of cancer growth and the smaller tumor sizes, a correlation between tumor size and weight loss would be expected. Thus, the lack of such correlation shows that uses of the anti-GDF-15 antibodies according to the invention result in two independent treatment effects: -an inhibition of cancer growth, and -a prevention of weight loss as an additional effect, * which independent from the inhibition of cancer growth, and which is expected to reflect a prevention of cancer cachexia.
Despite their mechanistic independence, it was observed that these effects can occur simultaneously in the same animals.
In addition to evaluating the mean body weight of the mice, the feed consumption of the mice was evaluated by pairwise comparisons of the study groups (Table 1). Notably, the feed consumption of the mice in the anti-GDF-l5 antibody groups (31-23, chimeric Bl-23 and humanized B1-23-H1LS) was significantly higher than the feed consumption of the mice in the groups which did not receive the anti-GDF-15 antibodies.
Table 1: ___________ __________ Feed vs. vs. vs. consumption chimeric humanized. 81-23 per mouse 81-23 81-23 ____________ and day __________ ___________ ___________ dacarbazine 2.8 ± 0.2 g ** ** ** PBS 2.6 ± 0.4 g ** ** ** Chimeric 3.5 ± 0.2 g B1-23 --n.s. n.s.
812 2.7 ± 0.2 g humanized 3.4 ± 0.2 g 81-23 ____________ n.s. --n.s.
81-23 3.6 ± 0.2 g n.s. n.s. -- (*pcQ.05; **p.cO.01; ***pcO.001 as assessed by unpaired two-sided Students t-test) Table 1: Comparative evaluation of the feed consumption between the different treatment groups. For the measured time intervals (day 17-20, day 20-24, day 24-27, day 27-31, day 31-34), the average feed consinuption per mouse and day was calculated for each respective group. The values are indicated together with their standard deviation.
the quality of the humanized anti-GDF-l5 antibody Bl-23-H1LS used in the study was tested by using gel electrophoresis and coomassie staining of the antibodies (see Figure 6). Notably, the band of the humanized anti-GDF-l5 antibody 81-23-U1LS was sharp and clear, whereas the bands of the murine 81-23 anti-GDF-lS antibody and the 812 control antibody appeared less sharp and at a higher molecular weight. This suggests that the humanized anti-GDF-l5 antibody Bl-23-H1LS is not prone to aggregation, and that some aggregation may have shifted the molecular weight of the other antibodies to higher values.
Additionally, by using a colorimetric assay, it was confirmed that all anti-GDF-lS antibodies used in the study bound to GDF-15 in a concentration-dependent manner. To determine the binding of 31-23 antibody variants to GDF-15, a colorimetric ELISA experiment was performed. The B12 antibody served as an isotype antibody, which does not bind to human GDF-15.
Therefore1 Maxisorp 96 well plates (Nunc) were coated with hrGDF-l5 (25 ng protein per well, 50 al volume) over night at 4°C. The following day, plates were washed to remove unbound protein (3 times with 150 4 of PBS 0.05 Tween) and non-specific binding sites were blocked with 150 jzl of PBS 1% BSA for 2 hours at room temperature. Again, plates were washed and different variants of Bl-23 test antibodies were applied (50 4 volume). To inquire specificity of the antibody binding, endpoint dilution was performed starting from 333 ng/ml and 1:3 serial dilution. As background control, PBS 1% ESA was applied. Following binding for 1 hour at room temperature, wells were washed as described above. As secondary antibody HRP conjugated Anti-human Igo (Life technologies, 1:5000) was applied for 1 hour at room temperature. Wells were washed as described above to remove unbound secondary antibody. For detection, 50 jil of peroxide substrate (TMB 1:100 in 0.1 M sodium acetate pH6) were added and following 10 minutes of incubation, 50 jzl of stop solution (2N H2S04) were added. As negative controls, wells without GDF-l5 coating and wells without secondary antibody were included. For analysis, optical densitiy at 450 nm was quantified using the ELISA reader (Tecan Sunrise) and the corresponding Magellan software). It was observed that in comparison to the 312 antibody, the humanized H1L5 antibody, the chimeric 81-23 antibody and the murine 81-23 antibody exhibited a clearly concentration-dependent binding to GDF-15.
Example 8: Determination of Rd values of anti-GDF-l5 antibodies. The Kd values of different anti-GDF-l5 antibodies were compared using Surface Acoustic Wave (SAW) gold chip biosensors technology (SAW Instruments GmbH, Schwertberger Str. 16, D-53177 Bonn, Germany): Antibody: Lcd value (nIvI) 31-23 anti-GDF-15 antibody 28.8 nM (murine, IgG2a) Chimeric 31-23 anti-GDF-15 antibody 14 nM (chimerized, human IgGi) H1L5 humanized 31-23 5,62 nM (humanized, human IgGl) Rituxumab 1116 nM (control antibody) Herceptin No binding (control antibody) The murine antibody (B].-23) as well as the chimeric 31-23 antibody were present in purified form. The H1L5 humanized 31-23 antibody was a serum-free CHO cell culture supernatant.
The Lcd value of the murine 31-23 deviates from the Lcd values determined by Biacore analyses (surface plasmon resonance) by a factor of 35.
This deviation may -apart from the differences in the measurement methods -be explained by a reduced availability of free murine 31-23 antibody, since it was found that this antibody can form aggregates in its form as mouse antibody.
The solution of the murine 31-23 antibody was therefore stabilized by addition of 0.2% BSA. Therefore, binding of the antibody to albumin may have reduced the availability of the murine 31-23 antibody and could explain the differences in the affinity values obtained by the different measurement methods. Compared to the murine 31-23 antibody, the I-11L5 hunanized Bl-23 antibody surprisingly showed no tendency to aggregate (see also Example 9 below).
In the present assay, the chimeric Bl-23 anti-GDF-l5 antibody and the H1LS humanized Bl-23 antibody exhibited affinities to human G]DF-l5 which were about 2-fold and 5-fold higher, respectively, than the affinity of the murine Bl-23 anti-GDF-antibody. Thus, the chimeric Bl-23 anti-GDF-l5 antibody and the ilLS humanized Bl-23 antibody are high affinity antibodies.
Example 9: Aggregation studies of the anti-GDF-15 antibodies In order to test the aggregation properties of anti-GOF-lS antibodies, antibody samples were shaken for 48 hours at room temperature in microcentrifuge tubes, and subsequently, the tubes were visually analyzed for aggregated antibody precipitates.
It was observed that compared to the murine Bl-23 antibody, the H1L5 humanized B1-23 antibody surprisingly showed no tendency to aggregate, even when the antibody was only present in phosphate-buffered saline (PBS), and when no stabilizing proteins such as ESA were present.
Moreover, in freeze/thaw and dilution experiments, it was observed that the ilLS humanized Bl-23 antibody did not aggregate during any of the dilution steps or freeze/thaw cycles These properties of the humanized antibody are expected to be advantageous for clinical formulation of the molecule.
G) Industrial Applicability
The antibodies, antigen-binding portions thereof, pharmaceutical compositions and kits according to the present inventjon may be industrially manufactured and sold as products for the claimed methods and uses (e.g. for treating cancer cachexia and cancer), in accordance with known standards for the manufacture of pharmaceutical products.
Accordingly, the present invention is industrially applicable.
Preferred Embodiments 1. A monoclonal antibody capable of binding to human GDF- 15, or an antigen-binding portion thereof, wherein the heavy chain variable domain comprises a CDR3 region comprising the amino acid sequence of SEQ ID NO: 5 or an amino acid sequence at least 90% identical thereto, and wherein the light chain variable domain comprises a CDR3 region comprising the amino acid sequence of SEQ ID NO: 7 or an amino acid sequence at least 85% identical thereto, for use in a method for treating cancer cachexia in a mammal.
2. A monoclonal antibody capable of binding to human GDF- 15, or an antigen-binding portion thereof, wherein the binding is binding to a conformational or discontinuous epitope on human GDF-15 comprised by the amino acid sequences of SEQ ID No: 25 and SEQ ID No: 26, for use in a method for treating cancer cachexia in a mammal.
3. The antibody or antigen-binding portion thereof of item 1 or 2 for the use according to item 1 or 2, wherein the method f or treating cancer cachexia is a method for completely preventing or completely reverting cancer cachexia.
4. The antibody or antigen-binding portion thereof of item 3 for the use according to item 3, wherein the method for treating cancer cachexia is a method for completely preventing cancer cachexia.
5. The antibody or antigen-binding portion thereof of item 3 for the use according to item 3, wherein the method for treating cancer cachexia is a method for completely reverting cancer cachexia.
6. The antibody or antigen-binding portion thereof of any one of items 1-5 for the use according to any one of items 1-5, wherein in the method, only mammals suffering from both i) the cancer, and ii) cancer cachexia are treated.
7. The antibody or antigen-binding portion thereof of any one of items 5-6 for the use according to any one of items 5-6, wherein the method increases body weight of the mammal compared to its body weight before the onset of cancer cachexia.
8. The antibody or antigen-binding portion thereof of item 7 for the use according to item 7, wherein the increase in body weight of the mammal is at least 1.5%, preferably at least 2.5%, more preferably at least 5% compared to its body weight before the onset of cancer cachexia.
9. The antibody or antigen-binding portion thereof of any one of items 1-8 for the use according to any one of items 1-8, wherein the method is a method for both treating cancer and treating cancer cachexia in the same mammal.
lO.The antibody or antigen-binding portion thereof of any one of items 1-9 for the use according to any one of items 1-9, wherein the antibody has a size of more than kna, preferably more than 110 kDa, more preferably more than 120 kiDa, still more preferably more than 130 kDa, and most preferably more than 140 kDa.
ll.The antibody or antigen-binding portion thereof of item for the use according to item 10, wherein the antibody is a full-length antibody.
12.The antibody or antigen-binding portion thereof of item 11 for the use according to item 11, wherein the antibody is a full-length IgG antibody.
13.The antibody or antigen-binding portion thereof of any one of items 1 to 12 for the use according to any one of items 1 to 12, wherein the antibody has an Fc portion which is capable of binding to the Fc receptor.
14.The antibody or antigen-binding portion thereof of any one of items 1 to 13 for the use according to any one of items 1 to 13, wherein the cancer cells of the mammal endogenously express GDF-15 and/or the cancer cells of the mammal stimulate endogenous expression of GDF-15 in non-cancerous cells of the mammal.
l5.The antibody or antigen-binding portion thereof of any one of items 1 to 14 for the use according to any one of items 1 to 14, wherein the cancer cells of the mammal endogenously express GDF-lS.
16.The antibody or antigen-binding portion thereof of any one of items 1 to 15 for the use according to any one of items 1 to 15, wherein the mammal is a human patient.
17.The antibody or antigen-binding portion thereof of any one of items 1-to 16 for the use according to any one of items 1 to 16, wherein the human GDF-lS is recombinant human GDF-15 having the amino acid sequence represented by SEQ ID No: 8.
l8.The antibody or antigen-binding portion thereof of any one of items 2-17 for the use according to any one of items 2-17, wherein the heavy chain variable domain comprises a CDR3 region comprising the amino acid sequence of SEQ ID NO; 5 or an amino acid sequence at least 90%-identical thereto, and wherein the light chain variable domain comprises a CDR3 region comprising the amino acid sequence of SEQ ID NO: 7 or an amino acid sequence at least 85% identical thereto.
19.The antibody or antigen-binding portion thereof of any one of items 1 and 3-17 for the use according to any one of items 1 and 3-17, wherein the binding is binding to a conformational or discontinuous epitope on human GDF-15 that is comprised by the amino acid sequences of SEQ ID No; 25 and SEQ ID No: 26.
20.The antibody or antigen-binding portion thereof of any one of items 1-19 for the use according to any one of items 1-19, wherein the method for treating cancer is a method comprising inhibition of cancer growth.
21.The antibody or antigen-binding portion thereof of any one of items 1-20 for the use according to any one of items 1-20, wherein the method for treating cancer comprises the induction of killing of cancer cells by NK cells and CD8÷ T cells in the human patient.
22.The antibody or antigen-binding portion thereof of any one of items 1-21 for the use according to any one of items 1-21, wherein the heavy chain variable domain comprises a CR1 region comprising the amino acid sequence of SEQ ID NO; 3 and a CDR2 region comprising the amino acid sequence of SEQ ID NO: 4, and wherein the light chain variable domain comprises a CDR1 region comprising the amino acid sequence of SEQ ID NO; 6 and a CR2 region comprising the amino acid sequence ser-ala-ser.
23.The antibody or antigen-binding portion thereof of any one of items 1-22 for the use according to any one of items 1-22, wherein the constant domain of the heavy chain comprises the amino acid sequence of SEQ ID No: 29, or an amino acid sequence at least 85%, preferably at least 90%, more preferably at least 95% identical thereto, and wherein the constant domain of the light chain comprises the amino acid sequence of SEQ ID No: 32, or an amino acid sequence at least 85%, preferably at least 90%, more preferably at least 95% identical thereto.
24.The antibody or antigen-binding portion thereof of any one of items 1-23 for the use according to any one of items 1-23, wherein the constant domain of the heavy chain comprises the amino acid sequence of SEQ ID No: 29, or an amino acid sequence at least 98%, preferably at least 99% identical thereto, and wherein the constant domain of the light chain comprises the amino acid sequence of SEQ ID No: 32, or an amino acid sequence at least 98%, preferably at least 99% identical thereto.
25. The antibody or antigen-binding portion thereof of any one of items 1-24 for the use according to any one of items 1-24, wherein the constant domain of the heavy chain comprises the amino acid sequence of SEQ ID No: 29, and wherein the constant domain of the light chain comprises the amino acid sequence of SEQ ID No: 32.
26. The antibody or antigen-binding portion thereof of any one of items 1-25 for the use according to any one of items 1-25, wherein the antibody is a humanized antibody, and wherein all of the variable domains of the antibody are humanized variable domains.
27.The antibody or antigen-binding portion thereof of any one of items 1-26 for the use according to any one of items 1-26, wherein the heavy chain variable domain comprises the amino acid sequence of SEQ ID No: 28, or an amino acid sequence at least 90%, preferably at least 95%, more preferably at least 98, still more preferably at least 991 identical thereto, and wherein the light chain variable domain comprises the amino acid sequence of SEQ ID No: 31, or an amino acid sequence at least 90%, preferably at least 95%, more preferably at least 98%, still more preferably at least 991 identical thereto.
28.. The antibody or antigen-binding portion thereof of any one of items 1-27 for the use according to any one of items 1-27, wherein the heavy chain variable domain comprises the amino acid sequence of SEQ ID No: 28, and wherein the light chain variable domain comprises the amino acid sequence of SEQ ID No: 31.
29.The antibody or antigen-binding portion thereof of any one of items 1-28 for the use according to any one of items 1-28, wherein the heavy chain comprises the amino acid sequence of SEQ ID No: 27, and wherein the light chain comprises the amino acid sequence of SEQ ID No: 30.
30.The antibody or antigen-binding portion thereof of any one of items 1-25 for the use according to any one of items 1-25, wherein the heavy chain variable domain comprises the amino acid sequence of SEQ ID No: 34, or an amino acid sequence at least 751, more preferably at least 901, more preferably at least 95%, more preferably at least 981, still more preferably at least 99% identical thereto, and wherein the light chain variable domain comprises the amino acid sequence of SEQ ID No: 37, or an amino acid sequence at least 801, more preferably at least 90%, more preferably at least 95%, more preferably at least 98%, still more preferably at least 99% identical thereto.
31.The antibody or antigen-binding portion thereof of item for the use according to item 30, wherein the heavy chain variable domain comprises the amino acid sequence of SEQ ID No: 34, and wherein the light chain variable domain comprises the amino acid sequence of SEQ ID No: 37.
32. The antibody or antigen-binding portion thereof of any one of items 1-22 for the use according to any one of items 1-22, wherein the antibody is the antibody to human GDF-1S obtainable from the cell line Bl-23 deposited with the Deutsche Sammlung für Mikroorganismen und Zellkulturen GmbH (DMSZ) under the accession No. DSM ACC3142 or an antigen-binding portion thereof.
33.The antibody or antigen-binding portion thereof of any one of items 1-31 for the use according to any one of items 1-31, wherein the heavy chain variable domain comprises a CR3 region comprising the amino acid sequence of SEQ ID NO: 5, or wherein the light chain variable domain comprises a C]DR3 region comprising the amino acid sequence of SEQ ID NO: 7.
34.The antibody or antigen-binding portion thereof of any one of items 1-31 and 33 for the use according to any one of items 1-31 and 33, wherein the heavy chain variable domain comprises a CR3 region comprising the amino acid sequence of SEQ ID NO: 5, and wherein the light chain variable domain comprises a CDR3 region comprising the amino acid sequence of SEQ ID NO: 7.
35.The antibody or antigen-binding portion thereof of any one of items 1-25 and 33-34 for the use according to any one of items 1-25 and 33-34, wherein the heavy chain variable domain comprises a region comprising an FRi. a CR1, an FR2, a CDR2 and an FR3 region and comprising the amino acid sequence of SEQ ID NO: 1 or a sequence 95% identical thereto, and wherein the light chain variable domain comprises a region comprising an FR1, a CDR1, an FR2, a CDR2 and an FR3 region and comprising the amino acid sequence of SEQ ID NO: 2 or a sequence 95% identical thereto.
36.The antibody or antigen-binding portion thereof of any one of items 1-25 and 33-35 for the use according to any one of items 1-25 and 33-35, wherein the heavy chain variable domain comprises a region comprising an FR1, a CR1, an FR2, a CR2 and an FR3 region and comprising the amino acid sequence of SEQ ID NO: 1 or a sequence 98% identical thereto, and wherein the light chain variable domain comprises a region comprising an FR1, a CDR1, an FR2, a CDR2 and an FR3 region and comprising the amino acid sequence of SEQ ID NO: 2 or a sequence 98% identical thereto.
37.The antibody or antigen-binding portion thereof of any one of items 1-36 for the use according to any one of items 1-36, wherein the antibody or antigen-binding portion thereof has an equilibrium dissociation constant for human GDF-l5 that is equal to or less than nM, preferably less than 10 nM, more preferably less than 5 nM and most preferably between 0.1 nM and 2 nM.
38.The antibody or antigen-binding portion thereof of any one of items 1-31 and 33-37 for the use according to any one of items 1-31 and 33-37, wherein the antibody or antigen-binding portion thereof binds to the same human GDF-l5 epitope as the antibody to human GDF-lS obtainable from the cell line B1-23 deposited with the Deutsche Sammiung für Mikroorganismen und Zellkulturen GTUbH (DMSZ) under the accession No. DSM ACC3142.
39.The antibody or antigen-binding portion thereof of any one of items 1-38 for the use according to any one of items 1-38, wherein the human patient has elevated GDF-levels in blood serum before administration.
40..The antibody or antigen-binding portion thereof of any one of items 1-39 for the use according to any one of items 1-39, wherein the antibody or antigen-binding portion thereof is A) *the sole ingredient pharmaceutically active against cancer used in the method, or B)used in combination with one or more further ingredients pharmaceutically active against cancer.
4l.The antibody or antigen-binding portion thereof of any one of items 1-40 for the use according to any one of items 1-40, wherein the cancer is selected from the group consisting of brain cancers including glioma, cancers of the nervous system, melanoma, lung cancer, lip and oral cavity cancer, hepatic carcinoma, leukemia, Hodgkin lymphoma, Non-Hodgkin lymphoma, bladder cancer, cervix uteri cancer, corpus uteri cancer, testis cancer, thyroid cancer, kidney cancer, gallbladder cancer, multiple myeloma, nasopharynx cancer, larynx cancer, pharynx cancer, oesophagus cancer, gastrointestinal tumors including stomach and colorectal cancer, pancreatic cancer, prostate cancer, ovarian cancer and breast cancer, preferably from the group consisting of melanoma, prostate cancer, breast cancer, brain cancers including glioma, colorectal cancer, stomach cancer, oesophagus cancer and ovarian cancer, and most preferably is melanoma.
42.The antibody or antigen-binding portion thereof of any one of items 1-41 for the use according to any one of items 1-41, wherein prior to administration, the tumor or tumors formed by the cancer have higher human GDF-15 levels compared to a control sample of the same patient obtained from a non-cancerous part of the tissue which is the tissue of origin of the cancer, preferably 1.2-fold higher levels, more preferably 1.5-fold higher levels, still more preferably 2-fold higher levels and most preferably 5-fold higher levels.
43.The antibody or antigen-binding portion thereof of item for the use according to item 40, wherein the antibody or antigen-binding portion thereof is used in combination with one or more further ingredients pharmaceutical1' active against cancer, and wherein the one or more further ingredients pharmaceutically active against cancer are selected from the group consisting of: alkylating agents; anti-meta.bolites; alkaloids, taxanes; topoisomerase inhibitors; cytotoxic antibiotics; and radioisotopes.
44.The antibody or antigen-binding portion thereof of item 43 for the use according to item 43, wherein the one or more further ingredients pharmaceutically active against cancer are selected from the group consisting of: cisplatin, carboplatin, oxaliplatin, mechlorethamine, cyclophosphamide, chlorambucil, and ifosfamide; azathioprine and mercaptopurine; vincristine, vinblastine, vinorelbine, and vindesine, paclitaxel, docetaxel, etoposide and teniposide; irinotecan and topotecan; actinomycin, anthracyclines, doxorubicin, daunorubicin, vainthicin, idarubicin, epirubicin, bleomycin, plicamycin and mitomycin.
References ArbaJDi Ghahroudi M et al.: "Selection and identification of single domain antibody fragments from camel heavy-chain antibodies." FEES Lett. 1997 Sep 15;4l4(3):521-6.
Ausubel et al.: "current Protocols in Molecular Biology." Greene Publishing Associates and Wiley Interscience; New York 1992.
Bauskin AR et al..: "The propeptide mediates formation of stromal stores of PROMIC-1: role in determining prostate cancer outcome." cancer Res. 2005 Mar l5;65(6) :2330-6.
Brown DA et al.: "Macrophage inhibitory cytokine 1: a new prognostic marker in prostate cancer." din Cancer Res. 2009 Nov l;15(21):6658-64.
Chothia C et al.: Conformations of immunoglobulin hypervariable regions. Nature. 1989 Dec 21-28;342(6252) :877-Clackson T et al.: "Making antibody fragments using phage display libraries." Nature. 1991 Aug 15;352 (6336) :624-8.
Fearon KC.: Cancer cachexia: developing multimodal therapy for a multidimensional problem. Eur j cancer. 2008 May;44(8) :1124-32 Fearon K. et al.: Definition and classification of cancer cachexia: an international consensus. Lancet Oncol. 2011 May; 12(5) :489-95.
Giudicelli V et al.: IMGT/V-QtJEST, an integrated software program for immunoglobulin and T cell receptor V-J and V-D-J rearrangement analysis. Nucleic Acids Res. 2004 Jul l;32(Web Server issue) :W435-40.
Harlow and Lane: "Antibodies: A Laboratory Manual" Cold Spring Harbor Laboratory Press, Cold Spring Harbor, New York 1988.
Holliger P et al.: "Diabodies": small bivalent and bispecific antibody fragments." Proc Nati Acad Sd U S A. 1993 Jul l5;90(14):6444-8.
Halt U et al.: "Domain antibodies: proteins for therapy." Trends Biotecimol. 2003 Nov;2l(1]j:484-90.
Huang a et al.: "Molecular alterations in prostate carcinomas that associate with in vivo exposure to chemotherapy: identification of a cytoprotective mechanism involving growth differentiation factor 15." din Cancer Res. 2007 Oct 1;13(l9) :5825-33.
Johnen H et al.,: "Tumor-induced anorexia and weight loss are mediated by the TGF-beta superfamily cytokine MIC-l." Nat Med. 2007 Nov;13(ll) :1333-40.
Jones PT et al.: "Replacing the complementarity-determining regions in a human antibody with those from a mouse." Nature.
1986 May 29-Jun 4;32l(6069):522-5.
Ka.bat et al.: Sequences of proteins of immunological interest, U.S. Dept. of Health and Human Services, Public Health Service, National Institutes of Health, Bethesda, Md. 1983.
Kâhler C and Milstein C: "Continuous cultures of fused cells secreting antibody of predefined specificity." Nature. 1975 Aug 7;256(5517) :495-7.
Marks 3D et al.: "By-passing immunization. Human antibodies from V-gene libraries displayed on phage." 3 Mol Biol. 1991 Dec 5;222(3):581-97.
Mimeault M and Batra 3K: "Divergent molecular mechanisms underlying the pleiotropic functions of macrophage inhibitory cytokine-1 in cancer." 3 Cell Physiol. 2010 Sep;224(3) ;626-35.
Murphy KT and Lynch GS: Update on emerging drugs for cancer cachexia. Expert Opin Emerg Drugs. 2009 Dec;14(4) :619-32.
Paul, W.E. (Ed.).: "Fundamental Immunology" 2nd Ed. Raven Press, Ltd., New York 1989.
Remington's Pharmaceutical Sciences, Ed. .ZR Gennaro, 20th edition, 2000, williams & Wilkins, PA, USA.
Riechmann L et al.: "Reshaping human antibodies for therapy." Nature. 1988 Mar 24;332(6162):323-7.
Roth P et al.: "GDF-15 contributes to proliferation and immune escape of malignant gliomas." din Cancer Res. 2010 Aug l;16(15):3851-9.
Saerens D et al.: "Single-domain antibodies as building blocks for novel therapeutics." Curr Opin Pharmacol. 2008 Oct;8(S):600-8. Epub 2008 Aug 22.
Sambrook et al.: "Molecular Cloning: A Laboratory Manual.", 2nd Ed., Cold Spring Harbor Laboratory Press, Cold Spring Harbor, New York 1989.
Siegel DL: "Recombinant monoclonal antibody technology." Transfus din Biol. 2002 Jan;9(l) :15-22.
Stefanescu IL et al., Eur.J.Mass Spectrom. 13, 69-75 (2007) Suckau et al. Proc Nati Acad Sd U S A. 1990 December; 87(24): 9848-9852.
Tisdale MJ.: Mechanisms of cancer cachexia. Physiol Rev. 2009 Apr;89(2) :381-410.
Weinberg It. et al..: The Biology of Cancer. Garland Science: New York 2006. 850p.
SEQUENCE LISTING
<110> Juiius-Maximiiians-UniversitAt wUrzburg <120> Monoclonal Antibodies to Growth and Differentiation Factor 15 (GDF-15), and uses thereof for treating cancer cachexia <130> 172099 <160> 38 <170> Patentln version 3.5 <210> 1 <211> 97 <212> PRT <213> Mus musculus <400> 1 Gin Va] Lys Leu Gin Gin ser Giy Pro Giy lie Leu Gin ser ser Gin 1 5 10 15 Thr Lea Ser Leu Thr cys ser Phe Ser Gly Phe Ser Leu Ser Thr ser 25 30 Giy Met Giy vai ser lie A9 Gin pro Ser Giy Lys Giy Leu Glu 40 45 Lea Ala His lie Tyr Asp Asp Asp Lys Arg Tyr Asn Pro Thr 55 60 Leu Lys Ser Arg Leu Thr lie Ser Lys ASP Pro Ser Arg Asn Gin val 70 75 80 Phe Lea Lys lie Thr ser vai Asp Thr Ala Asp Thr Ala Thr Tyr Tyr 90 95 cys <210> 2 <211> 88 <212> PRT <213> MUS muscuius <400> 2 Asp lie Val Lea Thr Gin ser Pro LyS Phe Met Ser Thr ser val Gly 1 5 10 15 Asp A9 val ser val Thr cys Lys Aia ser Gin Asn vai Giy Thr Asn 25 30 Va] Ala Trp Phe LeU Gln LyS Pro Giy Gin ser Pro Lys Aia Leu Ile 40 45 Tyr Ser Ala ser Tyr Arg Tyr Ser Gly Va] Pro Asp Arg Phe Thr Gly 55 60 Ser Gly Ser Giy Thr Asp Phe Thr Leu Thr lie Ser Asn Va] Gin ser 70 75 80 Giu Asp Leu Ala Giu Tyr Phe Cys <210> 3 <211> 10 <212> PRT <213> Mus musculus <400> 3 Gly Phe Ser Leu Ser Thr Ser Gly Met Gly 1 5 10 <210> 4 <211> 7 <212> PRT <213> Mus muscuius <400> 4 lie TY Asp Asp Asp Lys 1 5 <210> 5 <211> 10 <212> PRT <213> Mus musculus <400> 5 Ala Arg Ser Ser Tyr Gly Ala Met Asp TY 1 5 10 <210> 6 <211> 6 <212> PRT <213> MUS musculus <400> 6 Gin Asn Val Gly Thr Asn 1 5 <210> 7 <211> 9 <212> PRT <213> Mus muscuius <400> 7 Gin Gin Tyr Asn Asn Phe Pro Tyr Thr <210> 8 <211> 114 <212> PRT <213> Artificial sequence <220> <223> recombinant mature human GDF-15 protein <400> 8 Gly ser Ala Arg Asn Gly Asp His Cys Pro Leu Gly Pro dy Arg cys 1 5 10 15 cys Arq Leu His Thr Vai Arg Aia Ser Leu Glu Asp Leu Gly Ala 25 30 Asp Va] Lea Ser Pro A9 Glu Va] Gin vaffiiiiiet Cys lie Gly 40 45 Ala CS Pro Ser Clii Ptie Arg Ala Ala Asn Met His Ala Girt lie Lys 55 60 Thr ser Leu His A9 Leu Lys Pro Asp Thr vai Pro Ala Pro Cys Cys 70 75 80 val Pro Ala ser Tyr Asn Pro Met Val Lea Ile Gin Lys Thr Asp Thr 90 95 Gly Val Ser Lea Gin Thr Tyr Asp Asp Leu Leu Ala Lys Asp Cys His 105 110 cys Lie <210> 9 <211> 308 <212> PRT <213> Homo sapiens <400> 9 Met Pro dy Gin Glu Leu Arg Thr val Asn dy ser Gin Met Lea Leu 1 5 10 15 vai Lea LeU vai Leu Ser Trp Lea Pro His Gly Gly Ala Lea ser Lea 25 30 Ala Giu Ala Ser Arg Ala Ser Phe Pro Giy Pro Ser Glu Leu His Ser 40 45 Giu Asp Ser Arg Plie Arg Giu Lea Arg Lys Arg Tyr cia Asp Lea Leu 55 60 Thr Arg Leu Arg Ala Asn Gin Ser Trp Glu Asp Ser Asn Thr Asp Leu 70 75 80 val Pro Ala Pro Aia Val Arg lie Leu Thr Pro Glu Val Arg Leu ciy 90 95 Ser dy Gly His Leu His Lea Arg lie ser Arg Ala Ala Lea Pro Glu 105 110 dy Lea Pro cia Ala Ser Arg Leu His Arg Ala Leu Phe Arg Leu ser 120 125 Pro Thr Ala Ser Arg Ser Asp val Thr Arg Pro Lea Arg Arg Gln 135 140 Lea Ser Leu Ala Arg Pro Gln Ala Pro Ala Leu His Leu Arg Leu Ser 150 155 160 Pro Pro Pro ser Gln Ser Asp Gin Leu Leu Ala Glu Ser Ser Ser Ala 170 175 Arg Pro Gln Lea Glu Leu His Lea A9 Pro Gin Ala Ala A9 dy Arg 185 190 Arg Arg Ala Arg Ala Arg Asn dy Asp His cys Pro Leu Gly Pro Gly 200 205 Arg cys cys Arg Leu His Thr Val A9 Ala Ser Leu Glu Asp Leu Gly 210 215 220 Trp Ala sp Trp val Leu ser Pro A9 Glu Val Gin Val Thr Met Cys 225 230 235 240 tie Gly Ala cys Pro Ser Gin Phe A9 Ala Ala Asn Met His Ala Gln 245 250 255 lie Lys Thr Ser Leu His Arg La Lys Pro Asp Thr Val Pro Ala Pro 260 265 270 cys cys vai Pro Ala Ser Tyr Asn Pro Met val Leu Tie Gin Lys Thr 275 280 285 Asp Thr Giy val Ser Leu Gin Thr Tyr Asp Asp Leu Leu Ala LS Asp 290 295 300 Cys His cys Ile <210> 10 <211> 322 <212> PRT <213> Artificial sequence <220> <223> human GDF-15 precursor protein + N-terminal and c-terminal GSGS linker <400> 10 Gly Ser Gly Ser Gly Ser Gly Met Pro Gly Gln Glu Leu Arg Thr Val 1 5 10 15 Asn dy ser Gln Met Leu LCU Val Leu Leu Val Leu ser Trp Leu Pro 25 30 His Gly Gly Ala Leu Ser Leu Ala Glu Ala Ser Arg Ala Ser Phe Pro 40 45 Gly pro Ser Glu Leu His ser Glu Asp Ser Arg Phe Arg Glu Leu Arg 55 60 Lys Arg Tyr Glu Asp Leu Leu Thr Arg Leu Arg Ala Asn Gln Ser Trp 70 75 80 Glu Asp Ser Asn Thr Asp Leu val Pro Ala Pro Ala Val Ary lie Leu 90 95 Thr Pro Glu val Arg Leu Gly ser dy Gly His Leu His Leu Arg Ile 105 110 ser A9 Ala Ala Leu Pro Glu dy Leu Pro Glu Ala Ser Arg Leu His 120 125 Arg Ala Leu Phe Arg Leu Ser Pro Thr Ala ser Arg ser Asp val 135 140 Thr Arg Pro Leu Arg Arg Gin Leu Ser Leu Ala A9 Pro Gin Ala Pro 150 155 160 Ala Leu His La Arg Leu ser Pro Pro Pro Ser Gin ser Asp Gin Leu 170 175 Leu Ala Glu Ser Ser Ser Ala Arg Pro Gin Leu Glu Leu His Leu Arg 185 190 Pro Gin Ala Ala A9 Gly Arg Arg Arg Ala A9 Ala Ary Asn Gly Asp 200 205 His cys Pro Leu Gly Pro Gly Arg cys Cys Arg Leu His Thr Val Arg 210 215 220 Ala ser Leu Giu Asp Leu Gly Ala Asp Val Leu ser Pro Ar 225 230 235 24 Glu Vai Gin Val Thr Met cys lie Gly Ala cys Pro ser Gin Phe Arg 245 250 255 Ala Ala Asfl Met His Ala Gln Ile Lys Thr ser Leu His Arg Leu Lys 260 265 270 Pro Asp Thr Val Pro Ala Pro Cys Cys Val Pro Ala Ser Tyr Asn Pro 275 280 285 Met Val Leu ile cln Lys Thr Asp Thr Gly Val ser Leu Gin Thr Tyr 290 295 300 Asp Asp Leu Leu Ala Lys Asp Cys His Cys tie Gly Ser Giy Ser Gly 305 310 315 320 ser Gly <210> 11 <211> 10 <212> PRT <213> Artificial sequence <220> <223> Flag peptide <400> 11 Asp Tyr Lys Asp Asp Asp Asp Lys Gly Gly 1 5 10 <210> 12 <211> 10 <212> PRT <213> Artificial sequence <220> <223> HA peptide <400> 12 Tyr Pro Tyr Asp Va] Pro Asp Tyr Ala Gly 1 5 10 <210> 13 <211> 13 <212> PRT <213> Artificial Sequence <220> <223> peptide derived from human GDF-15 <400> 13 Glu Leu His LeU Arg Pro Gin Ala Ala Arg Gly Arg Arg 1 5 10 <210> 14 <211> 13 <212> PRT <213> Artificial sequence <220> <223> peptide derived from human GDF-15 <400> 14 Leu His Leu Arg Pro Gln Ala Ala Arg Gly Arg Arg Arg 1 5 10 <210> 15 <211> 13 <212> PRT <213> Artificial sequence <220> <223> peptide derived from human GDF-15 <400> 15 His Leu A9 Pro Gln Ala Ala Arg dy Arg Arg Arg Ala 1 5 10 <210> 16 <211> 13 <212> PRT <213> Artificial sequence <220> <223> peptide derived from human GDF-15 <400> 16 Leu Ary Pro Gin Ala Ala A9 Gly Arg Arg Arg Ala Ary 1 5 10 <210> 17 <211> 13 <212> PRT <213> Artificial sequence <220> <223> peptide derived from human GDF-15 <400> 17 Arg Pro Gin Ala Ala Arg Gly Arg Ary A9 Ala Arg Ala 1 5 10 <210> 18 <211> 13 <212> PRT <213> Artificial sequence <220> <223> peptide derived from human GDF-15 <400> 18 Pro Gln Ala Ala Arg dy Arg Arg Arg Ala A9 Ala Arg 1 5 10 <210> 19 <211> 13 <212> PRT <213> Artificial sequence <220> <223> peptide derived from human GDF-15 <400> 19 Gln Ala Ala Arg Gly Arg Arg Arg Ala Arg Ala Arg Asn 1 5 10 <210> 20 <211> 13 <212> PRI <213> Artificial sequence <220> <223> peptide derived from human GDF-15 <400> 20 Met His Ala Gin Ile Lys Thr Ser Leu His A9 Leu Lys 1 5 10 <210> 21 <211> 291 <212> DNA <213> Mus musculus <400> 21 caagtgaagc tgcagcagtc aggccctggg atattgcagt cctcccagac cctcagtctg 60 acttgttctt tctctgggtt ttcactgagt acttctggta tgggtgtgag ctggattcgt 120 cagccttcay gaaagggtct ggagtggctg gcacacattt actgggatga tgacaagcgc 180 tataacccaa ccctgaagag ccggctcaca atctccaagg atccctccag aaaccaggta 240 ttcctcaaga tcaccagtgt ggacactgca gatactgcca catactactg t 291 <210> 22 <211> 264 <212> DNA <213> Mus musculus <400> 22 gacattgtgc tcacccagtc -tccaaaattc atgtccacat cagtaggaga cagggtcagc 60 gtcacctgca aggccagtca gaatgtgggt actaatgtgg cctggtttct acagaaacca 120 gggcaatctc ctaaagcact tatttactcg gca-tcctacc ggtacagtgg agtccctgat 180 cgcttcacag gcagtggatc tgggacagat ttcactctca ccatcagcaa cgtgcagtct 240 gaagacttgg cagagtattt ctgt 264 <210> 23 <211> 30 <212> DNA <213> Mus musculus <400> 23 gctcgaagtt cctacgggyc aatggactac 30 <210> 24 <211> 27 <212> DNA <213> Mus musculus <400> 24 caqcaatata acaactttcc gtacacg 27 <210> 25 <211> 16 <212> PRT <213> Homo sapiens <400> 25 Glu val Gin Val Thr Met Cys lie Gly Ala Cys Pro ser Gin Phe Arg 1 5 10 15 <210> 26 <211> 21 <212> PRT <213> Homo sapiens <400> 26 Thr Asp Thr Gly Mi 5cr Leu Gin Thr TYr Asp Asp Leu Lea Ala Lys 1 5 10 15 Asp Cys His CYS lie <210> 27 <211> 448 <212> PRT <213> Artificial Sequence <220> <223> amino acid sequence of the heavy chain of the H1LS humanized B1-23 anti-GDF-15 antibody <400> 27 Gin tie Thr Leu Lys Giu Ser dy Pro Thr Leu val Lys Pro Thr Gin 1 5 10 15 Thr Lea Thr Leu Thr cys Thr I'he ser Gly Phe ser Leu ser Thr ser 25 30 Gly Met Gly Mi ser T' lie Arg Gin Pro Pro Giy Lys Gly Leu Glu 40 45 Trp Leu Ala His lie Tyr TP Asp A5 AS Lys Arg Tyr Asn Pro Thr 55 60 Leu Lys Ser Arg Leu Thr lie Thr LyS Asp Pro ser Lys Asfl Gin vai 70 75 80 Mi Leu Thr Met Thr ASn Met Asp Pro vai Asp Thr Ala Thr Tyr Tyr 90 95 Cys Ala A9 ser Ser Tyr dy Ala Met Asp Tyr Trp Gly Gln Gly Thr 105 110 Leu Val Thr val ser Ser Ala ser Thr Lys Gly Pro ser Va) Pile Pro 120 125 Leti Ala Pro Ser Ser Lys Ser Thr ser dy dy Thr Ala Ala Leu dy 135 140 Cys Leu Val LyS Asp Tyr Pile Pro Glu Pro Val TIlr Va) Ser Trp Asn 150 155 160 Ser dy Ala Leu Thr ser dy Va) His Thr Pile Pro Ala Val Leu Gin 170 175 Ser Ser Gly Leu Tyr Ser Leu Ser Ser Va) Val Thr vai Pro ser Ser 185 190 Ser Leu Gly Thr Gln Thr Tyr lie Cys Asn Val Asn His Lys Pro Ser 200 205 Asn Thr Lys Val Asp Lys Lys Val Glu Pro Lys ser CyS Asp LyS Thr 210 215 220 His Thr Cys Pro Pro Cys Pro Ala Pro Glu Leu Leu Gly Gly pro ser 225 230 235 240 val Phe Leu Phe Pro Pro Lys Pro Lys Asp Thr Leu Met Ile Ser Arg 245 250 255 Thr pro Glu Va) Thr cys Va) vai Val Asp val ser His Glu Asp Pro 260 265 270 Glu Val Lys Phe Asn Trp Tyr vai Asp Gly val Glu Val His Asn Aia 275 280 285 Lys Tflr Lys Pro Ary Glu Glu Gin Tyr Asn Ser Thr Tyr Arg Val Va) 290 295 300 ser Va) Leu Thr val Leu His Gin Asp Leu Asn Gly Lys Glu Tyr 305 310 315 320 Lys Cys Lys Va) Ser Asn LS Ala Leu Pro Ala Pro lie Glu Lys Thr 325 330 335 Ile ser Lys Ala ys dy Gin Pro Arg Q)u Pro Gln Va) Tyr Thr Leu 340 345 350 Pro Pro Ser Arg Glu Glu Met Thr Lys Asn Gln val Ser Leu Thr Cys 355 360 365 Leu Va) L5 Gly Phe Tyr Pro ser Asp Ile Ala val Glu Trp Giu Ser 370 375 380 Mn Gly Gln Pro Glu Asn Asn Tyr Lys Thr Thr Pro Pro Val Leu Asp 385 390 395 400 Ser Asp dy ser Phe Phe Leu Tyr Ser Lys Leu Thr val Asp Lys Ser 405 410 415 Arg Gin Gin Giy Asn Va] Phe ser Cys ser Va] Met His Glu Ala 420 425 430 Leu His Asn His TY Thr Gin Lys ser Leu ser Leu Ser Pro Gly Lys 435 440 445 <210> 28 <211> 118 <212> PRT <213> Artificial Sequence <220> <223> amino acid sequence of the heavy chain variable domain of the H1LS humanized B1-23 anti-dDE-iS antibody <400> 28 Gin lie Thr Leu Lys Glu Ser dy Pro Thr Leu vai Lys Pro Thr Gin 1 5 10 15 Thr Leu Thr Leu Thr cys Thr Phe ser Giy Phe Ser Leu Ser Thr Ser 25 30 Gly Met Gly vai Ser lie A9 Gin Pro Pro Gly Lys dy eu G]u 40 45 Trp Leu Ala His Ile Tyr Tt'p Asp ASP Asp LyS Arg Tyr Asn Pro Thr 55 60 Leu Lys ser Arg Leu Thr lie Thr Lys Asp pro ser Lys ASn Gin vai 70 75 80 val LeU Thr Met Thr Asn Met Asp Pro Va] Asp Thr Ala Thr Tyr Tyr 90 95 Cys Ala A9 ser Ser Tyr Gly Ala Met Asp Tyr Trp dy Gin Gly Thr 105 110 Leu Va] Thr val ser ser <210> 29 <211> 330 <212> PRT <213> Artificial sequence <220> <223> amino acid sequence of the heavy chain constant domain of the H1LS humanized 81-23 anti-GDF-15 antibody <400> 29 Ala Ser Thr Lys Gly Pro ser val Phe Pro Leu Ala Pro ser ser Lys 1 5 10 15 Ser Thr ser Gly Gly Thr Ala Ala Leu Gly cys Leu Vai Lys Asp Tyr 25 30 Phe Pro Glu Pro val Thr Val Ser Trp nfl ser Giy Ala Leu Thr ser 40 45 Gly Val His Thr Phe Pro Ala val Leu Gin Ser Ser Gly Leu Tyr Ser 55 60 Leu Ser ser val val Thr Val Pro Ser Ser Ser Leu dy Thr Gin Thr 70 75 80 Tyr tie cys nn val ASn His Lys Pro Ser Asn Thr Lys val Asp Lys 90 95 Lys Val 6Th Pro Lys Ser CyS Asp Lys Thr His Thr CyS Pro Pro Cys 105 110 Pro Ala Pro Glu Leu Leu dy dy Pro Ser Vai Phe Leu Phe Pro Pro 120 125 Lys Pro Lys Asp Thr Leu Met lie ser Arg Thr Pro Glu val Thr Cys 135 140 vaiVal val Asp vai ser His Giu Asp Pro Glu Val Lys Phe Asn Trp 150 155 160 Tyr Vai Asp Gly Vai Giu val His Asn Ala Lys itir Lys Pro Arg Glu 170 175 Glu Gin Tyr Asn Ser Thr Tyr Arg val vai ser val Leu Thr Val Leu 185 190 His Gin Asp Trp Leu Asn Gly Lys Glu Tyr Lys Cys Lys Val Ser Asn 200 205 Lys Ala Leu Pro Ala Pro lie Glu Lys Thr lie Ser Lys Ala Lys dy 210 215 220 Gin Pro Arg Glu Pro Gin Val Tyr Thr Leu Pro Pro Ser Arg Giu Glu 225 230 235 240 Met Thr Lys Asn Gin val Ser Leu Thr Cys Leu val Lys Gly Phe Tyr 245 250 255 Pro ser Asp lie Ala val Glu Trp Giu ser Asn Gly Gin Pro Giu Asn 260 265 270 Asn Tyr Lys Thr Thr Pro Pro vai Leu Asp ser Asp Gly Ser Phe Phe 275 280 285 Leu Tyr ser Lys Leu Thr val AS Lys ser Arg rp Gin Gin Gly Asri 290 295 300 Vai Phe ser cys ser val Met His Giu Ala Leu His Asn His Tyr Thr 305 310 315 320 Gin Lys ser LeU ser Leu ser Pro Giy Lys 325 330 <210> 30 <211> 211 <212> PRT <213> Artificial sequence <220> <223> amino acid sequence of the light chain of the H1L5 humanized B1-23 anti -GDF-15 antibody <400> 30 Asp lie val Leu Thr Gin Ser Pro Ser Phe Leu ser Ala ser Val Giy 1 5 10 15 Asp Arg val Thr lie Thr Cys Lys Ala ser Gin Asn val dy Thr Asn 25 30 Vai Ala Phe Gin Gin LyS Pro Giy Lys Ser Pro Lys Ala Leu Ile 40 45 Tyr Ser Ala Ser Tyr Arg Tyr ser dy val Pro Asp Arg Phe Thr Giy 55 60 Ser Gly ser dy Thr Glu Phe Thr Leu Thr lie ser ser Leu Gln Pro 70 75 80 Giu Asp Phe Ala Ala Tyr Phe Cys Gin Gin TY Asn Asn Phe Pro Tyr 90 95 Thr Rue Giy Giy Giy Thr LyS Leu Giu lie LS Arg Ala Pro Ser val 105 110 Phe Ile Phe Pro Pro ser Asp Glu Gin Leu Lys ser Gly Thr Ala ser 120 125 Val Val Cys Leu Leu Asn Asn Phe TY Pro Arg Glu Ala Lys Val Gin 135 140 Trp Lys Val Asp Asn Ala Leu Gln Ser Giy Asn Ser Gin Giu Ser Val 150 155 160 Thr Glu Gln Asp ser LS Asp Ser Thr Tyr ser Leu Ser Ser Thr Leu 170 175 Thr Leu Ser Lys Ala Asp Tyr Glu Lys His Lys Vai Tyr Ala Cys Glu 185 190 Val Thr His Gin Giy Leu Ser ser Pro Val Thr Lys ser Phe Asn Arg 200 205 Giy Giu cys <210> 31 <211> 108 <212> PRT <213> Artificial sequence <220> <223> amino acid sequence of the light chain variable domain of the H1LS humanized B1-23 anti-GDF-15 antibody <400> 31 Asp lie Val Leu Thr Gin Ser Pro Ser Phe Leu Ser Ala ser val Gly 1 5 10 15 Asp Arg Val Thr tie Thr Cys Lys Ala Ser Gin Asn Val Gly Thr Asn 25 30 val Ala Trp Phe Gin Gin Lys Pro Gly Lys Ser Pro Lys Aia Leu lie 40 45 Tyr ser Ala Ser Tyr Arg Tyr ser Gly Val Pro ASP Arg Phe Thr Giy 55 60 ser dy Ser dy Thr Glu Phe Thr Leu Thr lie ser Ser Leu Gin Pro 70 75 80 Glu Asp Phe Ala Ala TY Phe Cys Gin Gin TY Asn Asn Phe Pro Tyr 90 95 ihr Phe Gly Gly Gly Thr Lys Leu Glu lie ys Arg 105 <210> 32 <211> 103 <212> PRT <213> Artificial sequence <220> <223> amino acid sequence of the light chain constant domain of the H1L5 humanized B1-23 anti-GDF-15 antibody <400> 32 Ala ro ser val Phe Tie Phe Pro Pro ser ASP Glu Gin Leu Lys Ser 1 5 10 15 Giy Thr Ala Ser val Val cys Leu Leu Asn Asn Phe Tyr Pro Arg Giu 25 30 Ala Lys val Gln Trp Lys Val Asp Asn Ala Leu Gin ser Giy Asn Ser 40 45 Gin Giu Ser val Thr Glu Gin sp Ser LS Asp Ser Thr Tyr Ser Leu 55 60 ser Ser Thr Leu Thr Leu Ser Lys Ala Asp Tyr Glu L5 His Lys val 70 75 80 Tyr Ala cys Glu val Thr His Gin dy eu Ser Ser Pro Val Thr Lys 90 95 Ser Plie Asn Arg Gly Glu Cys <210> 33 <211> 448 <212> PRT <213> Artificial sequence <220> <223> amino acid sequence of the heavy chain of the chimeric 61-23 anti -GDF-15 anti body <400> 33 Gin val Lys Leu Gin Gin Ser dy Pro Gly lie Leu Gin Ser Ser Gin 1 5 10 15 Thr Leu Ser Leu Thr cys Ser Phe ser Giy Phe Ser Leu Ser Thr Ser 25 30 Giy Met dy Vai Ser Trp lie Arg Gin Pro ser dy Lys dy Leu Glu 40 45 Trp Leu Ala His lie Tyr Trp Asp Asp Asp Lys Arg Tyr Asn Pro Thr 55 60 Leu Lys Ser Arg Leu Thr lie 5cr LyS Asp Pro Ser Arg Asn Gin val 70 75 80 Phe Leu Lys Ile Thr Ser Val Asp Thr Aia Asp Thr Ala Thr Tyr Tyr 90 95 cys Ala Ary ser ser Tyr Gly Ala Met Asp Tyr Trp Giy Gin Giy Thr 105 110 Ser val Thr Val Ser Ser Ala Ser Thr Lys dy Pro Ser val Phe Pro 120 125 Leu Ala Pro Ser Ser Lys Ser Thr Ser Gly Giy Thr Aia Ala Leu dy 135 140 Cys Leu Val Lys Asp TY Phe Pro Giu Pro val Thr val Ser Trp Asn 150 155 160 Ser Giy Ala Leu Thr Ser Gly val His Thr Phe Pro Ala val Leu Gln 170 175 ser Ser Gly Leu Tyr ser Leu ser ser val val Thr Val Pro Ser Ser 185 190 ser Leu Gly Thr Gln Thr Tyr lie cys Asn val Asn His Lys Pro Ser 200 205 Asn Thr Lys val Asp Lys Lys Val Glu Pro L5 Ser cys Asp ys Thr 210 215 220 His Thr Cys Pro Pro Cys Pro Ala Pro Glu Leu Leu Gly Gly Pro ser 225 230 235 240 Val Phe Leu Phe Pro Pro Lys Pro Lys Asp Thr Leu Met lie Ser Arg 245 250 255 Thr pro Glu val Thr cys Val vai Val Asp vai ser HIS Giu A5 ro 260 265 270 Giu val Lys Plie Asn Trp Tyr val Asp Giy Va] Giu Va] His Asn Ala 275 280 285 Lys Thr Lys Pro Ary Giu Glu Gin Tyr Asn ser Thr Tyr Arg Va] val 290 295 300 Ser vai Leu Thr Val Leu HIS Gin Asp Trp Leu Asn Giy Lys Giu Tyr 305 310 315 320 Lys cys Lys Val Ser Asn Lys Ala Leu Pro Ala Pro lie Glu Lys Thr 325 330 335 Ile Ser Lys Aia Lys dy Gin Pro Arg Giu Pro Gin Vai Tyr Thr Leu 340 345 350 Pro Pro Ser Arg Glu Glu Met Thr Lys Asn Gin vai Ser Leu Thr Cys 355 360 365 Leu val Lys Gly Phe Tyr Pro Ser Asp Tie Ala vai Glu Trp Glu Ser 370 375 380 Asn Gly Gin Pro Glu ASn Asn Tyr Lys Thr Thr Pro Pro Val Leu Asp 385 390 395 400 Ser Asp ciy ser Phe Phe Leu Tyr ser Lys Leu Thr Val Asp Lys Ser 405 410 415 Arg Trp Gin Gin dy Asn vai Phe ser Cys Ser Val Met His Giu Ala 420 425 430 Leu His ASn His Tyr Thr Gin Lys Ser Leu Ser Leu Ser Pro Giy Lys 435 440 445 <210> 34 <211> 118 <212> PRT <213> Artificial sequence <220> <223> amino acid sequence of the heavy chain variable domain of the chimeric B1-23 anti-GDF--15 antibody <400> 34 Gln Vai Lys Leu Gin Gln Ser Giy Pro Giy ile Leu Gin Ser ser Gln 1 5 10 15 Thr Leu ser LeU Thr cys Ser Phe Ser Giy Phe Ser Leu Ser Thr Ser 25 30 Gly Met Giy Vai Ser Trp lie Arg Gln Pro Ser ciy Lys ciy Leu Glu 40 45 Trp Leu Aia His Ile Tyr Trp Asp sp Asp ys Arg Tyr Asn Pro Thr 55 60 Leu Lys ser Arg Leu Thr lie Ser LyS Asp Pro Ser Arg Asn Gin val 70 75 80 Phe Leu LS lie Thr Ser Vai Asp Thr Aia AS Thr Aia Thr Tyr Tyr 90 95 cys Ala Arg ser ser Tyr Gly Ala Met Asp Tyr Trp dy Gin Gly Thr 105 110 ser s/al Thr val ser ser <210> 35 <211> 330 <212> PRT <213> Artificial sequence <220> <223> amino acid sequence of the heavy chain constant domain of the chimeric 61-23 anti-GDF-15 antibody <400> 35 Ala ser Thr Lys dy pro ser s/al Phe Pro Leu Ala Pro Ser ser Lys 1 5 10 15 Ser Thr Ser Gly Gly Thr Ala Ala Leu Gly Cys Leu s/al Lys Asp Tyr 25 30 Phe Pro Glu Pro val Thr s/al Ser Trp Asn Ser Gly Ala Leu Thr Ser 40 45 Gly val His Thr Phe Pro Ala s/al Leu Gln ser Ser Gly Leu Tyr ser 55 60 Leu ser ser s/al s/al Thr s/al Pro Ser Ser Ser Leu Gly Thr Gin Thr 70 75 80 TY lie Cys Asn Val Asn His Lys Pro Ser Asn Thr Lys s/al Asp Lys 90 95 Lys val Glu Pro LS ser cys Asp Lys Thr His Thr cys Pro Pro Cys 105 110 Pro Ala Pro Glu Leu Leu Gly Gly Pro ser s/al Phe Leu Phe Pro Pro 120 125 Lys Pro Lys Asp Thr Leu Met lie Ser Arg Thr Pro Glu s/al Thr cys 135 140 s/al s/al val Asp val Ser His Glu Asp Pro Glu val Lys Phe Asn Trp 150 155 160 Tyr s/al Asp Gly Val Glu val His Asn Ala Lys Thr Lys Pro Arg Glu 170 175 Glu Gin TY Asn ser Thr Tyr Arg s/al s/al Ser val Leu Thr val Leu 185 190 His Gin Asp Leu Asn Giy Lys Glu Tyr Lys Cys Lys Val ser Asn 200 205 Lys Ala Leu Pro Ala Pro lie Glu Lys Thr lie Ser Lys Ala Lys Gly 210 215 220 Gin Pro Arg Glu Pro Gin vai Tyr Thr Leu Pro Pro Ser Arg Giu Glu 225 230 235 240 Met Thr Lys Asn Gin val Ser Leu Thr Cys Leu Vai Lys Giy Phe Tyr 245 250 255 Pro ser Asp Tie Ala vai Glu Trp Glu Ser Asn Gly Gin Pro Giu Asn 260 265 270 Asn Tyr Lys Thr Thr Pro Pro Val Lea Asp Ser Asp Gly Ser Phe Plie 275 280 285 Leu Tyr Ser Lys Leu Thr vai Asp Lys ser Arg Gin Gin Giy Asn 290 295 300 Val Phe Ser cys Ser val Met His Glu Ala Leu His Asn His Tyr Thr 305 310 315 320 Gin Lys Ser Leu ser Leu Ser Pro dy Lys 325 330 <210> 36 <211> 214 <212> PRT <213> Artificiai sequence <220> <223> amino acid sequence of the light chain of the chimeric B1-23 anti-dEW-iS antibody <400> 36 Asp lie Val Leu Thr Gin ser Pro LS Phe Met Ser Thr ser vai Giy 1 5 10 15 ASP A9 Vai Ser val Thr Cys Lys Ala ser Gin Asn vai Giy Thr ASfl 25 30 vai Ala Trp Phe Leu Gin Lys Pro Gly Gin Ser Pro Lys Ala Leu lie 40 4S Tyr ser Aia ser Tyr Arg Tyr Ser dy Vai Pro Asp Arg Phe Thr Giy 55 60 ser dy Ser Gly Thr Asp Phe Thr Leu Thr lie ser Asn Vai Gin Ser 70 75 80 Glu Asp Leu Ala Giu Tyr Phe C5 Gin Gin TY Asn Asn Phe Pro Tyr 90 95 Thr Phe dy Giy GiyThr Lys Lea Giu lie L5 Arg Thr vai AiaAia 105 110 Pro Ser Vai Phe Ile Phe Pro Pro Ser Asp Giu Gin Lea Lys Ser Giy 120 125 ihr Aia Ser val vai Cys Leu Lea Asn Asn Phe Tyr Pro Arg Giu Ala 135 140 Lys Vai Gin Trp Lys Val Asp Asn Ala Lea Gin Ser Gly Asn ser Gin 150 155 160 * Glu Ser Val Thr Glu Gin Asp Ser Lys Asp Ser Thr Tyr ser Leu ser 170 175 Ser Thr Leu Thr Leu Ser Lys Ala Asp Tyr Glu Lys His Lys val Tyr 185 190 Ala cys Glu val Thr His Gin Gly Leu Ser Ser Pro Val Thr Lys Ser 200 205 Phe Asn Arg Gly Glu cys <210> 37 <211> 111 <212> PRT <213> Artificial sequence <220> <223> amino acid sequence of the light chain variable domain of the chimeric B1-23 anti-GDF-15 antibody <400> 37 Asp lie vai Leu Thr Gin Ser Pro Lys Phe Met ser Thr ser Va] ciy 1 5 10 15 Asp A9 Val Ser Val Thr Cys Lys Ala Ser Gin Asn val dy Thr Asn 25 30 val Ala Phe Leu Gin Lys Pro dy Gin ser Pro LS Ala Leu lie 40 45 Tyr Ser Ala ser Tyr Arg Tyr ser dy vai Pro AS Arg Phe Thr Gly 55 60 Ser dy Ser dy Thr Asp Phe Thr Leu Thr lie Ser Asn Val Gin Ser 70 75 80 Glu Asp Leu Ala Glu Tyr Phe cys Gin Gin Tyr Asn Asn Phe Pro Tyr 90 95 Thr Phe Ely dy Gly Thr Lys Leu Glu lie Lys Arg Thr Va] Ala 105 110 <210> 38 <211> 103 <212> PRT <213> Artificial Sequence <220> <223> amino acid sequence of the light chain constant domain of the chimeric B1-23 anti-GDF-15 antibody <400> 38 Ala Pro ser val Phe lie Phe Pro Pro Ser Asp Giu Gin Leu Lys ser 1 5 10 15 Giy Thr Ala ser vai val cys Leu Leu Asn Asn Phe Tyr Pro Arg Giu 25 30 Ala Lys val Gin Trp Lys val Asp Asn Ala Leu Gin Ser Gly Asn Ser 40 45 Gin Glu ser val Thr Glu Gin Asp Ser LS Asp Ser Thr Tyr Ser Leu 55 60 Ser ser Thr Leu Thr Leu Ser Lys Ala sp Tyr Glu Lys His Lys val 70 75 80 Tyr Ala cys Glu val Thr His Gin dy Leu Ser ser Pro Val Thr Lys 90 95 Ser Phe Asn Arq dy Glu cys

Claims (32)

  1. Claims 1. A monoclonal antibody capable of binding to human GDF- 15, or an antigen-binding portion thereof, wherein the heavy chain variable domain comprises a CDR3 region comprising the amino acid sequence of SEQ ID NO: S or an amino acid sequence at least 90% identical thereto, and wherein the light chain variable domain comprises a CDR3 region comprising the amino acid sequence of SEQ ID NO: 7 or an amino acid sequence at least 85% identical thereto, for use in a method for treating cancer cachexia in a mammal.
  2. 2. A monoclonal antibody capable of binding to human GDF- 15, or an antigen-binding portion thereof, wherein the binding is binding to a conformational or discontinuous epitope on human GDF-15 comprised by the amino acid sequences of SEQ lID No: 25 and SEQ ID No: 26, for use in a method for treating cancer cachexia in a mammal.
  3. 3. The antibody or antigen-binding portion thereof of claim 1 or 2 for the use according to claim 1 or 2, wherein the method for treating cancer cachexia is a method for completely preventing or completely reverting cancer cachexia.
  4. 4. The antibody or antigen-binding portion thereof of claim 3 for the use according to claim 3, wherein the method for treating cancer cachexia is a method for completely preventing cancer cachexia.
  5. 5. The antibody or antigen-binding portion thereof of claim 3 for the use according to claim 3, wherein the method for treating cancer cachexia is a method for completely reverting cancer cachexia.
  6. 6. The antibody or antigen-binding portion thereof of any one of claims 1-5 for the use according to any one of claims 1-5, wherein in the method, only mammals suffering from both i) the cancer, and ii) cancer cachexia are treated.
  7. 7. The antibody or antigen-binding portion thereof of any one of claims 5-6 for the use according to any one of claims 5-6, wherein the method increases body weight of the mammal compared to its body weight before the onset of cancer cachexia.
  8. 8. The antibody or antigen-binding portion thereof of claim 7 for the use according to claim 7, wherein the increase in body weight of the mammal is at least 1.5%, preferably at least 2.5%, more preferably at least 5% compared to its body weight before the onset of cancer cachexia.
  9. 9. The antibody or antigen-binding portion thereof of any one of claims 1-8 for the use according to any one of claims 1-8, wherein the method is a method for both treating cancer and treating cancer cachexia in the same mammal.
  10. 10.The antibody or antigen-binding portion thereof of any one of claims 1-9 for the use according to any one of claims 1-9, wherein the antibody has a size of more than 100 kDa, preferably more than 110 kDa, more preferably more than 120 kDa, still more preferably more than 130 kDa, and most preferably more than 140 kna.
  11. 11.The antibody or antigen-binding portion thereof of claim 10 for the use according to claim 10, wherein the antibody is a full-length antibody.
  12. 12.The antibody or antigen-binding portion thereof of claim 11 for the use according to claim 11, wherein the antibody is a full-length IgG antibody.
  13. 13..The antibody or antigen-binding portion thereof of any one of claims 1 to 12 for the use according to any one of claims 1 to 12, wherein the antibody has an Fc portion which is capable of binding to the Fc receptor.
  14. 14.The antibody or antigen-binding portion thereof of any one of claims 1 to 13 for the use according to any one of claims 1 to 13, wherein the cancer cells of the mammal endogenously express GDF-15 and/or the cancer cells of the mammal stimulate endogenous expression of GDF-l5 in non-cancerous cells of the mammal.
  15. 15.The antibody or antigen-binding portion thereof of any one of claims 1 to 14 for the use according to any one of claims 1 to 14, wherein the cancer cells of the mammal endogenously express GDF-15.
  16. 16.The antibody or antigen-binding portion thereof of any one of claims 1 to 15 for the use according to any one of claims 1 to 15, wherein the mammal is a human patient.
  17. 17.The antibody or antigen-binding portion thereof of any one of claims 1 to 16 for the use according to any one of claims 1 to 16, wherein the human GDF-1S is recombinant human GDF-15 having the amino acid sequence represented by SEQ ID No: 8.
  18. 18.The antibody or antigen-binding portion thereof of any one of claims 2-17 for the use according to any one of claims 2-17, wherein the heavy chain variable domain comprises a CDR3 region comprising the amino acid sequence of SEQ ID NO: 5 or an amino acid sequence at least 90% identical thereto, and wherein the light chain variable domain comprises a CDR3 region comprising the amino acid sequence of SEQ ID NO: 7 or an amino acid sequence at least 85% identical thereto.
  19. 19.The antibody or antigen-binding portion thereof of any one of claims 1 and 3-17 for the use according to any one of claims 1 and 3-17, wherein the binding is binding to a conformational or discontinuous epitope on human GDF-15 that is comprised by the amino acid sequences of SEQ ID No: 25 and SEQ ID No: 26.
  20. 20.The antibody or antigen-binding portion thereof of any one of claims 1-19 for the use according to any one of claims 1-19, wherein the method for treating cancer is a method comprising inhibition of cancer growth.
  21. 21.The antibody or antigen-binding portion thereof of any one of claims 1-20 for the use according to any one of claims 1-20, wherein the method for treating cancer comprises the induction of killing of cancer cells by NK cells and CDB+ T cells in the human patient.
  22. 22.The antibody or antigen-binding portion thereof of any one of claims 1-21 for the use according to any one of claims 1-21, wherein the heavy chain variable domain comprises a CDR1 region comprising the amino acid sequence of SEQ ID NO: 3 and a CDR2 region comprising the amino acid sequence of SEQ ID NO: 4, and wherein the light chain variable domain comprises a CDR1 region comprising the amino acid sequence of SEQ ID NO: 6 and a CDR2 region comprising the amino acid sequence ser-ala-ser.
  23. 23.The antibody or antigen-binding portion thereof of any one of claims 1-22 for the use according to any one of claims 1-22, wherein the constant domain of the heavy chain comprises the amino acid sequence of SEQ ID No: 29, or an amino acid sequence at least 85%, preferably at least 90%, more preferably at least 95% identical thereto, and wherein the constant domain of the light chain comprises the amino acid sequence of SEQ ID No: 32, or an amino acid sequence at least 85%, preferably at least 90%, more preferably at least 95% identical thereto.
  24. 24.The antibody or antigen-binding portion thereof of any one of claims 1-23 for the use according to any one of claims 1-23, wherein the constant domain of the heavy chain comprises the amino acid sequence of SEQ ID No: 29, or an amino acid sequence at least 98%, preferably at least 99% identical thereto, and wherein the constant domain of the light chain comprises the amino acid sequence of SEQ ID No: 32, or an amino acid sequence at least 98%, preferably at least 99% identical thereto.
  25. 25. The antibody or antigen-binding portion thereof of any one of claims 1-24 for the use according to any one of claims 1-24, wherein the constant domain of the heavy chain comprises the amino acid sequence of SEQ ID No: 29, and wherein the constant domain of the light chain comprises the amino acid sequence of SEQ ID No: 32.
  26. 26. The antibody or antigen-binding portion thereof of any one of claims 1-25 for the use according to any one of* claims 1-25, wherein the antibody is a humanized antibody, and wherein all of the variable domains of the antibody are humanized variable domains.
  27. 27.The antibody or antigen-binding portion thereof of any one of claims 1-26 for the use according to any one of claims 1-26, wherein the heavy chain variable domain comprises the amino acid sequence of SEQ ID No: 28, or an amino acid sequence at least 90%, preferably at least 95%, more preferably at least 98%, still more preferably at least 99% identical thereto, and wherein the light chain variable domain comprises the amino acid sequence of SEQ ID No: 31, or an amino acid sequence at least 90%, preferably at least 95%, more preferably at least 98%, still more preferably at least 99% identical thereto.
  28. 28.The antibody or antigen-binding portion thereof of any one of claims 1-27 for the use according to any one of claims 1-27, wherein the heavy chain variable domain comprises the amino acid sequence of SEQ ID No: 28, and wherein the light chain variable domain comprises the amino acid sequence of SEQ ID No: 31.
  29. 29.The antibody or antigen-binding portion thereof of any one of claims 1-28 for the use according to any one of claims 1-28, wherein the heavy chain comprises the amino acid sequence of SEQ ID No: 27, and wherein the light chain comprises the amino acid sequence of SEQ ID No: 30.
  30. 30.The antibody or antigen-binding portion thereof of any one of claims 1-25 for the use according to any one of claims 1-25, wherein the heavy chain variable domain comprises the amino acid sequence of SEQ ID No: 34, or an amino acid sequence at least 75%, more preferably at least 90%, more preferably at least 95%, more preferably at least 98%, still more preferably at least 99% identical thereto, and wherein the light chain variable domain comprises the amino acid sequence of SEQ ID No: 37, or an amino acid sequence at least 80%, more preferably at least 90%, more preferably at least 95%, more preferably at least 98%, still more preferably at least 99% identical thereto.
  31. 31.The antibody or antigen-binding portion thereof of claim 30 for the use according to claim 30, wherein the heavy chain variable domain comprises the amino acid sequence of SEQ ID No: 34, and wherein the light chain variable domain comprises the amino acid sequence of SEQ ID No: 37.
  32. 32. The antibody or antigen-binding portion thereof of any one of claims 1-22 for the use according to any one of claims 1-22, wherein the antibody is the antibody to human GDF-15 obtainable from the cell line B1-23 deposited with the Deutsche Samrnlung für Mikroorganismen und Zellkulturen GmbH (DMSZ) under the accession No. DSM ACC3142 or an antigen-binding portion thereof.
GB1405477.9A 2014-03-26 2014-03-26 Monoclonal antibodies to growth and differentiation factor 15 (GDF-15), and uses thereof for treating cancer cachexia Active GB2524553C (en)

Priority Applications (41)

Application Number Priority Date Filing Date Title
GB1405477.9A GB2524553C (en) 2014-03-26 2014-03-26 Monoclonal antibodies to growth and differentiation factor 15 (GDF-15), and uses thereof for treating cancer cachexia
BR112016022013A BR112016022013A2 (en) 2014-03-26 2015-03-26 monoclonal antibodies for growth factor and differentiation 15 (gdf-15) and uses of them to treat cancer cachexia
LTEP19205321.3T LT3653644T (en) 2014-03-26 2015-03-26 Monoclonal antibodies to growth and differentiation factor 15 (gdf-15), and uses thereof for treating cancer cachexia and cancer
PL15712629T PL3122775T3 (en) 2014-03-26 2015-03-26 Monoclonal antibodies to growth and differentiation factor 15 (gdf-15), and uses thereof for treating cancer cachexia and cancer
PT192053213T PT3653644T (en) 2014-03-26 2015-03-26 Monoclonal antibodies to growth and differentiation factor 15 (gdf-15), and uses thereof for treating cancer cachexia and cancer
ES19205321T ES2965626T3 (en) 2014-03-26 2015-03-26 Monoclonal antibodies against growth and differentiation factor 15 (GDF-15), and uses thereof to treat cancer cachexia and cancer
NZ724649A NZ724649A (en) 2014-03-26 2015-03-26 Monoclonal antibodies to growth and differentiation factor 15 (gdf-15), and uses thereof for treating cancer cachexia and cancer
RS20191652A RS59700B1 (en) 2014-03-26 2015-03-26 Monoclonal antibodies to growth and differentiation factor 15 (gdf-15), and uses thereof for treating cancer cachexia and cancer
CN201580027932.8A CN106536553B (en) 2014-03-26 2015-03-26 Monoclonal antibodies to growth and differentiation factor 15(GDF-15) and their use for treating cancer cachexia and cancer
PL19205321.3T PL3653644T3 (en) 2014-03-26 2015-03-26 Monoclonal antibodies to growth and differentiation factor 15 (gdf-15), and uses thereof for treating cancer cachexia and cancer
DK15712629.3T DK3122775T3 (en) 2014-03-26 2015-03-26 MONOCLONAL ANTIBODIES FOR GROWTH AND DIFFERENTIATION FACTOR 15 (GDF-15), AND ITS APPLICATIONS FOR TREATING CANCER KAKEKSI AND CANCER
HUE15712629A HUE047398T2 (en) 2014-03-26 2015-03-26 Monoclonal antibodies to growth and differentiation factor 15 (gdf-15), and uses thereof for treating cancer cachexia and cancer
DK19205321.3T DK3653644T3 (en) 2014-03-26 2015-03-26 MONOCLONAL ANTIBODIES AGAINST GROWTH AND DIFFERENTIATION FACTOR 15 (GDF-15) AND USES THEREOF FOR TREATMENT OF CANCER-RELATED CACHESIA AND CANCER
BR122023027559-4A BR122023027559A2 (en) 2014-03-26 2015-03-26 MONOCLONAL ANTIBODIES TO GROWTH AND DIFFERENTIATION FACTOR 15 (GDF-15), CASE COMPRISING THEM, EXPRESSION VECTOR THAT CODES THEM AND CELL LINE CAPABLE OF PRODUCING THEM
US15/128,604 US20170204174A1 (en) 2014-03-26 2015-03-26 Monoclonal antibodies to growth and differentiation factor 15 (gdf-15), and uses thereof for treating cancer cachexia and cancer
EP19205321.3A EP3653644B1 (en) 2014-03-26 2015-03-26 Monoclonal antibodies to growth and differentiation factor 15 (gdf-15), and uses thereof for treating cancer cachexia and cancer
FIEP19205321.3T FI3653644T3 (en) 2014-03-26 2015-03-26 Monoclonal antibodies to growth and differentiation factor 15 (gdf-15), and uses thereof for treating cancer cachexia and cancer
KR1020167029710A KR102520978B1 (en) 2014-03-26 2015-03-26 Monoclonal antibodies to growth and differentiation factor 15 (gdf-15), and uses thereof for treating cancer cachexia and cancer
HRP20231514TT HRP20231514T1 (en) 2014-03-26 2015-03-26 Monoclonal antibodies to growth and differentiation factor 15 (gdf-15), and uses thereof for treating cancer cachexia and cancer
JP2016559286A JP6865585B2 (en) 2014-03-26 2015-03-26 Monoclonal antibody against growth differentiation factor 15 (GDF-15) and its use for treating cancer cachexia and cancer
AU2015238264A AU2015238264B2 (en) 2014-03-26 2015-03-26 Monoclonal antibodies to growth and differentiation factor 15 (GDF-15), and uses thereof for treating cancer cachexia and cancer
PCT/EP2015/056654 WO2015144855A1 (en) 2014-03-26 2015-03-26 Monoclonal antibodies to growth and differentiation factor 15 (gdf-15), and uses thereof for treating cancer cachexia and cancer
ES15712629T ES2763859T3 (en) 2014-03-26 2015-03-26 Monoclonal Antibodies to Growth and Differentiation Factor 15 (GDF-15), and Uses thereof to Treat Cancer Cachexia and Cancer
HUE19205321A HUE064573T2 (en) 2014-03-26 2015-03-26 Monoclonal antibodies to growth and differentiation factor 15 (gdf-15), and uses thereof for treating cancer cachexia and cancer
PT157126293T PT3122775T (en) 2014-03-26 2015-03-26 Monoclonal antibodies to growth and differentiation factor 15 (gdf-15), and uses thereof for treating cancer cachexia and cancer
EP15712629.3A EP3122775B1 (en) 2014-03-26 2015-03-26 Monoclonal antibodies to growth and differentiation factor 15 (gdf-15), and uses thereof for treating cancer cachexia and cancer
LTEP15712629.3T LT3122775T (en) 2014-03-26 2015-03-26 Monoclonal antibodies to growth and differentiation factor 15 (gdf-15), and uses thereof for treating cancer cachexia and cancer
CA2943694A CA2943694A1 (en) 2014-03-26 2015-03-26 Monoclonal antibodies to growth and differentiation factor 15 (gdf-15), and uses thereof for treating cancer cachexia and cancer
SI201531059T SI3122775T1 (en) 2014-03-26 2015-03-26 Monoclonal antibodies to growth and differentiation factor 15 (gdf-15), and uses thereof for treating cancer cachexia and cancer
EP23203704.4A EP4316596A3 (en) 2014-03-26 2015-03-26 Monoclonal antibodies to growth and differentiation factor 15 (gdf-15), and uses thereof for treating cancer cachexia and cancer
IL248038A IL248038B (en) 2014-03-26 2016-09-26 Monoclonal antibodies to growth and differentiation factor 15 (gdf-15), and uses thereof for treating cancer cachexia and cancer
US15/912,358 US10604565B2 (en) 2014-03-26 2018-03-05 Monoclonal antibodies to growth and differentiation factor 15 (GDF-15), and uses thereof for treating cancer cachexia and cancer
CY20201100008T CY1122456T1 (en) 2014-03-26 2020-01-08 MONOCLONAL ANTIBODIES AGAINST GROWTH AND DIFFERENTIATION FACTOR 15 (GDF-15), AND USES THEREOF FOR THE THERAPEUTIC TREATMENT OF CANCER CACHESIS AND CANCER
HRP20200034TT HRP20200034T1 (en) 2014-03-26 2020-01-09 Monoclonal antibodies to growth and differentiation factor 15 (gdf-15), and uses thereof for treating cancer cachexia and cancer
US16/794,406 US11760795B2 (en) 2014-03-26 2020-02-19 Monoclonal antibodies to growth and differentiation factor 15 (GDF-15), and uses thereof for treating cancer cachexia and cancer
US16/794,401 US11634482B2 (en) 2014-03-26 2020-02-19 Monoclonal antibodies to growth and differentiation factor 15 (GDF-15), and uses thereof for treating cancer cachexia and cancer
IL276405A IL276405B (en) 2014-03-26 2020-07-30 Monoclonal antibodies to growth and differentiation factor 15 (gdf-15), and uses thereof for treating cancer cachexia and cancer
AU2020260440A AU2020260440B2 (en) 2014-03-26 2020-10-28 Monoclonal antibodies to growth and differentiation factor 15 (GDF-15), and uses thereof for treating cancer cachexia and cancer
JP2021064572A JP2021119138A (en) 2014-03-26 2021-04-06 Monoclonal antibodies to growth and differentiation factor 15 (gdf-15), and uses thereof for treating cancer cachexia and cancer
JP2023062338A JP2023098966A (en) 2014-03-26 2023-04-06 Monoclonal antibodies to growth and differentiation factor 15 (gdf-15), and uses thereof for treating cancer cachexia and cancer
US18/364,173 US20230382987A1 (en) 2014-03-26 2023-08-02 Monoclonal antibodies to growth and differentiation factor 15 (gdf-15), and usesthereof for treating cancer cachexia and cancer

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
GB1405477.9A GB2524553C (en) 2014-03-26 2014-03-26 Monoclonal antibodies to growth and differentiation factor 15 (GDF-15), and uses thereof for treating cancer cachexia

Publications (4)

Publication Number Publication Date
GB201405477D0 GB201405477D0 (en) 2014-05-07
GB2524553A true GB2524553A (en) 2015-09-30
GB2524553B GB2524553B (en) 2017-06-28
GB2524553C GB2524553C (en) 2017-07-19

Family

ID=50686995

Family Applications (1)

Application Number Title Priority Date Filing Date
GB1405477.9A Active GB2524553C (en) 2014-03-26 2014-03-26 Monoclonal antibodies to growth and differentiation factor 15 (GDF-15), and uses thereof for treating cancer cachexia

Country Status (1)

Country Link
GB (1) GB2524553C (en)

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP3157942B1 (en) * 2014-06-23 2020-06-17 Novartis AG Site specific protein modifications
JP7138567B2 (en) * 2016-04-27 2022-09-16 ノバルティス アーゲー Antibodies against growth differentiation factor 15 and their uses

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20010010908A1 (en) * 1999-03-25 2001-08-02 Patricia A. Billing-Medel Reagents and methods useful for detecting diseases of the prostate
US20090004181A1 (en) * 2004-04-13 2009-01-01 St Vincent's Hospitalsydney Limited Method for Modulating Appetite
WO2013023557A1 (en) * 2011-08-12 2013-02-21 中国医学科学院肿瘤研究所 Humanized monoclone antibody of macrophage inhibition factor 1 and use thereof
WO2014049087A1 (en) * 2012-09-26 2014-04-03 Julius-Maximilians-Universität Würzburg Monoclonal antibodies to growth and differentiation factor 15 (gdf-15)
WO2014100689A1 (en) * 2012-12-21 2014-06-26 Aveo Pharmaceuticals, Inc. Anti-gdf15 antibodies

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20010010908A1 (en) * 1999-03-25 2001-08-02 Patricia A. Billing-Medel Reagents and methods useful for detecting diseases of the prostate
US20090004181A1 (en) * 2004-04-13 2009-01-01 St Vincent's Hospitalsydney Limited Method for Modulating Appetite
WO2013023557A1 (en) * 2011-08-12 2013-02-21 中国医学科学院肿瘤研究所 Humanized monoclone antibody of macrophage inhibition factor 1 and use thereof
WO2014049087A1 (en) * 2012-09-26 2014-04-03 Julius-Maximilians-Universität Würzburg Monoclonal antibodies to growth and differentiation factor 15 (gdf-15)
WO2014100689A1 (en) * 2012-12-21 2014-06-26 Aveo Pharmaceuticals, Inc. Anti-gdf15 antibodies

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
Biochemistry (2000); Vol 40, pp 65-73, "Epitope mapping of the transforming growth factor-beta...", Fairlie et al *
Cancer Science (Feb 2014); Vol 105, pp 176-185, "Macrophage inhibitory factor 1 acts as a potential biomarker...", Wang et al *
Nature Medicine (2007); Vol 13, pp 1333-1340, "Tumor-induced anorexia and weight loss...", Johnen et al *

Also Published As

Publication number Publication date
GB2524553C (en) 2017-07-19
GB201405477D0 (en) 2014-05-07
GB2524553B (en) 2017-06-28

Similar Documents

Publication Publication Date Title
AU2020260440B2 (en) Monoclonal antibodies to growth and differentiation factor 15 (GDF-15), and uses thereof for treating cancer cachexia and cancer
US11891436B2 (en) Monoclonal antibodies to growth and differentiation factor 15 (GDF-15)
GB2524553A (en) Monoclonal antibodies to growth and differentiation factor 15 (GDF-15), and use thereof for treating cancer cachexia
GB2524552A (en) Monoclonal antibodies to growth and differentiation factor 15 (GDF-15), and use thereof for treating cancer