WO2022136693A1 - Domaines variables d'anticorps et anticorps ayant une immunogénicité réduite - Google Patents

Domaines variables d'anticorps et anticorps ayant une immunogénicité réduite Download PDF

Info

Publication number
WO2022136693A1
WO2022136693A1 PCT/EP2021/087618 EP2021087618W WO2022136693A1 WO 2022136693 A1 WO2022136693 A1 WO 2022136693A1 EP 2021087618 W EP2021087618 W EP 2021087618W WO 2022136693 A1 WO2022136693 A1 WO 2022136693A1
Authority
WO
WIPO (PCT)
Prior art keywords
seq
sequence
antibody
amino acid
antibody variable
Prior art date
Application number
PCT/EP2021/087618
Other languages
English (en)
Original Assignee
Numab Therapeutics AG
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from EP20216957.9A external-priority patent/EP4019547A1/fr
Priority claimed from PCT/EP2021/064427 external-priority patent/WO2021239987A1/fr
Priority to EP21844734.0A priority Critical patent/EP4267622A1/fr
Priority to KR1020237024454A priority patent/KR20230125239A/ko
Priority to US18/258,957 priority patent/US20240084039A1/en
Priority to IL303171A priority patent/IL303171A/en
Priority to AU2021405066A priority patent/AU2021405066A1/en
Priority to JP2023537607A priority patent/JP2024501810A/ja
Priority to CN202180085944.1A priority patent/CN116783218A/zh
Priority to CA3205010A priority patent/CA3205010A1/fr
Application filed by Numab Therapeutics AG filed Critical Numab Therapeutics AG
Priority to CA3208781A priority patent/CA3208781A1/fr
Priority to JP2023545975A priority patent/JP2024504471A/ja
Priority to AU2022215847A priority patent/AU2022215847A1/en
Priority to EP22708399.5A priority patent/EP4288451A1/fr
Priority to MX2023009022A priority patent/MX2023009022A/es
Priority to CN202280023785.7A priority patent/CN117043185A/zh
Priority to KR1020237029806A priority patent/KR20230166075A/ko
Priority to PCT/EP2022/052425 priority patent/WO2022167460A1/fr
Priority to TW111104438A priority patent/TW202248211A/zh
Publication of WO2022136693A1 publication Critical patent/WO2022136693A1/fr
Priority to IL304403A priority patent/IL304403A/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/46Hybrid immunoglobulins
    • C07K16/468Immunoglobulins having two or more different antigen binding sites, e.g. multifunctional antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/30Immunoglobulins specific features characterized by aspects of specificity or valency
    • C07K2317/31Immunoglobulins specific features characterized by aspects of specificity or valency multispecific
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/30Immunoglobulins specific features characterized by aspects of specificity or valency
    • C07K2317/35Valency
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • C07K2317/567Framework region [FR]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/60Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments
    • C07K2317/62Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments comprising only variable region components
    • C07K2317/622Single chain antibody (scFv)

Definitions

  • the present invention relates to antibody variable domains, which exhibit a reduced binding to pre-existing anti-drug antibodies (ADA), and to antibodies comprising one or more of said antibody variable domains.
  • the present invention further relates to nucleic acids encoding said antibody variable domains or said antibodies, vector(s) comprising said nucleic acids, host cell(s) comprising said nucleic acids or said vector(s), and a method of producing said antibody variable domains or said multispecific antibodies. Additionally, the present invention relates to pharmaceutical compositions comprising said antibodies and to methods of use thereof.
  • antibodies have become an increasingly important class of biomolecules for research, diagnostic and therapeutic purposes. Initially, antibodies were exclusively obtained by immunizing animals with the corresponding antigen of interest. While antibodies of non-human origin can be used in research and diagnostics, in therapeutic approaches the human body typically recognize non- human antibodies as foreign and raise an immune response against the non-human antibody drug substance, rendering it less or not effective. Even if the administered antibody therapeutics have been humanized, e.g. by grafting of CDRs of non-human origin into human immunoglobulin frameworks to minimize the non-human component, they can still elicit an immune response, which compromises the efficacy and/or safety of these therapeutics.
  • ADAs anti-drug antibodies
  • ADAs can be antibodies, which are already present in human serum (so called pre-existing ADAs) and/or antibodies, which are formed during the course of the therapy.
  • ADA binding can be significantly enhanced for therapeutic antibodies that comprise or are built of portions of a naturally occurring human antibody, e.g. Fab or Fv antibody fragments. It is believed that one of the main reasons for this increase in ADA binding is that in antibody fragments, typically a significant number of amino acids that are formerly shielded by the contact to other antibody portions or domains, become exposed to the solvent and are present to the immune system as potential epitopes.
  • Zhao, L. and Li, J. (2010), BMC Struct. Biol., 10, S6, disclose a method for the prediction of potential B-cell epitopes on a protein surface, based on the structural information of antibody-antigen complexes.
  • the authors identified common structural elements that are often present in B-cell epitopes. In particular they found that in antigen epitopes recognized by antibodies, polar amino acids with flexible side chains such as arginine (R), lysin (K), asparagine (N), glutamine (Q), and histidine (H) are significantly overrepresented. Knowledge of these critical structural elements forms the basis for strategies to avoid them.
  • WO2011/075861 discloses a method for decreasing the immunogenicity of antibody variable domains, in particular scFvs, by mutating one or more amino acids located in the interface between the variable chain and the constant chain of a corresponding full-length antibody. It is further disclosed that (i) residues that are present in turn regions of secondary structures, (ii) residues that have large, flexible side chains or a bulky side chain, or (iii) residues that are hydrophobic, are prone to be B-cell epitopes and thus elicit an immunogenic reaction, and that removal of such amino acids interrupts B-cell epitopes.
  • the one or more amino acid residues to be substituted are Leucine (L), Valine (V), Aspartic acid (D), Phenylalanine (F), Arginine (R) and/or Glutamic Acid (E).
  • WO2011/075861 discloses one example of an scFv having the heavy chain point mutations L12S, L103T and L144T (AHo numbering) that, compared to the unmutated version, exhibits reduced binding to pre-existing ADAs present in human sera.
  • WO2011/075861 teaches a method for decreasing the immunogenicity of antibody variable domains towards preexisting ADAs by replacing small hydrophobic residues such as L and V located in the interface between the variable chain with small and weakly hydrophilic amino acids (such as S and T) and by avoiding large and bulky hydrophilic residues located in said interface.
  • the present invention relates to an antibody variable domain, which specifically binds to a target antigen, comprising:
  • variable heavy chain comprising from N-terminus to C-terminus, the regions HFW1-HCDR1-HFW2-HCDR2-HFW3-HCDR3-HFW4, wherein each HFW designates a heavy chain framework region, and each HCDR designates a heavy chain complementarity-determining region, wherein said variable heavy chain framework regions HFW1 , HFW2, HFW3 and HFW4 are selected from a human VH framework subtype, and wherein said HFW1 , HFW2, HFW3 and HFW4 have one of the following substitutions (AHo numbering): an arginine (R) at amino acid position 12; a glutamine (Q) at amino acid position 144; an arginine (R) at amino acid position 12 and a threonine (T) at amino acid position 103; an arginine (R) at amino acid position 12 and a (Q) at amino acid position 144; a threonine (T) at amino acid position 103 and
  • AHo numbering an
  • variable light chain (ii) a variable light chain (VL), wherein the variable light chain comprises, from N-terminus to C-terminus, the regions LFW1-LCDR1-LFW2-LCDR2-LFW3-LCDR3-LFW4, wherein each LFW designates a light chain framework region, and each LCDR designates a light chain complementarity-determining region, and wherein a. said variable light chain framework regions LFW1 , LFW2, LFW3 and LFW4 are selected from a human antibody VK framework subtype; or b. said variable light chain framework regions LFW1 , LFW2 and LFW3 are selected from a human antibody K framework subtype, and said variable light chain framework region LFW4 is selected from a V ⁇ framework subtype.
  • the present invention relates to an antibody comprising one or more antibody variable domains of the present invention.
  • the present invention relates to an antibody variable domain of the present invention, wherein said antibody variable domain specifically binds to CD137, and comprises: a) a VH sequence of SEQ ID NO: 3 and a VL sequence of SEQ ID NO: 5; b) a VH sequence of SEQ ID NO: 4 and a VL sequence of SEQ ID NO: 5; c) a VH sequence of SEQ ID NO: 8 and a VL sequence of SEQ ID NO: 10; or d) a VH sequence of SEQ ID NO: 9 and a VL sequence of SEQ ID NO: 10.
  • the present invention relates to an antibody variable domain of the present invention, wherein said antibody variable domain specifically binds to PDL1, and comprises: a) a VH sequence of SEQ ID NO: 13 and a VL sequence of SEQ ID NO: 15; b) a VH sequence of SEQ ID NO: 14 and a VL sequence of SEQ ID NO: 16; c) a VH sequence of SEQ ID NO: 19 and a VL sequence of SEQ ID NO: 21 ; or d) a VH sequence of SEQ ID NO: 20 and a VL sequence of SEQ ID NO: 22.
  • the present invention relates to an antibody variable domain of the present invention, wherein said antibody variable domain specifically binds to human serum albumin, and comprises: a) a VH sequence of SEQ ID NO: 25 and a VL sequence of SEQ ID NO: 27; b) a VH sequence of SEQ ID NO: 26 and a VL sequence of SEQ ID NO: 28; c) a VH sequence of SEQ ID NO: 31 and a VL sequence of SEQ ID NO: 33; or d) a VH sequence of SEQ ID NO: 32 and a VL sequence of SEQ ID NO: 34.
  • the present invention relates to an antibody variable domain of the present invention, wherein said antibody variable domain specifically binds to human serum albumin, and comprises a) a VH sequence of SEQ ID NO: 35 and a VL sequence of SEQ ID NO: 38; b) a VH sequence of SEQ ID NO: 36 and a VL sequence of SEQ ID NO: 39; c) a VH sequence of SEQ ID NO: 36 and a VL sequence of SEQ ID NO: 41 ; d) a VH sequence of SEQ ID NO: 37 and a VL sequence of SEQ ID NO: 40; e) a VH sequence of SEQ ID NO: 42 and a VL sequence of SEQ ID NO: 45; f) a VH sequence of SEQ ID NO: 43 and a VL sequence of SEQ ID NO: 46; g) a VH sequence of SEQ ID NO: 43 and a VL sequence of SEQ ID NO: 48; or h) a VH
  • the present invention relates to a nucleic acid or two nucleic acids encoding the antibody variable domain or the antibody of the present invention.
  • the present invention relates to a vector or two vectors comprising the nucleic acid or the two nucleic acids of the present invention.
  • the present invention relates to a host cell or host cells comprising the vector or the two vectors of the present invention.
  • the present invention relates to a method for producing the antibody variable domain of the present invention or the antibody of the present invention, comprising (i) providing the nucleic acid or the two nucleic acids of the present invention, or the vector or the two vectors of the present invention, expressing said nucleic acid or said two nucleic acids, or said vector or vectors, and collecting said antibody variable domain or said antibody from the expression system, or (ii) providing a host cell or host cells of the present invention, culturing said host cell or said host cells; and collecting said antibody variable domain or said antibody from the cell culture.
  • the present invention relates to a pharmaceutical composition
  • a pharmaceutical composition comprising the antibody of the present invention and a pharmaceutically acceptable carrier.
  • the present invention relates to the pharmaceutical composition of the present invention for use as a medicament.
  • An antibody variable domain which specifically binds to a target antigen, comprising:
  • variable heavy chain comprising from N-terminus to C-terminus, the regions HFW1-HCDR1-HFW2-HCDR2-HFW3-HCDR3-HFW4, wherein each HFW designates a heavy chain framework region, and each HCDR designates a heavy chain complementarity-determining region, wherein said variable heavy chain framework regions HFW1 , HFW2, HFW3 and HFW4 are selected from a human VH framework subtype, and wherein said HFW1 , HFW2, HFW3 and HFW4 have one of the following substitutions (AHo numbering): an arginine (R) at amino acid position 12; a glutamine (Q) at amino acid position 144; an arginine (R) at amino acid position 12 and a threonine (T) at amino acid position 103; an arginine (R) at amino acid position 12 and a (Q) at amino acid position 144; a threonine (T) at amino acid position 103 and
  • AHo numbering an
  • variable light chain (ii) a variable light chain (VL), wherein the variable light chain comprises, from N- terminus to C-terminus, the regions LFW1-LCDR1-LFW2-LCDR2-LFW3-LCDR3- LFW4, wherein each LFW designates a light chain framework region, and each LCDR designates a light chain complementarity-determining region, and wherein a. said variable light chain framework regions LFW1 , LFW2, LFW3 and LFW4 are selected from a human antibody K framework subtype; or b. said variable light chain framework regions LFW1 , LFW2 and LFW3 are selected from a human antibody VK framework subtype, and said variable light chain framework region LFW4 is selected from a VA framework subtype.
  • HFW1 , HFW2, HFW3 and HFW4 have one of the following substitutions (AHo numbering): an arginine (R) at amino acid position 12; an arginine (R) at amino acid position 12 and a threonine (T) at amino acid position 103; an arginine (R) at amino acid position 12 and a glutamine (Q) at amino acid position 144; an arginine (R) at amino acid position 12 and a (Q) at amino acid position 144; or an arginine (R) at amino acid position 12, a threonine (T) at amino acid position 103 and a glutamine (Q) at amino acid position 144; preferably wherein said HFW1 , HFW2, HFW3 and HFW4 have one of the following substitutions (AHo numbering): an arginine (R) at amino acid position 12; an arginine (R) at amino acid position 12 and a (Q) at amino acid
  • variable heavy chain framework regions HFW1 , HFW2, HFW3 and HFW4 are selected from the human VH framework subtypes VH1a, VH1 b, VH3 or VH4, in particular from the human VH framework subtype VH3.
  • variable heavy chain framework regions HFW1 , HFW2, HFW3 and HFW4 are selected from a. the combination of framework regions HFW1 , HFW2, HFW3 and HFW4 (/. e.
  • LFW1 , LFW2 and LFW3, and optionally also LFW4, if LFW4 is selected from a human antibody VK framework subtype, are selected from a. the combination of framework regions LFW1 , LFW2, LFW3 and optionally LFW4 (/. e. the non-italicized residues in Table 1 , 2 and 4) of any one of the SEQ ID NOs: 5, 10, 15, 16, 21 , 22, 27, 28, 33, 34, 38, 39, 40, 41 , 45, 46, 47, 48, 61 , 64, 73, 74, 78, 79, 82, 97, 98, 101 , 102, 118 and 119; and b.
  • LFW1 , LFW2, LFW3 and optionally LFW4 (/. e. the non-italicized residues in Table 1 , 2 and 4) of any one of the SEQ ID NOs: 5, 10, 15, 16, 21 , 22, 27, 28, 33, 34, 38, 39, 40, 41 , 45, 46, 47, 48, 61 , 64, 73, 74, 78, 79, 82, 97, 98, 101 , 102, 118 and 119 having 1 , 2 or 3 mutations within the framework region.
  • variable variable domain of any one of the preceding items wherein said variable light chain framework region LFW4 is selected from a VA framework subtype, particularly has a sequence selected from the group consisting of SEQ ID NOs: 123, 124, 125, 126, 127, 128, 129, 130 and 131.
  • the antibody variable domain of any one of the preceding items wherein said antibody variable domain, when being in scFv format, exhibits a reduced binding to pre-existing anti-drug antibodies (ADA) present in human sera when compared to a version of said antibody variable domain that does not comprise the substitutions defined in item 1 , as determined in a pre-existing ADA binding assay.
  • ADA anti-drug antibodies
  • the antibody variable domain of any one of the preceding items, wherein said antibody variable domain, when being in scFv format is further characterized by one or more of the following features: a.
  • Tm melting temperature
  • DSF differential scanning fluorimetry
  • one, two, three or four further antibody variable domains as defined in any of items 1 to 10, having, independently of each other, either specificity for one of the target antigens of the first and second variable domains, or specificity for a target antigen different from the target antigens of the first and second variable domains.
  • the antibody of item 14 wherein the IgG is selected from the IgG subclasses lgG1 and lgG4, in particular lgG4.
  • the antibody of item 16 wherein said antibody is in a format selected from the group consisting of: a tandem scDb (Tandab), a linear dimeric scDb (LD-scDb), a circular dimeric scDb (CD-scDb), a tandem tri-scFv, a tribody (Fab-(scFv)2), a Fab-Fv2, a triabody, an scDb-scFv, a tetrabody, a didiabody, a tandem-di-scFv and a MATCH.
  • the antibody of item 16, wherein said antibody does not comprise CH 1 and/or CL regions.
  • the antibody of item 18 wherein said antibody is in a scDb-scFv, a triabody, a tetrabody or a MATCH format, in particular wherein said antibody is in a MATCH or scDb-scFv format, more particularly wherein said antibody is in a MATCH format, more particularly a MATCH3 or a MATCH4 format.
  • CD137-BD CD137-BD
  • CD137-BD CD137-BD
  • CD137-BD CD137-BD
  • CD137-BD CD137-BD
  • CD137-BD CD137-BD
  • PDL1-BD PDL1
  • PDL1-BD PDL1
  • PDL1-BD PDL1
  • one human serum albumin binding domain comprising a) a VH sequence of SEQ ID NO: 23 and a VL sequence of SEQ ID NO: 27; b) a VH sequence of SEQ ID NO: 24 and a VL sequence of SEQ ID NO: 28; c) a VH sequence of SEQ ID NO: 25 and a VL sequence of SEQ ID NO: 27; or d) a VH sequence of SEQ ID NO: 26 and a VL sequence of SEQ ID NO: 28; with the proviso that at least one of the three binding domains comprises VH/VL sequence pairs selected from c) or d); or wherein the antibody comprises:
  • CD137-BD CD137-BD
  • CD137-BD CD137-BD
  • CD137-BD CD137-BD
  • CD137-BD CD137-BD
  • PDL1-BD PDL1
  • PDL1-BD PDL1
  • a VH sequence of SEQ ID NO: 17 and a VL sequence of SEQ ID NO: 21 a VH sequence of SEQ ID NO: 17 and a VL sequence of SEQ ID NO: 21 ; b) a VH sequence of SEQ ID NO: 18 and a VL sequence of SEQ ID NO: 22; c) a VH sequence of SEQ ID NO: 19 and a VL sequence of SEQ ID NO: 21 ; or d) a VH sequence of SEQ ID NO: 20 and a VL sequence of SEQ ID NO: 22; and
  • one human serum albumin binding domain comprising a) a VH sequence of SEQ ID NO: 29 and a VL sequence of SEQ ID NO: 33; b) a VH sequence of SEQ ID NO: 30 and a VL sequence of SEQ ID NO: 34; c) a VH sequence of SEQ ID NO: 31 and a VL sequence of SEQ ID NO: 33; or d) a VH sequence of SEQ ID NO: 32 and a VL sequence of SEQ ID NO: 34; with the proviso that at least one of the three binding domains comprises VH/VL sequence pairs selected from c) or d).
  • the antibody of item 20 wherein said antibody is a single chain protein in the scDb-scFvs (scMATCH3) format.
  • the antibody of item 21 wherein said antibody is a single-chain protein, wherein said single-chain protein comprises an amino acid sequence consisting of:
  • a first VH region comprising a sequence selected from SEQ ID NOs: 1 , 2, 3 and 4;
  • a second VH region comprising either a sequence selected from SEQ ID NOs: 11 and 13, in case the first VL region has the sequence of SEQ ID NO: 15, or a sequence selected from SEQ ID NOs: 12 and 14, in case the first VL region has the sequence of SEQ ID NO: 16; arranged one after another in the stated order; wherein said first VL region associates with said second VH region to form said PDL1- BD, and said second VL region associates with said first VH region to form said CD137- BD; and wherein said single-chain protein further comprises
  • a hSA-BD which is formed by a third VL region, comprising a sequence selected from SEQ ID NOs: 27 and 28; and a third VH region, comprising either a sequence selected from SEQ ID NOs: 23 and 25, in case the third VL region has the sequence of SEQ ID NO: 27, or a sequence selected from SEQ ID NOs: 24 and 26, in case the third VL region has the sequence of SEQ ID NO: 28; wherein said third VL region and said third VH region are connected via a fourth polypeptide linker; and wherein said hSA-BD is fused C-terminally or N-terminally via a fifth polypeptide linker to said amino acid sequence; or wherein said single-chain protein comprises an amino acid sequence consisting of:
  • a first VH region comprising a sequence selected from SEQ ID NOs: 6, 7, 8 and 9;
  • a second VH region comprising either a sequence selected from SEQ ID NOs: 17 and 18, in case the first VL region has the sequence of SEQ ID NO: 21 , or a sequence selected from SEQ ID NOs: 18 and 20, in case the first VL region has the sequence of SEQ ID NO: 22; arranged one after another in the stated order; wherein said first VL region associates with said second VH region to form said PDL1- BD, and said second VL region associates with said first VH region to form said CD137- BD; and wherein said single-chain protein further comprises
  • a hSA-BD which is formed by a third VL region, comprising a sequence selected from SEQ ID NOs: 33 and 34; and a third VH region, comprising either a sequence selected from SEQ ID NOs: 29 and 31 , in case the third VL region has the sequence of SEQ ID NO: 33, or a sequence selected from SEQ ID NOs: 30 and 32, in case the third VL region has the sequence of SEQ ID NO: 34; wherein said third VL region and said third VH region are connected via a fourth polypeptide linker; and wherein said hSA-BD is fused C-terminally or N-terminally via a fifth polypeptide linker to said amino acid sequence.
  • ADA anti-drug antibodies
  • the antibody variable domain of item 28, wherein said antibody variable domain, when being in scFv format a. binds to human serum albumin (hSA) with a monovalent dissociation constant (KD) of less than 10 nM, particularly with a KD of 0.05 to 10 nM, particularly of 0.05 to 5 nM, at a pH value of 5.5, as measured by surface plasmon resonance (SPR); b.
  • hSA human serum albumin
  • KD monovalent dissociation constant
  • SPR surface plasmon resonance
  • cSA Macaca fascicularis
  • mSA Mus musculus serum albumin
  • the antibody variable domain of item 29, wherein said antibody variable domain, when being in scFv format, is further characterized by one or more of the following features: d. binds to human serum albumin (hSA) with a monovalent dissociation constant (KD) of less than 10 nM, particularly with a KD of 0.05 to 10 nM, particularly of 0.05 to 5 nM, at a pH value of 7.4, as measured by surface plasmon resonance (SPR); e.
  • hSA human serum albumin
  • KD monovalent dissociation constant
  • SPR surface plasmon resonance
  • cSA Macaca fascicularis
  • mSA Mus musculus serum albumin
  • Tm melting temperature
  • DSF differential scanning fluorimetry
  • hSA-BD has a loss in monomer content, after storage for 28 days at 4°C, of less than 2 %, preferably less than 1 %, when said antigen-binding fragment is at a starting concentration of 10 mg/ml, and wherein said hSA-BD is formulated in 50 mM phosphate citrate buffer with 150 mM NaCI at pH 6.4; i. has a loss in protein content, after storage for 28 days at 4°C, of less than 2 %, preferably less than 1 %, when said antigen-binding fragment is at a starting concentration of 10 mg/ml, and wherein said hSA-BD is formulated in 50 mM phosphate citrate buffer with 150 mM NaCI at pH 6.4; j.
  • hSA-BD has a loss in protein content, after storage for 28 days at 40°C, of less than 2 %, preferably less than 1 %, when said antigen-binding fragment is at a starting concentration of 10 mg/ml, and wherein said hSA-BD is formulated in 50 mM phosphate citrate buffer with 150 mM NaCI at pH 6.4; and/or k. has a loss in monomer content, after storage for 14 days at 4°C, of less than 2 %, preferably less than 1 %, when said antigen-binding fragment is at a starting concentration of 50 mg/ml, and wherein said hSA-BD is formulated in 50 mM phosphate citrate buffer with 150 mM NaCI at pH 6.4.
  • a method for producing the antibody variable domain of any one of items 1 to 10 or 25 to 32, or the antibody of any one of items 11 to 24, comprising (i) providing the nucleic acid or the two nucleic acids of item 33, or the vector or the two vectors of item 34, expressing said nucleic acid sequence or nucleic acids, or said vector or vectors, and collecting said antibody variable domain or said antibody from the expression system, or (ii) providing a host cell or host cells of item 35, culturing said host cell or said host cells; and collecting said antibody variable domain or said antibody from the cell culture.
  • a pharmaceutical composition comprising the antibody of any one of items 11 to 24 and a pharmaceutically acceptable carrier. The antibody of any one of items 11 to 24, or the pharmaceutical composition of item 36, for use as a medicament.
  • a method for modifying an antibody where the antibody is fragment-based or is an antibody comprising one or more scFv fragments, the method comprises the step of introducing the following substitutions (AHo numbering) in the VH sequence(s) of said fragment-based antibody or in the VH sequence(s) of the scFv fragment(s) of said antibody: an arginine (R) at amino acid position 12; a glutamine (Q) at amino acid position 144; an arginine (R) at amino acid position 12 and a threonine (T) at amino acid position 103; an arginine (R) at amino acid position 12 and a (Q) at amino acid position 144; a threonine (T) at amino acid position 103 and a glutamine (Q) at amino acid position 144; or an arginine (R) at amino acid position 12; a threonine (T) at amino acid position 103 and a glutamine (Q) at amino acid position 144 to obtain a arg
  • FIG. 1 shows the absobtion levels of pre-existing ADAs in 20 human serum samples for PRO1922 (A), PRO2230 (C), PRO2922 (E) and PRO2925 (G), determined by the ELISA-based pre-existing ADA binding assay described in example 3.
  • the measurements were performed with spiked and unspike serum samples (confirmation assay setup). Further shown are the corresponding reduction of absorbance level (inhibition (%)) of spiked human serum samples for PRO1922 (B), PRO2230 (D), PRO2922 (F) and PRO2925 (H).
  • NC negative control.
  • PC positice control, /. e. a human/rabbit chimeric antibody exhibiting high binding to pre-existing antibodies.
  • FIG. 2 shows the absobtion levels of pre-existing ADAs in 40 human serum samples for NM21-1480 (A) and PRO2764 (B), determined by the ELISA-based pre-existing ADA binding assay described in example 3. The measurements were performed with spiked and unspike serum samples (confirmation assay setup). NC: negative control.
  • FIG. 3 shows the absobtion levels of pre-existing ADAs in 20 human serum samples for PRO2741 (A) and PRO2660 (B), determined by the ELISA-based pre-existing ADA binding assay described in example 3. The measurements were performed with spiked and unspike serum samples (confirmation assay setup). Further shown are the corresponding reduction of absorbance level (Inhibition (%)) of spiked human serum samples said molecules (C).
  • FIG. 4 shows the absobtion levels of pre-existing ADAs in 20 human serum samples (19 in case of PRO2589) for PRO2268 (A), PRO2269 (B), PRO2510 (C) and PRO2589 (D), determined by the ELISA-based pre-existing ADA binding assay described in example 3.
  • the measurements were performed with spiked and unspike serum samples (confirmation assay setup). Further shown are the corresponding reduction of absorbance level (Inhibition (%)) of spiked human serum samples said molecules (E).
  • antibody variable domains which are substituted at their heavy chain framework positions 12 and/or 144 by hydrophilic amino acids with large flexible and bulky side chains, i.e. an R at position 12 and/or a Q at position 144 (according to AHo numbering), and optionally are substituted at their heavy chain framework position 103 by a T, when being in scFv format, exhibit a reduced binding to preexisting anti-drug antibodies (ADA) present in human sera when compared to their unsubstituted versions.
  • ADA anti-drug antibodies
  • the present invention provides novel antibody variable domains, which are substituted with an R at heavy chain position 12 and/or with a Q at heavy chain position 144 (according to AHo numbering), and optionally are substituted with a T at heavy chain framework position 103.
  • novel antibody variable domain variants when being in scFv format, exhibit a significantly reduced binding to pre-existing ADAs when compared to their unsubstituted versions.
  • these antibody variable domains could successfully be applied in the construction of various scFvs and fragment-based multispecific antibodies, which exhibit low immunogenicity and excellent stability.
  • the antibody variable domain of the present invention could be successfully used for the construction of several scFvs and antibody fragment-based multispecific antibody formats.
  • the antibody variable domains of the present invention have been successfully applied in the construction of antibody fragment-based multispecific antibodies that target human mesothelin (MSLN), CD3 and human serum albumin (hSA) (MATCH4 format).
  • MSLN human mesothelin
  • hSA human serum albumin
  • the design, manufacturing and the functional and biophysical properties of these antibody fragment-based anti-MSLNxCD3xhSA antibodies are disclosed in detail in the patent application PCT/EP2021/064427. Specific examples are PRO2741 , PRO2745 and PRO2746.
  • the antibody variable domains of the present invention have been successfully incorporated in antibody fragment-based multispecific antibodies that target ROR1 , CD3 and hSA (scMATCH3 and MATCH4 format).
  • the design, manufacturing and the functional and biophysical properties of fragment-based anti-ROR1xCD3xhSA antibodies are disclosed in detail in the experimental part and in the priority document EP21154786.4. Specific examples are PRO2667, PRO2668, PRO2669 and PRO2670.
  • the antibody variable domains of the present invention have been successfully incorporated in Morrison-based multispecific antibodies that target IL-4R and IL- 31 (Morrison-H format).
  • the design, manufacturing and the functional and biophysical properties of the Morrison-based anti-IL4RxlL31 antibodies are disclosed in detail in the priority document EP20216957.9. Specific examples are PRO2198 and PRO2199.
  • the antibody variable domains of the present invention have been successfully incorporated into the antibody fragment-based multispecific antibody NM21- 1480 (scMATCH3 format), which targets PD-L1 , CD137 and hSA.
  • NM21- 1480 Several variants of NM21- 1480 have been prepared.
  • the design, manufacturing and the functional and biophysical properties of the parental fragment-based anti-PDL1xCD137xhSA antibodies are disclosed in detail in the experimental part and in the patent application WO 2019/072868. Specific examples are PRO2758, PRO2759, PRO2760, PRO2761 , PRO2762, PRO2763, PRO2764, PRO2765 and PRO3351.
  • the present invention relates to an antibody variable domain, which specifically binds to a target antigen, comprising:
  • variable heavy chain comprising from N-terminus to C-terminus, the regions HFW1-HCDR1-HFW2-HCDR2-HFW3-HCDR3-HFW4, wherein each HFW designates a heavy chain framework region, and each HCDR designates a heavy chain complementarity-determining region, wherein said variable heavy chain framework regions HFW1 , HFW2, HFW3 and HFW4 are selected from a human VH framework subtype, and wherein said HFW1 , HFW2, HFW3 and HFW4 have one of the following substitutions (AHo numbering): an arginine (R) at amino acid position 12; a glutamine (Q) at amino acid position 144; an arginine (R) at amino acid position 12 and a threonine (T) at amino acid position 103; a threonine (T) at amino acid position 103 and a glutamine (Q) at amino acid position 144; or an arginine (R) at amino acid position
  • variable light chain framework regions LFW1, LFW2, LFW3 and LFW4 are selected from a human antibody VK framework subtype; or b. said variable light chain framework regions LFW1 , LFW2 and LFW3 are selected from a human antibody VK framework subtype, and said variable light chain framework region LFW4 is selected from a VA framework subtype.
  • antibody and the like, as used herein, includes whole antibodies or single chains thereof; and any antigen-binding variable domain (/. e., “antigen-binding portion”) or single chains thereof; and molecules comprising antibody CDRs, VH regions or VL regions (including without limitation multispecific antibodies).
  • a naturally occurring “whole antibody” is a glycoprotein comprising at least two heavy (H) chains and two light (L) chains inter-connected by disulfide bonds.
  • Each heavy chain is comprised of a heavy chain variable region (abbreviated herein as VH) and a heavy chain constant region.
  • the heavy chain constant region is comprised of three domains, CH1 , CH2 and CH3.
  • Each light chain is comprised of a light chain variable region (abbreviated herein as VL) and a light chain constant region.
  • the light chain constant region is comprised of one domain, CL.
  • the VH and VL regions can be further subdivided into regions of hypervariability, termed complementarity determining regions (CDRs), flanked by regions that are more conserved, termed framework regions (FRs).
  • CDRs complementarity determining regions
  • FRs framework regions
  • Each VH and VL is composed of three CDRs and four FRs arranged from amino-terminus to carboxy-terminus in the following order: FR1 , CDR1, FR2, CDR2, FR3, CDR3, FR4.
  • the variable regions of the heavy and light chains contain a binding domain that interacts with an antigen.
  • the constant regions of the antibodies may mediate the binding of the immunoglobulin to host tissues or factors, including various cells of the immune system (e. g., effector cells) and the first component
  • antibody variable domain refers to one or more parts of an intact antibody that have the ability to specifically bind to a given antigen (e. g., PDL1, CD137, ROR1, MSLN, CD3, IL-4R, IL-31 or hSA).
  • a given antigen e. g., PDL1, CD137, ROR1, MSLN, CD3, IL-4R, IL-31 or hSA.
  • This can be any antigen-binding fragment (/. e., “antigen-binding portion”) of an intact antibody or single chains thereof; and molecules comprising antibody CDRs, VH regions or VL regions.
  • the term “antibody variable domain”, as used herein refers to a Fab fragment, /. e.
  • the antibody variable domain of the present invention is selected from a Fab fragment, an Fv fragment, a disulfide stabilized Fv fragment (dsFv) and an scFv fragment.
  • the antibody variable domain of the present invention is selected from a Fab fragment, an Fv fragment, a disulfide stabilized Fv fragment (dsFv) and an scFv fragment.
  • the antibody variable domain of the present invention is an Fv fragment, an scFv fragment or a disulfide stabilized Fv fragment (dsFv).
  • the antibody variable domain of the present invention is a single-chain Fv fragment (scFv).
  • the VL and VH domains of the scFv fragment are stabilized by an interdomain disulfide bond, in particular said VH domain comprises a single cysteine residue in position 51 (AHo numbering) and said VL domain comprises a single cysteine residue in position 141 (AHo numbering).
  • CDRs Complementarity Determining Regions
  • the CDR amino acids in the VH are numbered 26-32 (HCDR1), 52-56 (HCDR2), and 95-102 (HCDR3); and the amino acid residues in VL are numbered 24-34 (LCDR1), 50-56 (LCDR2), and 89-97 (LCDR3).
  • the CDRs consist of amino acid residues 26-35 (HCDR1), 50-65 (HCDR2), and 95-102 (HCDR3) in human VH and amino acid residues 24-34 (LCDR1), 50- 56 (LCDR2), and 89-97 (LCDR3) in human VL.
  • the CDR amino acid residues in the VH are numbered approximately 26-35 (HCDR1), 51-57 (HCDR2) and 93-102 (HCDR3), and the CDR amino acid residues in the VL are numbered approximately 27-32 (LCDR1), 50-52 (LCDR2), and 89-97 (LCDR3) (numbering according to “Kabat”).
  • the CDRs of an antibody can be determined using the program IMGT/DomainGap Align.
  • AHo Honegger & Pluckthun
  • CDRs are defined as CDRs according to AHo numbering scheme: LCDR1 (also referred to as CDR- L1): L24-L42; LCDR2 (also referred to as CDR-L2): L58-L72; LCDR3 (also referred to as CDR-L3): L107-L138; HCDR1 (also referred to as CDR-H1): H27-H42; HCDR2 (also referred to as CDR-H2): H57-H76; HCDR3 (also referred to as CDR-H3): H108-H138.
  • LCDR1 also referred to as CDR- L1
  • LCDR2 also referred to as CDR-L2
  • LCDR3 also referred to as CDR-L3
  • H108-H138 H108-H138.
  • LFW1-LFW4 light chain frameworks 1 to 4
  • HFW1- HFW4 heavy chain frameworks 1 to 4
  • AHo numbering scheme LFW1 : L1-L23; LFW2: L43-L57; LFW3: L73-L106; LFW4: L139-L149; HFW1 : H1-H26; HFW2: H43-H56; HFW3: H77-H107; HFW4: H139-H149.
  • binding specificity refers to the ability of an individual antibody to react with one antigenic determinant and not with a different antigenic determinant.
  • the term “specifically binds to” or is “specific for” refers to measurable and reproducible interactions such as binding between a target and an antibody, which is determinative of the presence of the target in the presence of a heterogeneous population of molecules including biological molecules.
  • an antibody that specifically binds to a target is an antibody that binds this target with greater affinity, avidity, more readily, and/or with greater duration than it binds to other targets.
  • “specific binding” is referring to the ability of the antibody to discriminate between the target of interest and an unrelated molecule, as determined, for example, in accordance with specificity assay methods known in the art. Such methods comprise, but are not limited to Western blots, ELISA, RIA, ECL, IRMA, SPR (Surface plasmon resonance) tests and peptide scans.
  • a standard ELISA assay can be carried out. The scoring may be carried out by standard color development (e. g. secondary antibody with horseradish peroxide and tetramethyl benzidine with hydrogen peroxide).
  • the reaction in certain wells is scored by the optical density, for example, at 450 nm.
  • an SPR assay can be carried out, wherein at least 10-fold, particularly at least 100-fold difference between a background and signal indicates on specific binding.
  • determination of binding specificity is performed by using not a single reference molecule, but a set of about three to five unrelated molecules, such as milk powder, transferrin or the like.
  • the antibody variable domains of the present invention bind to a target antigen, which can be any target antigen.
  • target antigens include, but are not limited to: a transmembrane molecule, a receptor, a ligand, a growth factor, a growth hormone, a clotting factor, an anti-clotting factor, a plasminogen activator, a serum albumin, a receptor for a hormone or a growth factor, a neurotrophic factor, a nerve growth factor, a fibroblast growth factor, transforming growth factor (TGF), a CD protein, an interferon, a colony stimulating factor (CSF), an interleukin (IL), a T-cell receptor, a T-cell co-stimulatory receptor, such as CD137, a surface membrane protein, a viral protein, a tumor associated antigen, an integrin or an interleukin, VEGF; a renin; a human growth hormone; a bovine growth hormone; a growth hormone releasing an enzyme
  • TAA tumor-associated antigen
  • a TAA is an antigen that is preferentially expressed on a tumor cell when compared to non-tumor cells, particularly wherein expression of the TAA on a tumor cell is at least more than 5-fold, at least more than 10-fold, at least more than 20-fold, at least more than 50- fold, or at least more than 100-fold higher than on non-tumor cells from the same organism or patient.
  • tumor associated antigen targets include, but are not limited to: ADRB3, AFP, ALK, BCMA, beta human chorionic gonadotropin, CA-125 (MUC16), CAIX, CD123, CD133, CD135, CD135 (FLT3), CD138, CD171, CD19, CD20, CD22, CD24, CD276, CD33, CD33, CD38, CD44v6, CD79b, CD97, CDH3 (cadherin 3), CEA, CEACAM6, CLDN6, CLEC12A (CLL1), CSPG4, CYP1 B1, EGFR, EGFRvlll, EPCAM, EPHA2, Ephrin B2, ERBBs (e. g.
  • ERBB2 ERBB2
  • FAP FAP
  • FGFR1 folate receptor alpha
  • folate receptor beta Fos-related antigen
  • GA733, GD2, GD3, GFRalpha4 globoH
  • GPC3, GPR20 GPRC5D
  • HAVCR1 Her2/neu (HER2)
  • HLA-A2, HMWMAA HPV E6 or E7
  • human telomerase reverse transcriptase IL-11 Ra, IL-13Ra2, intestinal carboxyl esterase, KIT, Legumain, LewisY, LMP2, Ly6k
  • MAD-CT-1 MAD-CT-2
  • ML-IAP MN-CA IX
  • MSLN MUC1, mut hsp 70-2, NA- 17, NCAM, neutrophil elastase, NY-BR-1, NY-ESO-1 , o-acetyl-GD2, OR51E2, PANX3, PDGFR-beta, PLAC1 , Polysialic acid, PS
  • Preferred examples are: CD138, CD79b, TPBG (5T4), HER2, MSLN, MUC1 , CA- 125 (MUC16), PSMA, BCMA, CD19, EpCAM, CLEC12A (CLL1), CD20, CD22, CEA, CD33, EGFR, GPC3, CD123, CD38, CD33, CD276, CDH3 (cadherin 3), FGFR1 , SSTR2, CD133, EPHA2, HLA-A2, IL13RA2, ROR1 , CEACAM6, CD135, GD-2, GA733, CD135 (FLT3), CSPG4 and TAG-72.
  • Particular examples are: CD138, CD79b, CD123, MSLN, PSMA, BCMA, CD19, CD20, CEA, CD38, CD33, CLEC12a, and ROR1.
  • the HFW1 , HFW2, HFW3 and HFW4 comprised in the antibody variable domains of the invention have one of the following substitutions (AHo numbering): an arginine (R) at amino acid position 12; an arginine (R) at amino acid position 12 and a (Q) at amino acid position 144; or an arginine (R) at amino acid position 12, a threonine (T) at amino acid position 103 and a glutamine (Q) at amino acid position 144.
  • AHo numbering an arginine (R) at amino acid position 12
  • an arginine (R) at amino acid position 12 and a (Q) at amino acid position 144 or an arginine (R) at amino acid position 12, a threonine (T) at amino acid position 103 and a glutamine (Q) at amino acid position 144.
  • the HFW1, HFW2, HFW3 and HFW4 comprised in the antibody variable domains of the invention have the following substitutions (AHo numbering): an arginine (R) at amino acid position 12, a threonine (T) at amino acid position 103, and a glutamine (Q) at amino acid position 144.
  • AHo numbering an arginine (R) at amino acid position 12
  • T threonine
  • Q glutamine
  • the VH domains of the binding domains of the invention belong to a human antibody VH family.
  • the VH domains of the binding domains of the invention belong to VH framework subtypes VH1a, VH1b, VH3 or VH4.
  • the binding domains of the invention comprises a VH domain belonging to the VH framework subtype VH3.
  • the term “belonging to or selected from a VHx framework subtype (or VK/V framework subtype)” means that the framework sequences HFW1 to HFW4 (or LF1 to LFW4) show the highest degree of homology to said human antibody VH or VL framework subtype.
  • a specific example of a VH domain belonging to the VH3 framework subtype is represented by SEQ ID NO: 114 or 115, and specific examples of a VH domain belonging to the VH1a, VH1b or VH4 framework subtype are represented by SEQ ID NO: 120, 121 and 122 (Table 8, framework regions are marked in non-bold).
  • VH1a, VH1b, VH3 and VH4 sequences may be found in Knappik et al., J. Mol. Biol. 296 (2000) 57-86 or in WO 2019/057787.
  • variable heavy chain framework regions HFW1, HFW2, HFW3 and HFW4 of the antibody variable domain of the present invention are selected from the combination of framework regions (the non-italicized residues in Tables 1 to 7, /. e.
  • VH regions further include VH domains comprising at least positions 5 to 140 (AHo numbering), particularly at least positions 3 to 145, more particularly at least positions 2 to 147 of one of the sequences shown in the SEQ ID NOs: 3, 4, 8, 9, 13, 14, 19, 20, 25, 26, 31, 32, 35, 36, 37, 42, 43, 44, 60, 63, 71, 72, 76, 77, 81, 95, 96, 99, 100, 116 and 117, provided that such VH domains exhibit the functional features defined above in items 9 and 10.
  • variable light chain frameworks LFW1 , LFW2, LFW3 and LFW4 of the antibody variable domain of the present invention are selected from a human antibody VK framework subtype (e.g. a VK1 , VK2, VK3 or VK4 framework subtype), particularly are of the VK1 framework subtype.
  • a VK1 framework subtype is represented by SEQ ID NO: 118 or 119 (Table 8, framework regions are marked in non-bold).
  • Alternative examples of VK1 sequences, and examples of VK2, VK3 or VK4 sequences, may be found in Knappik et al., J. Mol. Biol. 296 (2000) 57-86.
  • variable light chain frameworks LFW1, LFW2 and LFW3 are selected from a human antibody VK framework subtype, preferably a K1 framework subtype, and the variable light chain framework LFW4 is selected from a VA framework subtype.
  • the variable light chain framework LFW4 of the antibody variable domain of the present invention is selected from the group consisting of the VA framework 4 sequences of SEQ ID NOs: 123, 124, 125, 126, 127, 128, 129, 130 and 131.
  • VA framework 4 sequence of SEQ ID NO: 129 comprises a single cysteine residue at the variable light (VL) chain position 144 (AHo numbering) and is in particular applied in cases where a second single cysteine is present in the corresponding variable heavy (VH) chain, particularly in position 51 (AHo numbering) of VH, for the formation of an inter-domain disulfide bond.
  • variable light chain frameworks LFW1, LFW2, LFW3 and LFW4 of the antibody variable domain of the present invention are selected from the combination of framework regions (the non-italicized residues in Tables 1 to 7, /. e.
  • VL regions further include VL domains comprising at least positions 5 to 140 (AHo numbering), particularly at least positions 3 to 145, more particularly at least positions 2 to 147 of one of the sequences shown in the SEQ ID NOs: 5, 10, 15, 16, 21 , 22, 27, 28, 33, 34, 38, 39, 40, 41, 45, 46, 47, 48, 61 , 64, 73, 74, 78, 79, 82, 97, 98, 101, 102, 118 and 119, provided that such VL domains exhibit the functional features defined above in items 9 and 10.
  • the antibody variable domain of the present invention is in a format selected from a Fab fragment, /. e. a monovalent fragment consisting of the VL, VH, CL and CH1 domains; an Fv fragment consisting of the VL and VH domains of a single arm of an antibody; a disulfide stabilized Fv fragment (dsFv); and a single chain Fv fragment (scFv).
  • the antibody variable domain of the present invention is selected from an Fv fragment, a disulfide stabilized Fv fragment (dsFv) and a single-chain Fv fragment (scFv).
  • the antibody variable domain of the present invention is selected from an Fv fragment and a single-chain Fv fragment (scFv).
  • the VL and VH domains of the scFv fragment are stabilized by an interdomain disulfide bond, in particular said VH domain comprises a single cysteine residue in position 51 (AHo numbering) and said VL domain comprises a single cysteine residue in position 141 (AHo numbering).
  • the antibody variable domain of the present invention when being in scFv format, exhibits a reduced immunogenicity, when compared to a version of said antibody variable domain that does not comprise the above defined substitutions in the VH framework regions. More specifically, the antibody variable domain of the present invention, when being in scFv format, exhibits a reduced binding to pre-existing anti-drug antibodies (ADA) present in human sera, in particular reduced binding to pre-existing ADAs when compared to a version of said antibody variable domain that does not comprise the above defined substitutions in the VH framework regions, as determined in a pre-existing ADA binding assay, in particular as determined in a pre-existing ADA binding assay as defined in Example 3.
  • ADA anti-drug antibodies
  • Immunogenicity i.e. the tendency of a therapeutic protein to induce an antibody response within the patient's body, can e.g. be predicted by its capacity to be recognized by anti-drug antibodies (ADAs) that are already present in human sera of healthy and untreated individuals, herein referred to as “pre-existing ADAs”.
  • ADAs anti-drug antibodies
  • the term “immunogenicity”, as used herein, refers to the capacity of a therapeutic protein, e.g. an antibody, an antibody fragment or an antibody binding domain, to be recognized by pre-existing ADAs in human serum samples.
  • a therapeutic protein e.g. an antibody, an antibody fragment or an antibody binding domain
  • pre-existing ADA binding as well as the induction of the formation of ADAs during therapeutic treatment is linked with the occurrence of B cell and/or T cell epitopes on a therapeutic protein.
  • the extent of such immunogenicity can be determined by an ELISA assay and can be expressed by the percentage of human serum samples, which contain measurable amounts of pre-existing ADAs and/or ADAs formed during therapeutic treatment, that recognize, /. e.
  • a reduction of immunogenicity between a therapeutic protein and a corresponding therapeutic protein being modified with the goal to reduce its immunogenicity can be measured by comparing the percentage of positive serum samples against the modified therapeutic protein, with the percentage of positive serum samples against the original therapeutic protein. A lower number or percentage of positive serum samples for the modified therapeutic protein indicates a reduction of immunogenicity relative to the original therapeutic protein.
  • a serum sample is deemed to contain measurable amounts of pre-existing ADAs, when the ELISA signal surpasses a certain threshold.
  • This threshold is herein also referred to as the screening cut-point (SCP).
  • SCP can be calculated as defined below or set to an arbitrary value relative to the maximum ELISA signal obtained for the tested sera (e.g. 20 %, 15 %, 10 % or 5 % of the maximum ELISA signal obtained for the tested sera).
  • the SCP is calculated as defined below.
  • the antibody variable domains of the present invention when being in scFv format, further have advantageous biophysical properties, in particular an excellent stability.
  • the antibody variable domain of the present invention when being in scFv format, is further characterized by one or more of the following features: a. has a melting temperature (Tm), determined by differential scanning fluorimetry (DSF), of at least 65°C, when formulated in 50 mM phosphate citrate buffer with 150 mM NaCI at pH 6.4; b. has a loss in monomer content, after storage for 28 days at 4°C, of less than 5 %, when formulated at a concentration of 10 mg/ml in 50 mM phosphate citrate buffer with 150 mM NaCI at pH 6.4; c. has a loss in protein content, after storage for 28 days at 4°C, of less than 5 %, when formulated at a concentration of 10 mg/ml in 50 mM phosphate citrate buffer with 150 mM NaCI at pH 6.4.
  • Tm melting temperature
  • DSF differential scanning fluorimetry
  • DSF is described earlier (Egan, et al., MAbs, 9(1) (2017), 68-84; Niesen, et al., Nature Protocols, 2(9) (2007) 2212-2221).
  • the midpoint of transition for the thermal unfolding of the scFv constructs is determined by Differential Scanning Fluorimetry using the fluorescence dye SYPRO® Orange (see Wong & Raleigh, Protein Science 25 (2016) 1834- 1840).
  • Samples in phosphate-citrate buffer at pH 6.4 are prepared at a final protein concentration of 50 pg/ml and containing a final concentration of 5x SYPRO® Orange in a total volume of 100 pl.
  • the assay is performed in a qPCR machine used as a thermal cycler, and the fluorescence emission is detected using the software’s custom dye calibration routine.
  • the PCR plate containing the test samples is subjected to a temperature ramp from 25°C to 96°C in increments of 1°C with 30 s pauses after each temperature increment.
  • the total assay time is about 2 h.
  • the Tm is calculated by the software GraphPad Prism using a mathematical second derivative method to calculate the inflection point of the curve.
  • the reported Tm is an average of three measurements.
  • SE-HPLC is a separation technique based on a solid stationary phase and a liquid mobile phase as outlined by the US Pharmacopeia (USP), chapter 621. This method separates molecules based on their size and shape utilizing a hydrophobic stationary phase and aqueous mobile phase. The separation of molecules is occurring between the void volume (Vo) and the total permeation volume (VT) of a specific column. Measurements by SE-HPLC are performed on a Chromaster HPLC system (Hitachi High-Technologies Corporation) equipped with automated sample injection and a UV detector set to the detection wavelength of 280 nm.
  • the equipment is controlled by the software EZChrom Elite (Agilent Technologies, Version 3.3.2 SP2) which also supports analysis of resulting chromatograms. Protein samples are cleared by centrifugation and kept at a temperature of 4-6°C in the autosampler prior to injection.
  • EZChrom Elite Agilent Technologies, Version 3.3.2 SP2
  • Protein samples are cleared by centrifugation and kept at a temperature of 4-6°C in the autosampler prior to injection.
  • the column Shodex KW403-4F Showa Denko Inc., #F6989202
  • the target sample load per injection was 5 pg.
  • the present invention relates to an antibody comprising one or more antibody variable domains of the present invention.
  • the antibody of the present invention further comprises antibody variable domains that differ from the antibody variable domains of the present invention. More specifically, the antibody of the present invention further comprises antibody variable domains that do not have the substitutions in the framework regions as defined herein.
  • the antibody of the present invention exclusively comprises antibody variable domains of the present invention. More specifically, in these preferred embodiments, the antibody of the present invention exclusively comprises antibody variable domains that have the substitutions in the framework regions as defined herein.
  • binding domain or “monovalent binding domain”, as used herein, refers to a binding domain that binds to a single epitope on a target molecule.
  • bivalent antibody or “antibody that is bivalent for its target antigen”, as used herein, refers to a single antibody with two valencies, where “valency” is described as the number of antigen-binding moieties that binds to epitopes on a specific target molecule.
  • the single antibody can bind to two binding sites on a target molecule and/or to two target molecules due to the presence of two copies of the corresponding antigen-binding moieties.
  • trivalent antibody or “antibody that is trivalent for its target antigen”, as used herein, refers to a single antibody with three valencies. As such, the single antibody can bind to three binding sites on a target molecule and/or can bind up to three target molecules due to the presence of three copies of the corresponding antigen-binding moieties.
  • the antibodies of the invention comprise two or three binding domains
  • said two or three binding domains either bind the same epitope or different epitopes on the target molecules.
  • the two or three binding domains bind the same epitope on the target molecule.
  • allegene epitope refers to an individual protein determinant on the protein capable of specific binding to more than one antibody, where that individual protein determinant is identical, /. e. consist of identical chemically active surface groupings of molecules such as amino acids or sugar side chains having identical three-dimensional structural characteristics, as well as identical charge characteristics for each of said antibodies.
  • the format of the antibody is selected from bivalent bispecific IgG formats, trivalent bispecific IgG formats and tetravalent bispecific IgG formats.
  • the format of said antibody is selected from KiH-based IgGs, such as DuoBodies (bispecific IgGs prepared by the Duobody technology) (MAbs. 2017 Feb/Mar;9(2): 182-212.
  • IgG-scFv fusions such as CODV-IgG, Morrison (IgG CH3-scFv fusion (Morrison-H) or IgG CL-scFv fusion (Morrison-L)), bsAb (scFv linked to C-terminus of light chain), Bs1Ab (scFv linked to N- terminus of light chain), Bs2Ab (scFv linked to N-terminus of heavy chain), Bs3Ab (scFv linked to C-terminus of heavy chain), Ts1Ab (scFv linked to N-terminus of both heavy chain and light chain) and Ts2Ab (dsscFv linked to C-terminus of heavy chain).
  • IgG-scFv fusions such as CODV-IgG, Morrison (IgG CH3-scFv fusion (Morrison-H) or IgG CL-scFv fusion (Morrison-L)
  • bsAb s
  • the format of said antibody is selected from KiH-based IgGs, such as DuoBodies; DVD-lg; CODV-IgG and Morrison (IgG CH3-scFv fusion (Morrison-H) or IgG CL-scFv fusion (Morrison-L)), even more particularly from DVD-lg and Morrison (IgG CH3-scFv fusion (Morrison-H) or IgG CL-scFv fusion (Morrison-L)).
  • KiH-based IgGs such as DuoBodies
  • DVD-lg CODV-IgG and Morrison
  • IgG CH3-scFv fusion or IgG CL-scFv fusion (Morrison-L)
  • DVD-lg and Morrison IgG CH3-scFv fusion (Morrison-H) or IgG CL-scFv fusion (Morrison-L)
  • the IgG is preferably selected from the IgG subclasses lgG1 and lgG4, in particular lgG4.
  • the format of said antibody is selected from a Morrison format, /. e. a Morrison-L and a Morrison-H format.
  • the Morrison-L and Morrison-H format used in the present invention are tetravalent and bispecific molecular formats bearing an IgG Fc region, in particular an lgG4 Fc region.
  • Two highly stable scFv binding domains, wherein the light chain comprises VK FR1 to FR3 in combination with a VA FR4, and which are based on the antibody variable domains of the present invention, are fused via a linker L1 to the heavy chain (Morrison-H) or light chain (Morrison-L) C-termini.
  • the linker L1 is a peptide of 2-30 amino acids, more particularly 5-25 amino acids, and most particularly 10-20 amino acids.
  • the VH regions and the VL regions of the two scFv domains are connected by a linker L2.
  • the linker L2 is a peptide of 10-40 amino acids, more particularly 15-30 amino acids, and most particularly 20-25 amino acids.
  • the antibody of the invention does not comprise an immunoglobulin Fc region.
  • immunoglobulin Fc region or “Fc region”, as used herein, is used to define a C-terminal region of an immunoglobulin heavy chain, /. e. the CH2 and CH3 domains of the heavy chain constant regions.
  • Fc region includes native-sequence Fc regions and variant Fc regions, /. e. Fc regions that are engineered to exhibit certain desired properties, such as for example altered Fc receptor binding function and/or reduced or suppressed Fab arm exchange.
  • An example of such an engineered Fc region is the knob- into-hole (KiH) technology (see for example Ridgway et al., Protein Eng.
  • Native-sequence Fc regions include human lgG1 , lgG2 (lgG2A, lgG2B), lgG3 and lgG4.
  • Fc receptor or “FcR” describes a receptor that binds to the Fc region of an antibody.
  • the FcR is a native sequence human FcR, which binds an IgG antibody (a gamma receptor) and includes receptors of the FcyRI, FcyRII, and FcyRIII subclasses, including allelic variants and alternatively spliced forms of these receptors, FcyRII receptors including FcyRIIA (an “activating receptor”) and FcyRI IB (an “inhibiting receptor”), which have similar amino acid sequences that differ primarily in the cytoplasmic domains thereof.
  • Activating receptor FcyRIIA contains an immunoreceptor tyrosine-based activation motif (ITAM) in its cytoplasmic domain.
  • ITAM immunoreceptor tyrosine-based activation motif
  • Inhibiting receptor FcyRIIB contains an immunoreceptor tyrosine-based inhibition motif (ITIM) in its cytoplasmic domain, (see M. Daeron, Annu. Rev. Immunol. 5:203-234 (1997). FcRs are reviewed in Ravetch and Kinet, Annu. Rev. Immunol. 9: 457-92 (1991); Capet et al, Immunomethods 4: 25-34 (1994); and de Haas et al, J. Lab. Clin. Med. 126: 330-41 (1995). Other FcRs, including those to be identified in the future, are encompassed by the term “FcR” herein.
  • ITIM immunoreceptor tyrosine-based inhibition motif
  • Fc receptor or “FcR” also includes the neonatal receptor, FcRn, which is responsible for the transfer of maternal IgGs to the fetus.
  • FcRn the neonatal receptor
  • Methods of measuring binding to FcRn are known (see, e. g., Ghetie and Ward, Immunol. Today 18: (12): 592-8 (1997); Ghetie et al., Nature Biotechnology 15 (7): 637-40 (1997); Hinton et al., J. Biol. Chem.
  • Binding to FcRn in vivo and serum half-life of human FcRn high-affinity binding polypeptides can be assayed, e. g., in transgenic mice or transfected human cell lines expressing human FcRn, or in primates to which the polypeptides having a variant Fc region are administered.
  • WO 2004/42072 (Presta) describes antibody variants which improved or diminished binding to FcRs. See also, e. g., Shields et al., J. Biol. Chem. 9(2): 6591-6604 (2001).
  • the antibody is preferably in a format selected from the group consisting of: a tandem scDb (Tandab), a linear dimeric scDb (LD-scDb), a circular dimeric scDb (CD-scDb), a tandem tri-scFv, a tribody (Fab-(scFv)2), a Fab-Fv2, a triabody, an scDb-scFv, a tetrabody, a di-diabody, a tandem-di-scFv and a MATCH (described in WO 2016/0202457; Egan T., et al., MABS 9 (2017) 68-84).
  • the antibody of the invention does further not comprise CH1 and/or CL regions.
  • the antibody is in a scDb-scFv, a triabody, a tetrabody or a MATCH format, particularly in a MATCH or scDb-scFv format. More particularly, the antibody of the invention is in a MATCH3 or a MATCH4 format.
  • the antibody of the invention is trispecific and tetravalent.
  • the antibody of the invention is trispecific and trivalent.
  • the antibody variable domains comprised in the bispecific, trispecific tetraspecific or pentaspecific, antibodies of the invention are capable of binding to their respective antigens or receptors simultaneously.
  • the term “simultaneously”, as used in this connection refers to the simultaneous binding of one of the antibody variable domains, which for example specifically binds to ROR1 , and of one or two further antibody variable domains, which for example have specificity for CD3 and hSA.
  • the antibody variable domains comprised in the bispecific, trispecific tetraspecific or pentaspecific, antibodies of the invention are operably linked.
  • operably linked indicates that two molecules (e. g., polypeptides, domains, binding domains) are attached in a way that each molecule retains functional activity. Two molecules can be “operably linked” whether they are attached directly or indirectly (e. g., via a linker, via a moiety, via a linker to a moiety).
  • linker refers to a peptide or other moiety that is optionally located between binding domains or antibody variable domains used in the invention. A number of strategies may be used to covalently link molecules together.
  • the linker is a peptide bond, generated by recombinant techniques or peptide synthesis. Choosing a suitable linker for a specific case where two polypeptide chains are to be connected depends on various parameters, including but not limited to the nature of the two polypeptide chains (e. g., whether they naturally oligomerize), the distance between the N- and the C-termini to be connected if known, and/or the stability of the linker towards proteolysis and oxidation. Furthermore, the linker may contain amino acid residues that provide flexibility.
  • polypeptide linker refers to a linker consisting of a chain of amino acid residues linked by peptide bonds that is connecting two domains, each being attached to one end of the linker.
  • the polypeptide linker should have a length that is adequate to link two molecules in such a way that they assume the correct conformation relative to one another so that they retain the desired activity.
  • the polypeptide linker has a continuous chain of between 2 and 30 amino acid residues (e. g., 2, 3, 4, 5, 6, 7, 8, 9, 10, 11 , 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30 amino acid residues).
  • the amino acid residues selected for inclusion in the polypeptide linker should exhibit properties that do not interfere significantly with the activity of the polypeptide.
  • the linker peptide on the whole should not exhibit a charge that would be inconsistent with the activity of the polypeptide, or interfere with internal folding, or form bonds or other interactions with amino acid residues in one or more of the monomers that would seriously impede the binding of receptor monomer domains.
  • the polypeptide linker is non-structured polypeptide.
  • Useful linkers include glycine-serine, or GS linkers.
  • Gly-Ser or “GS” linkers is meant a polymer of glycines and serines in series (including, for example, (Gly-Ser) n , (GSGGS) n (GGGGS)n and (GGGS) n , where n is an integer of at least one), glycine-alanine polymers, alanine-serine polymers, and other flexible linkers such as the tether for the shaker potassium channel, and a large variety of other flexible linkers, as will be appreciated by those in the art. Glycine-serine polymers are preferred since oligopeptides comprising these amino acids are relatively unstructured, and therefore may be able to serve as a neutral tether between components. Secondly, serine is hydrophilic and therefore able to solubilize what could be a globular glycine chain. Third, similar chains have been shown to be effective in joining subunits of recombinant proteins such as single-chain antibodies.
  • the antibody variable domain of the invention is an isolated variable domain.
  • the antibodies of the invention are isolated antibodies.
  • isolated variable domain refers to an variable domain or an antibody that is substantially free of other variable domains or other antibodies having different antigenic specificities (e. g., an isolated antibody variable domain that specifically binds mesothelin is substantially free of antibody variable domains that specifically bind antigens other than mesothelin).
  • an isolated antibody variable domain or isolated antibody may be substantially free of other cellular material and/or chemicals.
  • the antibody variable domains and antibodies of the invention are monoclonal antibody variable domains and antibodies.
  • the term “monoclonal antibody variable domains” or “monoclonal antibody” as used herein refers to variable domains or antibodies that have substantially identical amino acid sequences or are derived from the same genetic source.
  • a monoclonal variable domain or antibody displays a binding specificity and affinity for a particular epitope, or binding specificities and affinities for specific epitopes.
  • the antibody variable domains and antibodies of the invention include, but are not limited to, chimeric, human and humanized antibody variable domains and antibodies.
  • chimeric antibody or “chimeric antibody variable domain”, as used herein, refers to an antibody molecule or antibody variable domain in which (a) the constant region, or a portion thereof, is altered, replaced or exchanged so that the antigen-binding site (variable region) is linked to a constant region of a different or altered class, effector function and/or species; or (b) the variable region, or a portion thereof, is altered, replaced or exchanged with a variable region having a different or altered antigen specificity.
  • a mouse antibody can be modified by replacing its constant region with the constant region from a human immunoglobulin.
  • human antibody or “human antibody variable domain” , as used herein, is intended to include antibodies or antibody variable domains having variable regions in which both the framework and CDR regions are derived from sequences of human origin. Furthermore, if the antibody or antibody variable domain contains a constant region, the constant region also is derived from such human sequences, e. g., human germline sequences, or mutated versions of human germline sequences.
  • the human antibodies and antibody variable domains of the invention may include amino acid residues not encoded by human sequences (e.
  • human antibody or antibody variable domain specifically excludes a humanized antibody or antibody variable domain comprising non-human antigen-binding residues.
  • Human antibodies and antibody variable domains can be produced using various techniques known in the art, including phage-display libraries (Hoogenboom and Winter, J. Mol. Biol, 227:381 (1992); Marks et al, J. Mol. Biol, 222:581 (1991)). Also available for the preparation of human monoclonal antibodies and human monoclonal antibody variable domains are methods described in Cole et al, Monoclonal Antibodies and Cancer Therapy, Alan R. Liss, p.
  • Human antibodies and human antibody variable domains can be prepared by administering the antigen to a transgenic animal that has been modified to produce such antibodies and antibody variable domains in response to antigenic challenge, but whose endogenous loci have been disabled, e. g., immunized xenomice (see, e. g., U.S. Pat. Nos. 6,075,181 and 6,150,584 regarding XENOMOUSETM technology). See also, for example, Li et al, Proc. Natl. Acad. Sci. USA, 103:3557- 3562 (2006) regarding human antibodies generated via a human B-cell hybridoma technology.
  • humanized antibody or “humanized” antibody variable domain refers to an antibody or antibody variable domain that retains the reactivity of a non- human antibody or antibody variable domain while being less immunogenic in humans. This can be achieved, for instance, by retaining the non-human CDR regions and replacing the remaining parts of the antibody or antibody variable domain with their human counterparts (/. e., the constant region as well as the framework portions of the variable region). Additional framework region modifications may be made within the human framework sequences as well as within the CDR sequences derived from the germline of another mammalian species.
  • the humanized antibodies and antibody variable domains of the invention may include amino acid residues not encoded by human sequences (e.
  • recombinant humanized antibody or “recombinant humanized antibody variable domain” as used herein, includes all human antibodies and human antibody variable domains that are prepared, expressed, created or isolated by recombinant means, such as antibodies and antibody variable domains isolated from a host cell transformed to express the humanized antibody or humanized antibody variable domain, e. g., from a transfectoma, and antibodies and antibody variable domains prepared, expressed, created or isolated by any other means that involve splicing of all or a portion of a human immunoglobulin gene, sequences to other DNA sequences.
  • the antibody variable domains and antibodies of the invention are humanized. More preferably, the antibody variable domains and antibodies of the invention are humanized and comprise rabbit-derived CDRs.
  • bispecific antibody refers to an antibody that binds to two or more different epitopes on at least two or more different targets, for example 2 different targets (bispecific), 3 different targets (trispecific), 4 different targets (tetraspecific), or 5 different targets (pentaspecific).
  • the antibodies of the invention are bispecific, trispecific or tetraspecific, particularly bispecific or trispecific, more particularly trispecific.
  • trispecific antibody refers to an antibody that binds to at least three different epitopes on three different targets (e. g., mesothelin, CD3 and hSA or ROR1, CD3 and hSA).
  • epitope means a protein determinant capable of specific binding to an antibody.
  • Epitopes usually consist of chemically active surface groupings of molecules such as amino acids or sugar side chains and usually have specific three-dimensional structural characteristics, as well as specific charge characteristics. “Conformational” and “linear” epitopes are distinguished in that the binding to the former but not the latter is lost in the presence of denaturing solvents.
  • formational epitope refers to amino acid residues of an antigen that come together on the surface when the polypeptide chain folds to form the native protein.
  • linear epitope refers to an epitope, wherein all points of interaction between the protein and the interacting molecule (such as an antibody) occurring linearly along the primary amino acid sequence of the protein (continuous).
  • the term “recognize” as used herein refers to an antibody or antibody variable domain that finds and interacts (e. g., binds) with its conformational epitope.
  • the antibody variable domain of the invention could be successfully applied in the construction of divers antibody fragments, e. g. scFv fragments, and multispecific antibodies, e. g. bispecific and trispecific antibodies, which exhibits significantly reduced immunogenicity, when compared to their unmodified counterparts, and which have an excellent stability.
  • divers antibody fragments e. g. scFv fragments
  • multispecific antibodies e. g. bispecific and trispecific antibodies
  • One specific group of embodiments relates to an antibody comprising one or more antibody variable domains of the present invention, wherein the antibody is trispecific and monovalent for each target antigen, and wherein the antibody comprises:
  • CD137-BD one binding domain, which specifically binds to CD137
  • PDL1-BD one binding domain, which specifically binds to PDL1
  • the one binding domain, which specifically binds to CD137 comprises a. a VH sequence of SEQ ID NO: 1 and a VL sequence of SEQ ID NO: 5; b. a VH sequence of SEQ ID NO: 2 and a VL sequence of SEQ ID NO: 5; c. a VH sequence of SEQ ID NO: 3 and a VL sequence of SEQ ID NO: 5; or d.
  • the one binding domain, which specifically binds to PDL1 comprises a. a VH sequence of SEQ ID NO: 11 and a VL sequence of SEQ ID NO: 15; b. a VH sequence of SEQ ID NO: 12 and a VL sequence of SEQ ID NO: 16; c. a VH sequence of SEQ ID NO: 13 and a VL sequence of SEQ ID NO: 15; or d. a VH sequence of SEQ ID NO: 14 and a VL sequence of SEQ ID NO: 16; the one human serum albumin binding domain (hSA-BD) comprises a.
  • the one binding domain, which specifically binds to PDL1 comprises a. a VH sequence of SEQ ID NO: 17 and a VL sequence of SEQ ID NO: 21 ; b.
  • the above definition further includes variants of said VH and VL domains, /. e. variants of SEQ ID NOs: 3, 4, 5, 13,14,15,16, 25, 26, 27 and 28 or variants of SEQ ID NOs: 8, 9, 10, 19, 20, 21 , 22, 31 , 32, 33 and 34, wherein no more than 5 amino acids, particularly no more than 4 amino acids, particularly no more than 3 amino acids, particularly no more than 2 amino acids, particularly no more than 1 amino acid within the framework regions (the non-italicized residues in Table 1) at positions different from heavy chain positions 12, 103 and 144 (AHo numbering) have been mutated, provided that the VH and VL domains selected from these variant sequences still exhibit the respective binding properties to CD3, PDL1 or hSA as well as the functional properties as defined above in item 24.
  • mutation means, as various non-limiting examples, an addition, substitution or deletion.
  • binding domain of an antibody, as used herein, or the terms “antigenbinding fragment thereof’ or “antigen-binding portion” of an antibody, and the like, refer to one or more parts of an intact antibody that have the ability to specifically bind to a given antigen. Antigen-binding functions of an antibody can be performed by fragments of an intact antibody. Specifically, in case of the antibodies of the present invention, the terms “binding domain”, as used herein, or the terms “antigen-binding fragment thereof” or “antigen-binding portion”, and the like, refer to a Fab fragment, /. e.
  • the binding domains of the antibodies of the present invention are independently of each other selected from an Fv fragment, a disulfide stabilized Fv fragment (dsFv) and a single-chain Fv fragment (scFv).
  • the binding domains of the antibodies of the present invention are independently of each other selected from an Fv fragment and a single-chain Fv fragment (scFv).
  • the VL and VH domains of the scFv fragment are stabilized by an interdomain disulfide bond, in particular said VH domain comprises a single cysteine residue in position 51 (AHo numbering) and said VL domain comprises a single cysteine residue in position 141 (AHo numbering).
  • the antibodies of said specific group of embodiments do not comprise an immunoglobulin Fc region.
  • the antibodies of said specific group of embodiments do not comprise an immunoglobulin Fc region and do also not comprises CH1 and/or CL regions.
  • binding domains are constructed from an antibody variable domain of the present invention, /. e. comprise heavy chain framework regions having the specific substitutions as defined herein for the antibody variable domains of the present invention.
  • the antibodies of this specific group of embodiments comprise VH/VL sequences as defined herein, which can be found in Table 3.
  • the antibodies of this specific group of embodiments are in a MATCH3 format, particularly have the scMATCH3 format.
  • the antibodies of this specific group of embodiments are variants of the trispecific trivalent antibody NM21-1480.
  • Specific examples are PRO2758, PRO2759, PRO2760, PRO2761, PRO2762, PRO2763, PRO2764, PRO2765 and PRO3351, whose sequences can be found in Table 3.
  • the anti-PDL1xCD137xhSA antibodies of the present invention exhibit a significantly reduced immunogenicity, /. e. exhibit a significantly reduced binding to pre-existing ADAs present in human serum samples of healthy untreated individuals when compared to NM21-1480, which does not comprise the substitutions as defined in item 1.
  • the assay used for determining the binding of ADAs in said serum samples to the anti-PDL1xCD137xhSA antibodies of the present invention, the NM21-1480 variants and NM21-1480 is described in detail in Example 3.
  • these fragment-based anti-PDL1xCD137xhSA antibodies have advantageous biophysical properties, in particular excellent stability.
  • the present invention relates to an antibody variable domain as defined herein, wherein said antibody variable domain specifically binds to human serum albumin.
  • serum albumin refers in particular to human serum albumin with UniProt ID number P02768 or a variant thereof.
  • Human serum albumin (herein abbreviated as hSA) is a 66.4 kDa abundant protein in human serum (50 % of total protein) comprised of 585 amino acids (Sugio, Protein Eng, Vol. 12, 1999, 439-446).
  • the structure of multifunctional hSA protein allows to bind and transport a number of metabolites such as fatty acids, metal ions, bilirubin and some drugs (Fanali, Molecular Aspects of Medicine, Vol. 33, 2012, 209- 290).
  • HSA concentration in serum is around 3.5-5 g/dl.
  • Said hSA binding antibody variable domains of the present invention may thus be used, for example, to extend the in vivo serum half-life of drugs or proteins conjugated thereto.
  • said antibody variable domain that specifically binds to human serum albumin comprises a) a VH domain selected from any one of the SEQ ID NOs: 35, 36 and 37, and from variants of SEQ ID NOs: 35, 36 and 37, wherein no more than 5 amino acids, particularly no more than 4 amino acids, particularly no more than 3 amino acids, particularly no more than 2 amino acids, particularly no more than 1 amino acid within the framework regions (the non-italicized residues in Table 2) at positions different from 12, 103 and 144 (AHo numbering) have been mutated, provided that the VH domains selected from these variant sequences exhibit the functional features defined above in any one of the items 29 to 31 ; and b) a VL domain selected from any one of the SEQ ID NOs: 38, 39 and 40, and from variants of SEQ ID NOs: 38, 39 and 40, wherein no more than 5 amino acids, particularly no more than 4 amino acids, particularly no more than 3 amino acids, particularly no more than 2 amino acids, particularly no more
  • said antibody variable domain that specifically binds to human serum albumin comprises a) a VH domain selected from any one of the SEQ ID NOs: 35, 36 and 37, and from variants of SEQ ID NOs: 35, 36 and 37, wherein no more than 5 amino acids, particularly no more than 4 amino acids, particularly no more than 3 amino acids, particularly no more than 2 amino acids, particularly no more than 1 amino acid within the framework regions (the non-italicized residues in Table 2) at positions different from 12, 103 and 144 (AHo numbering) have been mutated, provided that the VH domains selected from these variant sequences exhibit the functional features defined above in any one of the items 29 to 31 ; and b) a VL domain selected from any one of the SEQ ID NOs: 38, 39 and 40, and from variants of SEQ ID NOs: 38, 39 and 40, wherein no more than 5 amino acids, particularly no more than 4 amino acids, particularly no more than 3 amino acids, particularly no more than 2 amino acids, particularly no
  • VH and VL regions further include VH and VL domains comprising at least positions 5 to 140 (AHo numbering), particularly at least positions 3 to 145, more particularly at least positions 2 to 147 of one of the sequences shown in the SEQ ID NOs: 35, 36 and 37, provided that such VL domains exhibit the functional features defined above in items 9 and 10.
  • said antibody variable domain that specifically binds to human serum albumin comprises a) a VH sequence of SEQ ID NO: 35 and a VL sequence of SEQ ID NO: 38; b) a VH sequence of SEQ ID NO: 36 and a VL sequence of SEQ ID NO: 39; c) a VH sequence of SEQ ID NO: 36 and a VL sequence of SEQ ID NO: 41 ; d) a VH sequence of SEQ ID NO: 37 and a VL sequence of SEQ ID NO: 40; e) a VH sequence of SEQ ID NO: 42 and a VL sequence of SEQ ID NO: 45; f) a VH sequence of SEQ ID NO: 43 and a VL sequence of SEQ ID NO: 46; g) a VH sequence of SEQ ID NO: 43 and a VL sequence of SEQ ID NO: 48; or h) a VH sequence of SEQ ID NO: 44 and a VL sequence of SEQ ID NO:
  • this hSA-binding antibody variable domain of the present invention is cross-reactive to other species.
  • the antibody variable domains of the invention are cross-reactive to Cynomolgus (Macaca fascicularis) serum albumin (herein abbreviated as cSA) and mouse (Mus musculus) serum albumin (herein abbreviated as mSA).
  • the hSA-binding antibody variable domain of the present invention when being in scFv format, is further characterized by the following parameters: a. binds to human serum albumin (hSA) with a monovalent dissociation constant (KD) of less than 10 nM, particularly with a KD of 0.05 to 10 nM, particularly of 0.05 to 5 nM, at a pH value of 5.5, as measured by surface plasmon resonance (SPR); b.
  • KD monovalent dissociation constant
  • cSA Macaca fascicularis
  • mSA Mus musculus serum albumin
  • the hSA-binding antibody variable domain of the present invention when being in scFv format, is further characterized by the following parameters: a. binds to human serum albumin (hSA) with a monovalent dissociation constant (KD) of less than 10 nM, particularly with a KD of 0.05 to 10 nM, particularly of 0.05 to 5 nM, at a pH value of 5.5, as measured by surface plasmon resonance (SPR); b.
  • KD monovalent dissociation constant
  • cSA Macaca fascicularis
  • mSA Mus musculus serum albumin
  • Tm melting temperature
  • DSF differential scanning fluorimetry
  • hSA-BD has a loss in protein content, after storage for 28 days, at 4°C, of less than 2 %, preferably less than 1 %, when said antigen-binding fragment is at a starting concentration of 10 mg/ml, and wherein said hSA-BD is formulated in 50 mM phosphate citrate buffer with 150 mM NaCI at pH 6.4; g. has a loss in protein content, after storage for 28 days, at 40°C, of less than 2 %, preferably less than 1 %, when said antigen-binding fragment is at a starting concentration of 10 mg/ml, and wherein said hSA-BD is formulated in 50 mM phosphate citrate buffer with 150 mM NaCI at pH 6.4; and/or h.
  • hSA-BD has a loss in monomer content, after storage for 14 days at 4°C, of less than 2 %, preferably less than 1 %, when said antigen-binding fragment is at a starting concentration of 50 mg/ml, and wherein said hSA-BD is formulated in 50 mM phosphate citrate buffer with 150 mM NaCI at pH 6.4.
  • the hSA-binding antibody variable domain of the present invention when being in scFv format, is further characterized by the following parameters: a. binds to human serum albumin (hSA) with a monovalent dissociation constant (KD) of less than 10 nM, particularly with a KD of 0.05 to 10 nM, particularly of 0.05 to 5 nM, at a pH value of 5.5, as measured by surface plasmon resonance (SPR); b.
  • KD monovalent dissociation constant
  • cSA Macaca fascicularis
  • mSA Mus musculus serum albumin
  • hSA human serum albumin
  • KD monovalent dissociation constant
  • cSA Macaca fascicularis
  • cSA monovalent KD of less than 10 nM, particularly with a KD of 0.05 to 10 nM, particularly of 0.05 to 5 nM, at a pH value of 7.4, as measured by SPR; and/or f.
  • mSA Mus musculus serum albumin
  • the hSA-binding antibody variable domain of the present invention is further characterized by the following parameters: a. binds to human serum albumin (hSA) with a monovalent dissociation constant (KD) of less than 10 nM, particularly with a KD of 0.05 to 10 nM, particularly of 0.05 to 5 nM, at a pH value of 5.5, as measured by surface plasmon resonance (SPR); b.
  • KD monovalent dissociation constant
  • cSA Macaca fascicularis
  • mSA Mus musculus serum albumin
  • hSA human serum albumin
  • KD monovalent dissociation constant
  • cSA Macaca fascicularis
  • cSA monovalent KD of less than 10 nM, particularly with a KD of 0.05 to 10 nM, particularly of 0.05 to 5 nM, at a pH value of 7.4, as measured by SPR; and f.
  • mSA Mus musculus
  • mSA Mus musculus serum albumin
  • g. has a melting temperature (Tm), determined by differential scanning fluorimetry (DSF), of at least 70°C, preferably at least 75°C, wherein said hSA-BD is formulated in 50 mM phosphate citrate buffer with 150 mM NaCI at pH 6.4; h.
  • hSA-BD has a loss in monomer content, after storage for 28 days at 4°C, of less than 2 %, preferably less than 1 %, when said antigen-binding fragment is at a starting concentration of 10 mg/ml, and wherein said hSA-BD is formulated in 50 mM phosphate citrate buffer with 150 mM NaCI at pH 6.4; i. has a loss in protein content, after storage for 28 days, at 4°C, of less than 2 %, preferably less than 1 %, when said antigen-binding fragment is at a starting concentration of 10 mg/ml, and wherein said hSA-BD is formulated in 50 mM phosphate citrate buffer with 150 mM NaCI at pH 6.4; j.
  • hSA-BD has a loss in protein content, after storage for 28 days, at 40°C, of less than 2 %, preferably less than 1 %, when said antigen-binding fragment is at a starting concentration of 10 mg/ml, and wherein said hSA-BD is formulated in 50 mM phosphate citrate buffer with 150 mM NaCI at pH 6.4; and/or k. has a loss in monomer content, after storage for 14 days at 4°C, of less than 2 %, preferably less than 1 %, when said antigen-binding fragment is at a starting concentration of 50 mg/ml, and wherein said hSA-BD is formulated in 50 mM phosphate citrate buffer with 150 mM NaCI at pH 6.4.
  • affinity refers to the strength of interaction between the antibody or the antibody variable domain and the antigen at single antigenic sites. Within each antigenic site, the variable region of the antibody variable domain or the antibody “arm” interacts through weak non-covalent forces with antigen at numerous sites; the more interactions, the stronger the affinity.
  • Binding affinity generally refers to the strength of the total sum of non-covalent interactions between a single binding site of a molecule (e. g., of an antibody or an antibody variable domain) and its binding partner (e. g., an antigen or, more specifically, an epitope on an antigen).
  • binding affinity refers to intrinsic binding affinity that reflects a 1 :1 interaction between members of a binding pair (e. g., an antibody variable domain and an antigen).
  • the affinity of a molecule X for its partner Y can generally be represented by the dissociation constant (KD).
  • Affinity can be measured by common methods known in the art, including those described herein. Low-affinity antibodies and antibody variable domains generally bind antigens slowly and tend to dissociate readily, whereas high-affinity antibodies generally bind antigens faster and tend to remain bound longer. A variety of methods of measuring binding affinity are known in the art, any of which can be used for purposes of the present invention. Specific illustrative and exemplary embodiments for measuring binding affinity, /. e. binding strength are described in the following.
  • K asS oc “K a ” or “K on ”, as used herein, are intended to refer to the association rate of a particular antibody-antigen interaction
  • Kdis “Kd” or “Koff”, as used herein, is intended to refer to the dissociation rate of a particular antibodyantigen interaction
  • KD is intended to refer to the dissociation constant, which is obtained from the ratio of Kd to K a (/. e. Kd/K a ) and is expressed as a molar concentration (M).
  • M molar concentration
  • the “KD” or “KD value” or "KD” or “KD value” according to this invention is in one embodiment measured by using surface plasmon resonance assays.
  • the hSA-binding antibody variable domains of the present invention are selected from the group consisting of: a Fab, an Fv, a dsFv and an scFv.
  • the hSA-binding antibody variable domains of the present invention when being in scFv format, exhibit a significantly reduced immunogenicity, /. e. exhibit a significantly reduced binding to pre-existing ADAs present in human serum samples, when compared to the corresponding hSA-binding scFvs that do not comprise the framework substitutions as defined above in item 1.
  • the assay used for determining the binding of ADAs in said serum samples to said hSA-binding scFvs is described in detail in Example 3.
  • the antibody variable domains and antibodies of the invention can be produced using any convenient antibody-manufacturing method known in the art (see, e. g., Fischer, N. & Leger, O., Pathobiology 74 (2007) 3-14 with regard to the production of bispecific constructs; Hornig, N. & Farber-Schwarz, A., Methods Mol. Biol. 907 (2012)713-727, and WO 99/57150 with regard to bispecific diabodies and tandem scFvs).
  • suitable methods for the preparation of the multispecific constructs further include, inter alia, the Genmab (see Labrijn et al., Proc. Natl. Acad. Sci.
  • the antibodies of the invention that are multispecific, e.g. bispecific, trispecific, tetraspecific or pentaspecific, and/or multivalent, can be prepared by conjugating the constituent binding specificities, using methods known in the art. For example, each binding specificity of these antibodies can be generated separately and then conjugated to one another. When the binding specificities are proteins or peptides, a variety of coupling or cross-linking agents can be used for covalent conjugation.
  • cross-linking agents examples include protein A, carbodiimide, N-succinimidyl-5-acetyl-thioacetate (SATA), 5,5'-dithiobis (2-nitrobenzoic acid) (DTNB), o-phenylenedimaleimide (oPDM), N-succinimidyl-3-(2- pyridyldithio)propionate (SPDP), and sulfosuccinimidyl 4-(N-maleimidomethyl)-cyclohexane- 1-carboxylate (sulfo-SMCC) (see e. g., Karpovsky et al., 1984 J. Exp. Med.
  • two or more binding specificities can be encoded in the same vector and expressed and assembled in the same host cell.
  • This method is particularly useful where the bispecific molecule is a mAb x Fab, a mAb x scFv, a mAb x dsFv or a mAb x Fv fusion protein.
  • Methods for preparing multispecific and/or multivalent antibodies and molecules are described for example in US 5,260,203; US 5,455,030; US 4,881,175; US 5,132,405; US 5,091 ,513; US 5,476,786; US 5,013,653; US 5,258,498; and US 5,482,858.
  • binding of the antibody variable domains and multispecific antibodies to their specific targets can be confirmed by, for example, enzyme-linked immunosorbent assay (ELISA), radioimmunoassay (REA), FACS analysis, bioassay (e. g., growth inhibition), or Western Blot assay.
  • ELISA enzyme-linked immunosorbent assay
  • REA radioimmunoassay
  • FACS fluorescence-activated cell sorting
  • bioassay e. g., growth inhibition
  • Western Blot assay Western Blot assay.
  • Each of these assays generally detects the presence of proteinantibody complexes of particular interest by employing a labeled reagent (e. g., an antibody) specific for the complex of interest.
  • a labeled reagent e. g., an antibody
  • the invention provides a nucleic acid or two nucleic acids encoding the antibody variable domain or the antibody of the invention. Such nucleic acids can be optimized for expression in mammalian cells
  • nucleic acid is used herein interchangeably with the term “polynucleotide(s)” and refers to one or more deoxyribonucleotides or ribonucleotides and polymers thereof in either single- or double-stranded form.
  • the term encompasses nucleic acids containing known nucleotide analogs or modified backbone residues or linkages, which are synthetic, naturally occurring, and non-naturally occurring, which have similar binding properties as the reference nucleic acid, and which are metabolized in a manner similar to the reference nucleotides.
  • Examples of such analogs include, without limitation, phosphorothioates, phosphoramidates, methyl phosphonates, chiral-methyl phosphorates, 2-O-methyl ribonucleotides, peptide-nucleic acids (PNAs).
  • PNAs peptide-nucleic acids
  • a particular nucleic acid sequence also implicitly encompasses conservatively modified variants thereof (e. g., degenerate codon substitutions) and complementary sequences, as well as the sequence explicitly indicated.
  • degenerate codon substitutions may be achieved by generating sequences in which the third position of one or more selected (or all) codons is substituted with mixed-base and/or deoxyinosine residues (Batzer et al., Nucleic Acid Res. 19:5081, 1991 ; Ohtsuka et al., J. Biol. Chem. 260:2605- 2608, 1985; and Rossolini et al., Mol. Cell. Probes 8:91-98, 1994).
  • the invention provides substantially purified nucleic acid molecules which encode polypeptides comprising segments or domains of the antibody variable domains or the antibodies described above. When expressed from appropriate expression vectors, polypeptides encoded by these nucleic acid molecules are capable of exhibiting antigenbinding capacities of the antibody variable domains or the antibodies of the present invention.
  • the polynucleotide sequences can be produced by de novo solid-phase DNA synthesis or by PCR mutagenesis of an existing sequence (e. g., sequences as described in the Examples below) encoding the antibody variable domain or the antibody of the invention.
  • Direct chemical synthesis of nucleic acids can be accomplished by methods known in the art, such as the phosphotriester method of Narang et al., 1979, Meth. Enzymol. 68:90; the phosphodiester method of Brown et al., Meth. Enzymol.
  • Also provided in the invention are expression vectors and host cells for producing the antibody variable domain or the antibody of the invention.
  • vector is intended to refer to a polynucleotide molecule capable of transporting another polynucleotide to which it has been linked.
  • plasmid refers to a circular double stranded DNA loop into which additional DNA segments may be ligated.
  • viral vector Another type of vector is a viral vector, wherein additional DNA segments may be ligated into the viral genome.
  • Certain vectors are capable of autonomous replication in a host cell into which they are introduced (e. g., bacterial vectors having a bacterial origin of replication and episomal mammalian vectors). Other vectors (e. g., non- episomal mammalian vectors) can be integrated into the genome of a host cell upon introduction into the host cell, and thereby are replicated along with the host genome.
  • vectors are capable of directing the expression of genes to which they are operatively linked. Such vectors are referred to herein as “recombinant expression vectors” (or simply, “expression vectors”).
  • expression vectors of utility in recombinant DNA techniques are often in the form of plasmids.
  • plasmid and vector may be used interchangeably as the plasmid is the most commonly used form of vector.
  • the invention is intended to include such other forms of expression vectors, such as viral vectors (e. g., replication defective retroviruses, adenoviruses and adeno- associated viruses), which serve equivalent functions.
  • operably linked refers to a functional relationship between two or more polynucleotide (e. g., DNA) segments. Typically, it refers to the functional relationship of a transcriptional regulatory sequence to a transcribed sequence.
  • a promoter or enhancer sequence is operably linked to a coding sequence if it stimulates or modulates the transcription of the coding sequence in an appropriate host cell or other expression system.
  • promoter transcriptional regulatory sequences that are operably linked to a transcribed sequence are physically contiguous to the transcribed sequence, /. e., they are cis-acting.
  • some transcriptional regulatory sequences, such as enhancers need not be physically contiguous or located in close proximity to the coding sequences whose transcription they enhance.
  • Non-viral vectors and systems include plasmids, episomal vectors, typically with an expression cassette for expressing a protein or RNA, and human artificial chromosomes (see, e. g., Harrington et al., Nat Genet. 15:345, 1997).
  • non-viral vectors useful for expression of the hSA-binding polypeptides, or of polynucleotides encoding such polypeptides, in mammalian e.
  • g., human cells include pThioHis A, B and C, pcDNA3.1/His, pEBVHis A, B and C, (Invitrogen, San Diego, Calif.), MPS V vectors, and numerous other vectors known in the art for expressing other proteins.
  • Useful viral vectors include vectors based on retroviruses, adenoviruses, adeno-associated viruses, herpes viruses, vectors based on SV40, papilloma virus, HBP Epstein Barr virus, Vaccinia virus vectors and Semliki Forest virus (SFV). See, Brent et al., supra; Smith, Annu. Rev. Microbiol. 49:807, 1995; and Rosenfeld et al., Cell 68: 143, 1992.
  • the choice of expression vector depends on the intended host cells in which the vector is to be expressed.
  • the expression vectors contain a promoter and other regulatory sequences (e. g., enhancers) that are operably linked to the polynucleotides encoding a multispecific antibody chain or a variable domain.
  • an inducible promoter is employed to prevent expression of inserted sequences except under inducing conditions.
  • Inducible promoters include, e. g., arabinose, lacZ, metallothionein promoter or a heat shock promoter. Cultures of transformed organisms can be expanded under non-inducing conditions without biasing the population for coding sequences whose expression products are better tolerated by the host cells.
  • promoters In addition to promoters, other regulatory elements may also be required or desired for efficient expression of a multispecific antibody chain or a variable domain. These elements typically include an ATG initiation codon and adjacent ribosome binding site or other sequences. In addition, the efficiency of expression may be enhanced by the inclusion of enhancers appropriate to the cell system in use (see, e. g., Scharf et al., Results Probl. Cell Differ. 20: 125, 1994; and Bittner et al., Meth. Enzymol., 153:516, 1987). For example, the SV40 enhancer or CMV enhancer may be used to increase expression in mammalian host cells.
  • Vectors to be used typically encode the antibody variable domain or the antibody light and heavy chain including constant regionsor parts thereof, if present. Such vectors allow expression of the variable regions as fusion proteins with the constant regions thereby leading to production of intact antibodies and antibody variable domains thereof. Typically, such constant regions are human.
  • the term “recombinant host cell” refers to a cell into which a recombinant expression vector has been introduced. It should be understood that such terms are intended to refer not only to the particular subject cell but to the progeny of such a cell. Because certain modifications may occur in succeeding generations due to either mutation or environmental influences, such progeny may not, in fact, be identical to the parent cell, but are still included within the scope of the term “host cell” as used herein.
  • the host cells for harboring and expressing the antibody variable domain or the antibody of the invention can be either prokaryotic or eukaryotic. E.
  • coli is one prokaryotic host useful for cloning and expressing the polynucleotides of the present invention.
  • Other microbial hosts suitable for use include bacilli, such as Bacillus subtilis, and other enterobacteriaceae, such as Salmonella, Serratia, and various Pseudomonas species.
  • bacilli such as Bacillus subtilis
  • enterobacteriaceae such as Salmonella, Serratia, and various Pseudomonas species.
  • prokaryotic hosts one can also make expression vectors, which typically contain expression control sequences compatible with the host cell (e. g., an origin of replication).
  • any number of a variety of well-known promoters will be present, such as the lactose promoter system, a tryptophan (trp) promoter system, a beta-lactamase promoter system, or a promoter system from phage lambda.
  • the promoters typically control expression, optionally with an operator sequence, and have ribosome binding site sequences and the like, for initiating and completing transcription and translation.
  • Other microbes, such as yeast can also be employed to express the antibody variable domain or multispecific antibodies of the invention. Insect cells in combination with baculovirus vectors can also be used.
  • mammalian host cells are used to express and produce the antibody variable domain or the antibody of the invention.
  • they can be either a hybridoma cell line expressing endogenous immunoglobulin genes or a mammalian cell line harboring an exogenous expression vector.
  • These include any normal mortal or normal or abnormal immortal animal or human cell.
  • suitable host cell lines capable of secreting intact immunoglobulins have been developed including the CHO cell lines, various COS cell lines, HeLa cells, myeloma cell lines, transformed B-cells and hybridomas.
  • the use of mammalian tissue cell culture to express polypeptides is discussed generally in, e.
  • Expression vectors for mammalian host cells can include expression control sequences, such as an origin of replication, a promoter, and an enhancer (see, e. g., Queen, et al., Immunol. Rev. 89:49-68, 1986), and necessary processing information sites, such as ribosome binding sites, RNA splice sites, polyadenylation sites, and transcriptional terminator sequences.
  • expression control sequences such as an origin of replication, a promoter, and an enhancer (see, e. g., Queen, et al., Immunol. Rev. 89:49-68, 1986)
  • necessary processing information sites such as ribosome binding sites, RNA splice sites, polyadenylation sites, and transcriptional terminator sequences.
  • These expression vectors usually contain promoters derived from mammalian genes or from mammalian viruses.
  • Suitable promoters may be constitutive, cell type-specific, stage-specific, and/or modulatable or regulatable.
  • Useful promoters include, but are not limited to, the metallothionein promoter, the constitutive adenovirus major late promoter, the dexamethasone-inducible MMTV promoter, the SV40 promoter, the MRP poll II promoter, the constitutive MPS V promoter, the tetracycline-inducible CMV promoter (such as the human immediate-early CMV promoter), the constitutive CMV promoter, and promoter-enhancer combinations known in the art. [0149] Methods for introducing expression vectors containing the polynucleotide sequences of interest vary depending on the type of cellular host.
  • calcium chloride transfection is commonly utilized for prokaryotic cells, whereas calcium phosphate treatment or electroporation may be used for other cellular hosts.
  • calcium phosphate treatment or electroporation may be used for other cellular hosts.
  • cell lines which stably express the antibody variable domain or the antibody of the invention can be prepared using expression vectors of the invention which contain viral origins of replication or endogenous expression elements and a selectable marker gene. Following the introduction of the vector, cells may be allowed to grow for 1 to 2 days in an enriched media before they are switched to selective media.
  • the purpose of the selectable marker is to confer resistance to selection, and its presence allows growth of cells which successfully express the introduced sequences in selective media.
  • Resistant, stably transfected cells can be proliferated using tissue culture techniques appropriate to the cell type.
  • the present invention thus provides a method of producing the antibody variable domain or the antibody of the invention, wherein said method comprises the step of culturing a host cell comprising a nucleic acid or a vector encoding the antibody variable domain or the antibody of the invention, whereby said antibody variable domain or said antibody of the disclosure is expressed.
  • the present invention relates to a method of producing the antibody variable domain or the antibody of the invention, the method comprising the step of culturing a host cell expressing a nucleic acid or two nucleic acids encoding the antibody variable domain or the antibody of the invention.
  • the present invention relates to a method of producing the antibody variable domain or the antibody of the invention, the method comprising (i) providing a nucleic acid or two nucleic acids encoding the antibody variable domain or the antibody of the invention or one or two vectors encoding the antibody variable domain or the antibody of the invention, expressing said nucleic acid or nucleic acids, or said vector or vectors, and collecting said antibody variable domain or said antibody from the expression system, or (ii) providing a host cell or host cells expressing a nucleic acid or two nucleic acids encoding the antibody variable domain or the antibody of the invention, culturing said host cell or said host cells; and collecting said antibody variable domain or said multispecific antibody from the cell culture.
  • the present invention relates to a pharmaceutical composition
  • a pharmaceutical composition comprising the antibody of the invention, and a pharmaceutically acceptable carrier.
  • “Pharmaceutically acceptable carrier” means a medium or diluent that does not interfere with the structure of the antibodies.
  • Pharmaceutically acceptable carriers enhance or stabilize the composition, or facilitate preparation of the composition.
  • Pharmaceutically acceptable carriers include solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, and the like that are physiologically compatible.
  • compositions enable pharmaceutical compositions to be formulated as, for example, tablets, pills, dragees, capsules, liquids, gels, syrups, slurries, suspension and lozenges for the oral ingestion by a subject.
  • Certain of such carriers enable pharmaceutical compositions to be formulated for injection, infusion or topical administration.
  • a pharmaceutically acceptable carrier can be a sterile aqueous solution.
  • compositions in accordance with the present disclosure may further routinely contain pharmaceutically acceptable concentrations of salt, buffering agents, preservatives, supplementary immune potentiating agents such as adjuvants and cytokines and optionally other therapeutic agents.
  • the composition may also include antioxidants and/or preservatives.
  • antioxidants may be mentioned thiol derivatives (e. g. thioglycerol, cysteine, acetylcysteine, cystine, dithioerythreitol, dithiothreitol, glutathione), tocopherols, butylated hydroxyanisole, butylated hydroxytoluene, sulfurous acid salts (e. g.
  • Suitable preservatives may for instance be phenol, chlorobutanol, benzylalcohol, methyl paraben, propyl paraben, benzalkonium chloride and cetylpyridinium chloride.
  • the pharmaceutical composition of the invention can be administered by a variety of methods known in the art.
  • the route and/or mode of administration vary depending upon the desired results. Administration can be intravenous, intramuscular, intraperitoneal, or subcutaneous, or administered proximal to the site of the target.
  • the pharmaceutically acceptable carrier should be suitable for intravenous, intramuscular, subcutaneous, parenteral, spinal or epidermal administration (e. g., by injection or infusion).
  • the active compound, /. e., the antibody of the invention may be coated in a material to protect the compound from the action of acids and other natural conditions that may inactivate the compound.
  • compositions of the invention can be prepared in accordance with methods well known and routinely practiced in the art. See, e. g., Remington: The Science and Practice of Pharmacy, Mack Publishing Co., 20th ed., 2000; and Sustained and Controlled Release Drug Delivery Systems, J. R. Robinson, ed., Marcel Dekker, Inc., New York, 1978. Pharmaceutical compositions are preferably manufactured under GMP conditions. Typically, a therapeutically effective dose or efficacious dose of the antibody of the invention is employed in the pharmaceutical compositions of the invention.
  • the antibodies of the invention are formulated into pharmaceutically acceptable dosage forms by conventional methods known to those of skill in the art. Dosage regimens are adjusted to provide the optimum desired response (e.
  • Dosage unit form refers to physically discrete units suited as unitary dosages for the subjects to be treated; each unit contains a predetermined quantity of active compound calculated to produce the desired therapeutic effect in association with the required pharmaceutical carrier.
  • compositions of the invention can be varied so as to obtain an amount of the active ingredient which is effective to achieve the desired therapeutic response for a particular patient, composition, and mode of administration, without being toxic to the patient.
  • the selected dosage level depends upon a variety of pharmacokinetic factors including the activity of the particular compositions of the present invention employed, or the ester, salt or amide thereof, the route of administration, the time of administration, the rate of excretion of the particular compound being employed, the duration of the treatment, other drugs, compounds and/or materials used in combination with the particular compositions employed, the age, sex, weight, condition, general health and prior medical history of the patient being treated, and like factors.
  • the antibody of the invention is usually administered on multiple occasions. Intervals between single dosages can be weekly, monthly or yearly. Intervals can also be irregular as indicated by measuring blood levels of the multispecific antibody of the invention in the patient. Alternatively, the antibody of the invention can be administered as a sustained release formulation, in which case less frequent administration is required. Dosage and frequency vary depending on the half-life of the antibody in the patient. In general, humanized antibodies show longer half-life than that of chimeric antibodies and non-human antibodies. The dosage and frequency of administration can vary depending on whether the treatment is prophylactic or therapeutic. In prophylactic applications, a relatively low dosage is administered at relatively infrequent intervals over a long period of time. Some patients continue to receive treatment for the rest of their lives. In therapeutic applications, a relatively high dosage at relatively short intervals is sometimes required until progression of the disease is reduced or terminated, and preferably until the patient shows partial or complete amelioration of symptoms of disease. Thereafter, the patient can be administered a prophylactic regime.
  • the present invention relates to the antibody of the invention or the pharmaceutical composition of the invention for use as a medicament.
  • the present invention provides the multispecific antibody or the pharmaceutical composition for use in the treatment of a proliferative disease, such as cancer, or a disease selected from allergic, inflammatory and autoimmune diseases.
  • the present invention provides the pharmaceutical composition of the invention for use in the manufacture of a medicament for the treatment of a proliferative disease, such as cancer, or a disease selected from allergic, inflammatory and autoimmune diseases.
  • the present invention relates to the use of the antibody or the pharmaceutical composition of the present invention for treating a proliferative disease, such as cancer, or a disease selected from allergic, inflammatory and autoimmune diseases, in a subject in need thereof.
  • a proliferative disease such as cancer
  • a disease selected from allergic, inflammatory and autoimmune diseases in a subject in need thereof.
  • the present invention relates to a method of treating a subject comprising administering to the subject a therapeutically effective amount of the antibody of the present invention.
  • the present invention relates to a method for the treatment of a proliferative disease, such as cancer, or a disease selected from allergic, inflammatory and autoimmune diseases, in a subject comprising administering to the subject a therapeutically effective amount of the antibody of the present invention.
  • subject includes human and non-human animals.
  • mammals include all vertebrates, e. g., non-human mammals and nonmammals, such as non-human primates, sheep, dog, cow, chickens, amphibians, and reptiles. Except when noted, the terms “patient” or “subject” are used herein interchangeably.
  • treatment refers to obtaining a desired pharmacologic and/or physiologic effect.
  • the effect may be therapeutic in terms of a partial or complete cure for a disease and/or adverse effect attributable to the disease or delaying the disease progression.
  • Treatment covers any treatment of a disease in a mammal, e. g., in a human, and includes: (a) inhibiting the disease, /. e., arresting its development; and (b) relieving the disease, /. e., causing regression of the disease.
  • terapéuticaally effective amount refers to the amount of an agent that, when administered to a mammal or other subject for treating a disease, is sufficient to affect such treatment for the disease.
  • the “therapeutically effective amount” will vary depending on the agent, the disease and its severity and the age, weight, etc., of the subject to be treated.
  • the present invention relates to a method for modifying an antibody, where the antibody is fragment-based or is an antibody comprising one or more scFv fragments, the method comprises the step of introducing the following substitutions (AHo numbering) in the VH sequence(s) of said fragment-based antibody or in the VH sequence(s) of the scFv fragment(s) of said antibody:
  • T threonine
  • Q glutamine
  • modified antibody exhibits a decreased binding to pre-existing anti-drug- antibodies (ADAs) present in human sera from healthy donors when compared to its unmodified version, and wherein the decrease in binding is determined by an ELISA- based pre-existing anti-drug-antibody binding assay.
  • ADAs pre-existing anti-drug- antibodies
  • said modified antibody comprises an antibody variable domain in accordance with the present invention, /. e. as defined in the claims, in items 1 to 32 or in the detailed description of the invention.
  • Sequence listing (residues designated according to AHo numbering scheme; the CDRs defined according to Numab CDR definition, unless specified otherwise)
  • VH/VL sequences of NM21 -1480 variants according to the present invention and reference VH/VL sequences of unmodified NM21-1480 variants (modifications relative to NM21-1480 references are shown in bold; CDR residues are shown in bold and italic letters).
  • NM21-1480 reference and examples of NM21 -1480 variants according to the present invention (linker residues and modifications relative to unmodified NM21-1480 are shown in bold).
  • VH/VL sequences of NM26 variants according to the present invention and reference VH/VL sequences of unmodified NM26 variants (modifications relative to unmodified NM26 and linker residues, if present, are shown in bold; CDR residues in the VH/VL sequences are shown in bold and italic letters).
  • VH/VL sequences ofNM28 variants according to the present invention and reference VH/VL sequences of unmodified NM28 references (modifications relative to NM28 references and linker residues, if present, are shown in bold; CDR residues in the VH/VL sequences are shown in bold and italic letters).
  • VH/VL sequences ofNM32 variants according to the present invention and reference VH/VL sequences of unmodified NM32 references (modifications relative to NM32 references and linker residues, if present, are shown in bold; CDR residues in the VH/VL sequences are shown in bold and italic letters).
  • Example 1 Manufacturing of scFv variants according to the present invention
  • PRO1922 The manufacturing as well as the functional and biophysical char acterization of PRO1922 are disclosed in detail in the patent application PCT/EP2021/064427.
  • PRO2230 is a scFv of the PD-L1 binding domain of the multispecific anti- PDL1xCD137xhSA antibody PRO1480.
  • the design, characterization and manufacturing of PRO1480 and its binding domains is disclosed in detail in the patent application WO 2019/072868.
  • Proteins were purified from clarified harvest by affinity chromatography. If necessary, variant scFvs were polished by SE-chromatography to a final monomeric content > 95 %.
  • standard analytical methods such as SE-HPLC, U 280 and SDS- PAGE were applied.
  • PRO2155 and PRO2317 are the scFvs of the hSA binding domain present in the multispecific anti-MSLNxCD3xhSA antibodies PRO2576 and PRO2660, and the multispecific anti-ROR1xCD3xhSA antibodies PRO2510, PRO2589, PRO2658 and PRO2659.
  • the variants of PRO2155 and PRO2317 according to the present invention are the scFvs of the hSA binding domain present in the anti-MSLNxCD3xhSA and anti-ROR1xCD3xhSA multispecific antibody variants PRO2741, PRO2745, PRO2746, PRO2667, PRO2668, PRO2669 and PRO2670.
  • PRO2155 and PRO2317 are disclosed in detail in the patent application WO/2021/089609.
  • variants of PRO2155 and PRO2317 of the present invention have been produced according to the methods described in said patent applications and above in section 1.1.
  • NM21-1480 variants of the present invention have been produced according to the methods described therein. [0182] Briefly, the expression of the NM21-1480 variants (scMATCH3 constructs) as listed in Table 11 has been performed at 0.5 L scale using CHOgro expression kit (Mirus) and mammalian CHO-S cells.
  • Protein A Purified from clarified culture supernatants by Protein A (MabSelect PrismA, Cytiva) affinity chromatography either followed by size exclusion chromatography (SEC) in 50 mM phosphate-citrate buffer with 300 mM sucrose at pH 6.5 or, where applicable, capture fractions with >95% purity were directly pooled and buffer exchanged to 50 mM phosphate-citrate buffer with 300 mM sucrose at pH 6.5 buffer. Monomeric content of SEC fractions was assessed by SE-HPLC analysis and fractions with a monomeric content >95% were pooled. For quality control of the manufactured material, standard analytical methods such as SE-HPLC, UV280 and SDS- PAGE were applied. Molecule composition and a manufacture summary of scMATCH3 molecules are shown in Table 2. Thermal stability of selected molecules has been assessed by nDSF using Prometheus NT.48 device (NanoTemper) as also summarized in Table 12.
  • NM32 variants scMATCH3 and MATCH4 constructs
  • Table 13 the expression of the NM32 variants (scMATCH3 and MATCH4 constructs) as listed in Table 13 has been performed at 1 L scale at Evitria AG (Schlieren, Switzerland) using their proprietary mammalian expression system.
  • Proteins were purified from clarified culture supernatants by Protein L (CaptoL, Cytiva) affinity chromatography followed by SEC in 50 mM phosphate-citrate buffer with 300 mM sucrose at pH 6.5. Monomeric content of SEC fractions was assessed by SE-HPLC analysis and fractions with a monomeric content >95% were pooled.
  • standard analytical methods such as SE-HPLC, UV280 and SDS-PAGE were applied.
  • NM21-1480 variants of the present invention NM21-1480 variants of the present invention.
  • able 12 Manufacture summary table of NM21-1480 variants including thermal stability by nDSF
  • a method was developed at Numab to detect pre-existing anti-drug-antibodies in human serum, using a direct assay format.
  • 96 well half-area plates were coated with 100 ng/ml of the test molecule (MATCH3 or scFv format) for 2 hours at room temperature. The plates were blocked for 1 hour with PBS containing 0.2% Tween and 1% BSA. Individual human sera were then added at a dilution of 1 :20 (5% serum) or 1 :100 (1% serum), either unspiked (screening assay) or spiked (confirmatory assay) with the same molecule as coated in the corresponding well. The spiking concentration ranged from 60 to 115 nM and spiked samples were pre-incubated for 1 hour.
  • Antibodies bound to the molecules coated on the plate where then detected with 100 ng/ml rabbit anti-human IgG-HRP for 1 hour.
  • the initial screening assay was not performed. Instead, the test samples were directly analysed using the confirmatory assay procedure. For data analysis however, the same calculations were performed, which at least involves the calculation of the SCP and the %CCP.
  • SCP Screening Cut Point
  • NF Normalization Factor
  • FCP Floating Cut Point
  • test compound 40 individual human serum samples of healthy untreated subjects were analyzed. In other cases 20 individual human serum samples of healthy untreated subjects were analyzed.
  • the screening cut point is the threshold at which a signal is considered positive (screening positive). It is calculated such that 5% false positive sera are included. [0195]
  • Mean N corresponds to mean signal from all unspiked individual sera measured for a specific test compound
  • SDN corresponds to standard deviation calculated from all unspiked individual sera measured for a specific test compound.
  • NF normalization factor
  • negative control mean corresponds to mean signal from the negative controls (pooled individual human sera, same on each plate) analyzed in duplicates (/. e. 2 wells) per plate.
  • the Floating Cut Point (FCP) for each plate was used as the reference cut point.
  • the FCP takes into account the analytical variability of each analytical run, by normalizing the SCP with the negative controls of the plate.
  • the FCP is calculated for each analytical run as follows:
  • Mean NC refers to the mean signal of the negative control samples; “NF” refers to the normalization factor, as defined above.
  • PRO1922 variants and seven PRO2230 variants according to the present invention have been measured for their immunogenic properties using the pre-existing ADA binding assay described above.
  • Two references i. e. PRO1922-L12S-V103T-L144T (PRO2990) and PRO2230-L12S-V103T-L144T (PRO2984) have been analyzed as well. The measurements were directly performed in the confirmatory assay setup using 20 human serum samples.
  • Table 14 Number of positive serum samples above 30 % CCP of PRO1922 (MSLN binding scFv) variants and of PRO2230 (PDL1 binding scFv):
  • NM21-1480 variants including NM21-1480 (PRO1480) as reference have been measured for their immunogenic properties using the pre-existing ADA binding assay described above. The measurements were directly performed in the confirmatory assay setup using 40 human serum samples.
  • Table 15 Number of positive serum samples above 31,56 % CCP of PRO1922 (MSLN binding scFv) variants and of PRO2230 (PDL1 binding scFv):
  • NM28 variant PRO2741 and, as comparison, the corresponding unmodified reference PRO2660 have been measured for their immunogenic properties using the pre- existing ADA binding assay described above. The measurements were directly performed in the confirmatory assay setup using 20 human serum samples.
  • Table 17 Number of positive serum samples above 30 % CCP of PRO2668 and PRO2669 and the references PRO2510 and PRO2589: PRO2510 3 15

Landscapes

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

Abstract

La présente invention concerne des domaines variables d'anticorps, qui présentent une liaison réduite à des anticorps anti-médicament préexistants (ADA), et des anticorps comprenant un ou plusieurs desdits domaines variables d'anticorps. La présente invention concerne également des séquences d'acide nucléique codant pour lesdits domaines variables d'anticorps ou lesdits anticorps, un ou plusieurs vecteurs comprenant lesdites séquences d'acide nucléique, une ou plusieurs cellules hôtes comprenant lesdites séquences d'acide nucléique ou ledit/lesdits vecteur(s) ainsi qu'une méthode de production desdits domaines variables d'anticorps ou lesdits anticorps multispécifiques. La présente invention concerne également des compositions pharmaceutiques comprenant lesdits anticorps et des méthodes d'utilisation associées.
PCT/EP2021/087618 2020-12-23 2021-12-23 Domaines variables d'anticorps et anticorps ayant une immunogénicité réduite WO2022136693A1 (fr)

Priority Applications (18)

Application Number Priority Date Filing Date Title
EP21844734.0A EP4267622A1 (fr) 2020-12-23 2021-12-23 Domaines variables d'anticorps et anticorps ayant une immunogénicité réduite
KR1020237024454A KR20230125239A (ko) 2020-12-23 2021-12-23 면역원성이 감소된 항체 가변 도메인 및 항체
US18/258,957 US20240084039A1 (en) 2020-12-23 2021-12-23 Antibody variable domains and antibodies having decreased immunogenicity
IL303171A IL303171A (en) 2020-12-23 2021-12-23 Variable antibody domains and antibodies that have decreased immunogenicity
AU2021405066A AU2021405066A1 (en) 2020-12-23 2021-12-23 Antibody variable domains and antibodies having decreased immunogenicity
JP2023537607A JP2024501810A (ja) 2020-12-23 2021-12-23 減少した免疫原性を有する抗体可変ドメイン及び抗体
CN202180085944.1A CN116783218A (zh) 2020-12-23 2021-12-23 具有降低的免疫原性的抗体可变结构域和抗体
CA3205010A CA3205010A1 (fr) 2020-12-23 2021-12-23 Domaines variables d'anticorps et anticorps ayant une immunogenicite reduite
PCT/EP2022/052425 WO2022167460A1 (fr) 2021-02-02 2022-02-02 Anticorps multispécifiques ayant une spécificité pour ror1 et cd3
CA3208781A CA3208781A1 (fr) 2021-02-02 2022-02-02 Anticorps multispecifiques ayant une specificite pour ror1 et cd3
KR1020237029806A KR20230166075A (ko) 2021-02-02 2022-02-02 Ror1 및 cd3에 대한 특이성을 가지는 다중 특이적 항체
JP2023545975A JP2024504471A (ja) 2021-02-02 2022-02-02 Ror1およびcd3に対する特異性を有する多重特異性抗体
AU2022215847A AU2022215847A1 (en) 2021-02-02 2022-02-02 Multispecific antibodies having specificity for ror1 and cd3
EP22708399.5A EP4288451A1 (fr) 2021-02-02 2022-02-02 Anticorps multispécifiques ayant une spécificité pour ror1 et cd3
MX2023009022A MX2023009022A (es) 2021-02-02 2022-02-02 Anticuerpos multiespecificos con especificidad para ror1 y cd3.
CN202280023785.7A CN117043185A (zh) 2021-02-02 2022-02-02 对ror1和cd3具有特异性的多特异性抗体
TW111104438A TW202248211A (zh) 2021-02-02 2022-02-07 針對ror1與cd3之多特異性抗體
IL304403A IL304403A (en) 2021-02-02 2023-07-11 Multispecific antibodies with specificity for ror1 and cd3

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
EP20216957.9 2020-12-23
EP20216957.9A EP4019547A1 (fr) 2020-12-23 2020-12-23 Anticorps multispécifiques ayant une spécificité pour il-4r et il-31
EP21154786 2021-02-02
EP21154786.4 2021-02-02
PCT/EP2021/064427 WO2021239987A1 (fr) 2020-05-29 2021-05-28 Anticorps multispécifique
EPPCT/EP2021/064427 2021-05-28

Publications (1)

Publication Number Publication Date
WO2022136693A1 true WO2022136693A1 (fr) 2022-06-30

Family

ID=79730292

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2021/087618 WO2022136693A1 (fr) 2020-12-23 2021-12-23 Domaines variables d'anticorps et anticorps ayant une immunogénicité réduite

Country Status (1)

Country Link
WO (1) WO2022136693A1 (fr)

Citations (22)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4458066A (en) 1980-02-29 1984-07-03 University Patents, Inc. Process for preparing polynucleotides
US4881175A (en) 1986-09-02 1989-11-14 Genex Corporation Computer based system and method for determining and displaying possible chemical structures for converting double- or multiple-chain polypeptides to single-chain polypeptides
US5013653A (en) 1987-03-20 1991-05-07 Creative Biomolecules, Inc. Product and process for introduction of a hinge region into a fusion protein to facilitate cleavage
US5091513A (en) 1987-05-21 1992-02-25 Creative Biomolecules, Inc. Biosynthetic antibody binding sites
US5132405A (en) 1987-05-21 1992-07-21 Creative Biomolecules, Inc. Biosynthetic antibody binding sites
US5258498A (en) 1987-05-21 1993-11-02 Creative Biomolecules, Inc. Polypeptide linkers for production of biosynthetic proteins
US5260203A (en) 1986-09-02 1993-11-09 Enzon, Inc. Single polypeptide chain binding molecules
US5476786A (en) 1987-05-21 1995-12-19 Creative Biomolecules, Inc. Biosynthetic antibody binding sites
US5766886A (en) 1991-12-13 1998-06-16 Xoma Corporation Modified antibody variable domains
WO1999057150A2 (fr) 1998-05-05 1999-11-11 Deutsches Krebsforschungszentrum Stiftung des öffentlichen Rechts Constructions d'anticorps multivalentes
US6075181A (en) 1990-01-12 2000-06-13 Abgenix, Inc. Human antibodies derived from immunized xenomice
US6150584A (en) 1990-01-12 2000-11-21 Abgenix, Inc. Human antibodies derived from immunized xenomice
WO2004042072A2 (fr) 2002-11-01 2004-05-21 The Regents Of The University Of Colorado, A Body Corporate Analyse quantitative d'isoformes de proteines utilisant la spectrometrie de masse a temps de vol par desorption/ionisation laser assistee par matrice
WO2004092219A2 (fr) 2003-04-10 2004-10-28 Protein Design Labs, Inc Modification d'affinites de liaison pour fcrn ou de demi-vies de serum d'anticorps par mutagenese
US7807162B2 (en) * 2005-05-20 2010-10-05 Ablynx N.V. Single domain VHH antibodies against von Willebrand factor
WO2011075861A1 (fr) 2009-12-23 2011-06-30 Esbatech, An Alcon Biomedical Research Unit Llc Procédé pour faire baisser l'immunogénicité
WO2015067636A1 (fr) * 2012-11-05 2015-05-14 Delenex Therapeutics Ag Éléments de liaison dirigés contre l'il-1 bêta
WO2016202457A1 (fr) 2015-06-15 2016-12-22 Numab Ag Format d'anticorps hétérodimères multispécifiques
WO2019057787A1 (fr) 2017-09-20 2019-03-28 Numab Innovation Ag Nouvelles combinaisons stables de charpentes de domaines variables d'anticorps
WO2019072868A1 (fr) 2017-10-10 2019-04-18 Numab Therapeutics AG Anticorps multispécifiques
CN110229230A (zh) * 2019-04-26 2019-09-13 上海科棋药业科技有限公司 一种靶向cd38的单域抗体及其应用
WO2021089609A1 (fr) 2019-11-04 2021-05-14 Numab Therapeutics AG Anticorps multispécifiques

Patent Citations (24)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4458066A (en) 1980-02-29 1984-07-03 University Patents, Inc. Process for preparing polynucleotides
US4881175A (en) 1986-09-02 1989-11-14 Genex Corporation Computer based system and method for determining and displaying possible chemical structures for converting double- or multiple-chain polypeptides to single-chain polypeptides
US5260203A (en) 1986-09-02 1993-11-09 Enzon, Inc. Single polypeptide chain binding molecules
US5455030A (en) 1986-09-02 1995-10-03 Enzon Labs, Inc. Immunotheraphy using single chain polypeptide binding molecules
US5013653A (en) 1987-03-20 1991-05-07 Creative Biomolecules, Inc. Product and process for introduction of a hinge region into a fusion protein to facilitate cleavage
US5091513A (en) 1987-05-21 1992-02-25 Creative Biomolecules, Inc. Biosynthetic antibody binding sites
US5132405A (en) 1987-05-21 1992-07-21 Creative Biomolecules, Inc. Biosynthetic antibody binding sites
US5258498A (en) 1987-05-21 1993-11-02 Creative Biomolecules, Inc. Polypeptide linkers for production of biosynthetic proteins
US5476786A (en) 1987-05-21 1995-12-19 Creative Biomolecules, Inc. Biosynthetic antibody binding sites
US5482858A (en) 1987-05-21 1996-01-09 Creative Biomolecules, Inc. Polypeptide linkers for production of biosynthetic proteins
US6075181A (en) 1990-01-12 2000-06-13 Abgenix, Inc. Human antibodies derived from immunized xenomice
US6150584A (en) 1990-01-12 2000-11-21 Abgenix, Inc. Human antibodies derived from immunized xenomice
US5766886A (en) 1991-12-13 1998-06-16 Xoma Corporation Modified antibody variable domains
WO1999057150A2 (fr) 1998-05-05 1999-11-11 Deutsches Krebsforschungszentrum Stiftung des öffentlichen Rechts Constructions d'anticorps multivalentes
WO2004042072A2 (fr) 2002-11-01 2004-05-21 The Regents Of The University Of Colorado, A Body Corporate Analyse quantitative d'isoformes de proteines utilisant la spectrometrie de masse a temps de vol par desorption/ionisation laser assistee par matrice
WO2004092219A2 (fr) 2003-04-10 2004-10-28 Protein Design Labs, Inc Modification d'affinites de liaison pour fcrn ou de demi-vies de serum d'anticorps par mutagenese
US7807162B2 (en) * 2005-05-20 2010-10-05 Ablynx N.V. Single domain VHH antibodies against von Willebrand factor
WO2011075861A1 (fr) 2009-12-23 2011-06-30 Esbatech, An Alcon Biomedical Research Unit Llc Procédé pour faire baisser l'immunogénicité
WO2015067636A1 (fr) * 2012-11-05 2015-05-14 Delenex Therapeutics Ag Éléments de liaison dirigés contre l'il-1 bêta
WO2016202457A1 (fr) 2015-06-15 2016-12-22 Numab Ag Format d'anticorps hétérodimères multispécifiques
WO2019057787A1 (fr) 2017-09-20 2019-03-28 Numab Innovation Ag Nouvelles combinaisons stables de charpentes de domaines variables d'anticorps
WO2019072868A1 (fr) 2017-10-10 2019-04-18 Numab Therapeutics AG Anticorps multispécifiques
CN110229230A (zh) * 2019-04-26 2019-09-13 上海科棋药业科技有限公司 一种靶向cd38的单域抗体及其应用
WO2021089609A1 (fr) 2019-11-04 2021-05-14 Numab Therapeutics AG Anticorps multispécifiques

Non-Patent Citations (73)

* Cited by examiner, † Cited by third party
Title
"PCR Protocols: A Guide to Methods and Applications", 1990, ACADEMIC PRESS
"Remington: The Science and Practice of Pharmacy", 2000, MACK PUBLISHING CO
"Sustained and Controlled Release Drug Delivery Systems", 1978, MARCEL DEKKER, INC.
"UniProt", Database accession no. P02768
AL-LAZIKANI ET AL., JMB, vol. 273, 1997, pages 927 - 948
BATZER ET AL., NUCLEIC ACID RES., vol. 19, 1991, pages 5081
BEAUCAGE ET AL., TETRAHEDRON LETT., vol. 22, 1981, pages 1859
BITTNER ET AL., METH. ENZYMOL., vol. 153, 1987, pages 516
BOEMER ET AL., J. IMMUNOL, vol. 147, no. l, 1991, pages 86 - 95
BRENNAN ET AL., SCIENCE, vol. 229, 1985, pages 81 - 83
CAPET ET AL., IMMUNOMETHODS, vol. 4, 1994, pages 25 - 34
CHAMESBATY, FEMS MICROBIOL. LETTERS, vol. 189, 2000, pages 1 - 8
DE HAAS ET AL., J. LAB. CLIN. MED., vol. 126, 1995, pages 330 - 41
ECKERT ET AL., PCR METHODS AND APPLICATIONS, vol. 1, 1991, pages 17
EGAN ET AL., MABS, vol. 9, no. 1, 2017, pages 68 - 84
EGAN T. ET AL., MABS, vol. 9, 2017, pages 68 - 84
ELLIOTO'HARE, CELL, vol. 88, 1997, pages 223
FANALI, MOLECULAR ASPECTS OF MEDICINE, vol. 33, 2012, pages 209 - 290
FISCHER, N.LEGER, O., PATHOBIOLOGY, vol. 74, 2007, pages 3 - 14
GHETIE ET AL., NATURE BIOTECHNOLOGY, vol. 15, no. 7, 1997, pages 637 - 40
GHETIEWARD, IMMUNOL. TODAY, vol. 18, no. 12, 1997, pages 592 - 8
GLENNIE ET AL., J. IMMUNOL., vol. 139, 1987, pages 2367 - 2375
GREEN, M. R.SAMBROOK, J.: "Molecular Cloning: A Laboratory Manual", 2012, COLD SPRING HARBOR LABORATORY PRESS
GUYER ET AL., J. IMMUNOL., vol. 117, 1976, pages 587
HARRINGTON ET AL., NAT GENET., vol. 15, 1997, pages 345
HINTON ET AL., J. BIOL. CHEM. TJI, no. 8, 2004, pages 6213 - 6
HONEGGERPLUCKTHUN, J. MOL. BIOL., vol. 309, 2001, pages 657 - 670
HOOGENBOOMWINTER, J. MOL. BIOL, vol. 227, 1992, pages 381
HORNIG, N.FARBER-SCHWARZ, A., METHODS MOL. BIOL., vol. 907, 2012, pages 713 - 727
KARPOVSKY ET AL., J. EXP. MED., vol. 160, 1984, pages 1686
KIGUCHI YUKI ET AL: "The VH framework region 1 as a target of efficient mutagenesis for generating a variety of affinity-matured scFv mutants", SCIENTIFIC REPORTS, vol. 11, no. 1, 1 December 2021 (2021-12-01), XP055915182, Retrieved from the Internet <URL:https://www.nature.com/articles/s41598-021-87501-7.pdf> DOI: 10.1038/s41598-021-87501-7 *
KIM ET AL., J. IMMUNOL., vol. 24, 1994, pages 249
KIRIK UFUK ET AL: "Antibody Heavy Chain Variable Domains of Different Germline Gene Origins Diversify through Different Paths", FRONTIERS IN IMMUNOLOGY, vol. 8, 13 November 2017 (2017-11-13), XP055915193, DOI: 10.3389/fimmu.2017.01433 *
KNAPPIK ET AL., J. MOL. BIOL., vol. 296, 2000, pages 57 - 86
KOHLERMILSTEIN, NATURE, vol. 256, 1975, pages 495 - 497
KRUIF ET AL., BIOTECHNOL. BIOENG., vol. 106, 2010, pages 741 - 750
LABRIJN ET AL., PROC. NATL. ACAD. SCI. USA, vol. 110, 2013, pages 5145 - 5150
LEFRANC, M.-P. ET AL., DEV. COMP. IMMUNOL., vol. 27, 2003, pages 55 - 77
LEFRANC, M.-P., THE IMMUNOLOGIST, vol. 7, 1999, pages 132 - 136
LI ET AL., PROC. NATL. ACAD. SCI. USA, vol. 103, 2006, pages 3557 - 3562
LIU, M A ET AL., PROC. NATL. ACAD. SCI. USA, vol. 82, 1985, pages 8648
M. DAERON, ANNU. REV. IMMUNOL., vol. 5, 1997, pages 203 - 234
MABS, vol. 9, no. 2, February 2017 (2017-02-01), pages 182 - 212
MARKS ET AL., J. MOL. BIOL, vol. 222, 1991, pages 581
MATTILA ET AL., NUCLEIC ACIDS RES., vol. 19, 1991, pages 967
MAZOR, R. ET AL., IMMUNOL REV., vol. 270, no. 1, 2016, pages 152 - 64
MORRISON ET AL., PROC. NATL. ACAD. SCI. USA, vol. 81, 1984, pages 6851 - 6855
MORRISONOI, ADV. IMMUNOL., vol. 44, 1988, pages 65 - 92
NARANG ET AL., METH. ENZYMOL., vol. 68, 1979, pages 109
NATAGA, S.PASTAN, I., ADV DRUG DELIV REV, 2009, pages 977 - 985
NIESEN ET AL., NATURE PROTOCOLS, vol. 2, no. 9, 2007, pages 2212 - 2221
OHTSUKA ET AL., J. BIOL. CHEM., vol. 260, 1985, pages 2605 - 2608
ONDA, M. ET AL., PNAS, vol. 105, no. 32, 2008, pages 1311 - 11316
PADLAN, MOLEC. IMMUN., vol. 28, 1991, pages 489 - 498
PADLAN, MOLEC. IMMUN., vol. 31, 1994, pages 169 - 217
PAULUS, BEHRING INS. MITT., vol. 78, 1985, pages 118 - 132
QUEEN ET AL., IMMUNOL. REV., vol. 89, 1986, pages 49 - 68
RAVETCHKINET, ANNU. REV. IMMUNOL., vol. 9, 1991, pages 457 - 92
RIDGWAY ET AL., PROTEIN ENG., vol. 9, 1996, pages 617 - 21
ROSENFELD ET AL., CELL, vol. 68, 1992, pages 143
ROSSOLINI ET AL., MOL. CELL. PROBES, vol. 8, 1994, pages 91 - 98
SCHARF ET AL., RESULTS PROBL. CELL DIFFER., vol. 20, 1994, pages 125
SHIELDS ET AL., J. BIOL. CHEM., vol. 9, no. 2, 2001, pages 6591 - 6604
SMITH, ANNU. REV. MICROBIOL., vol. 49, 1995, pages 807
SPIESS ET AL., J BIOL CHEM., vol. 288, no. 37, 2013, pages 26583 - 93
SUGIO, PROTEIN ENG, vol. 12, 1999, pages 439 - 446
VAN DIJKVAN DE WINKEL, CURR. OPIN. PHARMACOL, vol. 5, 2001, pages 368 - 74
VERHOEYEN ET AL., SCIENCE, vol. 239, 1988, pages 1534 - 1536
WINNACKER: "FROM GENES TO CLONES", 1987, VCH PUBLISHERS
WONGRALEIGH, PROTEIN SCIENCE, vol. 25, 2016, pages 1834 - 1840
YOKOYAMA ET AL., CURR. PROTOC. IMMUNOL., 2006
ZHAO, L.LI, J., BMC STRUCT. BIOL., vol. 10, 2010, pages S6
ZHOU JEFFREY O. ET AL: "The Effects of Framework Mutations at the Variable Domain Interface on Antibody Affinity Maturation in an HIV-1 Broadly Neutralizing Antibody Lineage", FRONTIERS IN IMMUNOLOGY, vol. 11, 1 January 2020 (2020-01-01), pages 1529, XP055915181, DOI: 10.3389/fimmu.2020.01529 *

Similar Documents

Publication Publication Date Title
CN107849136B (zh) 抗TfR抗体及其在治疗增殖性和炎性疾病中的用途
US11261254B1 (en) Antibodies
EP3915580A1 (fr) Anticorps multi-spécifique
AU2021405058A1 (en) Multispecific antibodies having specificity for il-4r and il-31
EP4288451A1 (fr) Anticorps multispécifiques ayant une spécificité pour ror1 et cd3
US20230303694A1 (en) Antibodies that bind gamma-delta t cell receptors
US20230203162A1 (en) Multispecific antibody
US20240084039A1 (en) Antibody variable domains and antibodies having decreased immunogenicity
WO2022136693A1 (fr) Domaines variables d&#39;anticorps et anticorps ayant une immunogénicité réduite
EP4273162A1 (fr) Domaines variables d&#39;anticorps et anticorps présentant une immunogénicité réduite
EP4136122A1 (fr) Constructions d&#39;anticorps se liant à 4-1bb et récepteurs alpha de folate et leurs utilisations
WO2023214047A1 (fr) Domaines variables d&#39;anticorps et anticorps ayant une immunogénicité réduite
CN116783218A (zh) 具有降低的免疫原性的抗体可变结构域和抗体
EP4292609A1 (fr) Compositions comprenant des anticorps se liant aux récepteurs de lymphocytes t gamma-delta
AU2021231712A1 (en) Anti-CD19 antibodies and methods of using and making thereof

Legal Events

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

Ref document number: 21844734

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 3205010

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 202180085944.1

Country of ref document: CN

WWE Wipo information: entry into national phase

Ref document number: 2023537607

Country of ref document: JP

ENP Entry into the national phase

Ref document number: 2021405066

Country of ref document: AU

Date of ref document: 20211223

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 18258957

Country of ref document: US

ENP Entry into the national phase

Ref document number: 20237024454

Country of ref document: KR

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 2021844734

Country of ref document: EP

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 2021844734

Country of ref document: EP

Effective date: 20230724