OA19651A - Anti-Pd-1 antibodies, method for producing same and method for using same. - Google Patents

Anti-Pd-1 antibodies, method for producing same and method for using same. Download PDF

Info

Publication number
OA19651A
OA19651A OA1201800540 OA19651A OA 19651 A OA19651 A OA 19651A OA 1201800540 OA1201800540 OA 1201800540 OA 19651 A OA19651 A OA 19651A
Authority
OA
OAPI
Prior art keywords
antibody
binding
fragment
human
antibodies
Prior art date
Application number
OA1201800540
Inventor
Viktoriia Mikhailovna EKIMOVA
Dmitry Valeryevich KORZHAVIN
Yulia Sergeevna CHERNYKH
Timofey Aleksandrovich Nemankin
Valery Vladimirovich SOLOVYEV
Anna Konstantinovna VLADIMIROVA
Irina Andreevna BULANKINA
Sergei Vasilyevich DIDUK
Anna Vladimirovna EROSHOVA
Iakov Iurevich USTIUGOV
Marina Vladimirovna ARTIUKHOVA
Andrei Borisovich ULITIN
Roman Alekseevich IVANOV
Dmitry Valentinovich MOROZOV
Olga Vladimirovna GONCHAROVA
Original Assignee
Joint Stock Company "Biocad"
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Joint Stock Company "Biocad" filed Critical Joint Stock Company "Biocad"
Publication of OA19651A publication Critical patent/OA19651A/en

Links

Abstract

The present invention relates to biotechnology and comprises isolated monoclonal antibodies, in particular human monoclonal antibodies, which specifically bind to PD-1 with high affinity. The antibodies according to the invention may be chimeric, humanized or human antibodies, or antigen-binding fragments thereof, and may be used as a medicinal agent in oncology and immuno-oncology, for treating diseases associated with various cell proliferation or development disorders. The invention also relates to methods for producing said antibodies and a method for treating human diseases using said antibodies.

Description

ANTI-PD-l ANTIBODIES, A METHOD OF PRODUCTION AND A METHOD OF USE THEREOF
Field of invention
The présent invention relates to biotechnology and provides isolated monoclonal antibodies, in particular human monoclonal antibodies that specifically bind to high affmity PD-1. The antibodies of the invention can be chimeric, humanized or human antibodies, or antigen-binding fragments thereof, and can be used as a médicinal agent in oncology and immuno-oncology, and for treating diseases associated with various cell prolifération or development disorders. The invention also relates to methods of producing said antibodies and a method of treating human diseases with said antibodies.
Background of the invention
Programmed death 1 (PD-1) protein is an inhibitory member of the CD28 receptor family that also includes CD28, CTLA-4, ICOS and BTLA. PD-1 is expressed on activated B cells, T cells, and myeloid cells (Agata et al., supra; Okazaki et al. (2002) Curr. Opin. Immunol. 14: 391779-82; Bennet et al. (2003) J Immunol 170:711-8). The initial members of this family, CD28 and ICOS, were detected by functional effects on increase in T cell prolifération following the addition of monoclonal antibodies (Hutloff et al. (1999) Nature 397:263266; Hansen et al. (1980) Immunogenics 10:247-260). PD-1 was detected by screening for differential expression in apoptotic cells (Ishida et al. (1992) EMBO J 11:3887-95). Other members of this family, CTLA-4 and BTLA, were detected by screening for differential expression in cytotoxic T-lymphocytes and TH1 cells, respectively. CD28, ICOS and CTLA-4, ail hâve an unpaired cysteine residue that allows them to homodimerize. In contrast, PD-1 is believed to exist as a monomer, lacking the unpaired cysteine residue characteristic in other CD28 family members.
PD-1 is a 55 kDa type I transmembrane protein that is part of the Ig gene superfamily (Agata et al. (1996) Int Immunol 8:765-72). PD-1 comprises a membrane proximal immunoreceptor tyrosine inhibitory motif (ITIM) and a membrane distal tyrosine-based switch motif (ITSM) (Thomas, M.L. (1995) J Exp Med 181:1953-6; Vivier, E n Daeron, M (1997) Immunol Today 18:28691). Although structurally similar to CTLA-4, PD-1 lacks the MYPPY motif that is critical for B7-1 and B7-2 binding. It has been detected that PD-1 has two ligands, PD-L1 and PD-L2, which hâve been shown to negatively regulate T cell activation after binding to PD-1 (Freeman et al. (2000) J Exp Med 192:1027-34; Latchman et al. (2001) Nat Immunol 2:261-8; Carter et al. (2002) Eur J Immunol 32:634-43). Both PD-L1 and PD-L2 are B7 homologs that bind to PD1, but do not bind to other members of the CD28 family.
One PD-1 ligand, PD-L1, is abundant in various human cancers (Dong et al. (2002) Nat. Med. 8:787-9). The interaction between PD-1 and PD-L1 leads to a réduction in the number of tumor-infiltrating lymphocytes, decrease in T cell receptor-mediated prolifération, and escape from immunological surveillance of cancer cells (Dong et al. (2003) J. Mol. Med. 81:281-7; Blank et al. (2005) Cancer Immunol. Immunother. 54:307-314; Konishi et al. (2004) Clin. Cancer Res. 10:5094-100). Immunosuppression may be reversed by inhibiting the local interaction of PD-L1 with PD-1, and this effect is additive when the interaction of PD-L2 with PD-1 is blocked (Iwai et al. (2002) Proc. Nat’l. Acad. Sci. USA 99:12293-7; Brown et al. (2003) J. Immunol. 170:1257-66).
PD-1 is an inhibitory member of the CD28 family and is expressed on activated B cells, T cells and myeloid cells (Agata et al., supra; Okazaki et al. (2002) Curr Opin Immunol 14: 391779-82; Bennett et al. (2003) J Immunol 170:711-8). PD-1-déficient animais are prone to developing various autoimmune diseases including autoimmune cardiopathy, and lupus-like syndrome comprised of arthritis and nephritis (Nishimura et al. (1999) Immunity 11:141-51; Nishimura et al. (2001) Science 291:319-22). In addition, PD-1 was found to play a rôle in autoimmune encephalomyelitis, systemic lupus erythematosus, graft-versus-host disease (GVHD), type I diabètes and rheumatoid arthritis (Salama et al. (2003) J Exp Med 198:71-78; Prokunina and
Alarcon-Riquelme (2004) Hum Mol Genet 13:R143; Nielsen et al. (2004) Lupus 13:510). In a murine B cell tumor line, the ITSM of PD-1 was shown to be essential to block BCR-mediated Ca2+-flux and tyrosine phosphorylation of downstream effector molécules (Okazaki et al. (2001) PNAS 98:13866-71).
Today, there are a number of anti-PD-1 antibodies, such as nivolumab (BMS-936558, MDX-1106 or ONO-4538; BMS), pembrolizumab (Merck).
The prior art discloses monoclonal anti-PD-1 antibodies comprising certain amino acid sequences according to WO 2006/121168 (nivolumab, BMS), which exhibit some useful properties, such as high affinity binding to human PD-1, but no significant cross-reactivity with human CD28, CTLA-4 or ICOS. In addition, it has been shown that these antibodies modulate immune responses. Thus, the présent application also describes a method for modulating immune responses using anti-PD-1 antibodies. In particular, the présent invention provides a method for inhibiting in vivo growth of tumor cells using anti-PD-1 antibodies.
The prior art also discloses an isolated PD-1 binding protein described in WO2009/114335 (pembrolizumab, Merck) which comprises a first variable région and a second variable région. The first variable région is a heavy chain comprising various CDRs, and the second variable région is a light chain also comprising various CDRs.
Thus, it is important to develop antibodies that recognize PD-1, and methods of use of such agents. The présent invention provides antibodies that specifically bind to PD-1 and hâve advantageous characteristics of functional activity, affinity, specificity, and stability in the test assays.
Brief summary of invention
The présent invention relates to human monoclonal antibodies that specifically bind to PD-1. Such antibodies can be used in the treatment of oncological and infectious diseases. The monoclonal antibodies of the présent invention are believed to provide the best clinical response, as compared with current methods of treatment of said diseases, including treatment with antibodies.
In one aspect, the présent invention relates to an antibody or antigenbinding fragment thereof that is capable of binding to a human PD-1 receptor and comprises an amino acid sequence that is at least 75% homologous to the sequence of SEQ ID NO: 3.
In one embodiment, the présent invention relates to an antibody or fragment thereof which comprises the amino acid sequence of SEQ ID NO:3.
In some embodiments, the présent invention relates to an antibody or fragment thereof which comprises the following:
- a sequence of a heavy chain variable domain that is at least 75% homologous to the sequence of SEQ ID NO:7, and
- a sequence of a light chain variable domain that is at least 75% homologous to the sequence of SEQ ID NO:8.
In one embodiment, the présent invention relates to an antibody or fragment thereof which comprises the amino acid sequences of SEQ ID NO: 13.
In some embodiments, a binding fragment competes for binding or binds to the same epitope as a binding domain comprising the amino acid sequence of SEQ ID NO: 7.
In some embodiments, a binding fragment is at least 90% homologous to the amino acid sequence of SEQ ID NO: 7. In one embodiment of the invention, a binding domain comprises the amino acid sequence of SEQ ID NO: 7. In one embodiment of the invention, a binding domain can be humanized.
In some embodiments of the invention, an antibody or antigen-binding fragment thereof is characterized in that it relates to human IgGl, IgG2, IgG3, IgG4 isotypes.
In some embodiments, an antibody or fragment thereof has a heavy chain sequence that is at least 90% homologous to the sequence of SEQ ID NO 9.
In some embodiments, an antibody or fragment thereof has a light chain sequence that is at least 90% homologous to the sequence of SEQ ID NO: 10.
In some embodiments, the Fc constant région of an antibody or fragment thereof comprises any mutations that reduce or eliminate any of the effector functions (ADCC, ADCP or CDC) as compared with the natural sequence.
In some embodiments, the Fc constant région of an antibody or fragment thereof comprises mutations that increase animal or human pharmacokinetic parameters, such as ί1/2β (h) or Cmax (pg/ml).
In some embodiments, an antibody or antigen-binding fragment thereof hâve at least one of the following properties:
a) aggregation stability: the aggregate content does not increase by more than 5% of the initial content in solution at concentrations above 10 mg/ml and at storage température T = 4 C for more than 6 months;
b) aggregation stability: the aggregate content does not increase by more than 5% of the initial content in solution at concentrations above 10 mg/ml and with an increase in température to 37°C for more than 2 weeks;
c) aggregation stability: the aggregate content does not increase by more than 5% of the initial content in solution at concentrations above 10 mg/ml and with an increase in température to 50°C for more than 6 hours;
d) the dissociation constant KD of not more than 10'9 (M) when binding to human PD-1 ;
e) the kinetic association constant kon (1/Ms) of at least 105 (1/Ms) when binding to human PD-1 ;
f) the kinetic dissociation constant dis (1/s) of not more than 10'4 (1/s) when binding to human PD-1.
In one aspect, the présent invention relates to a bispecific antibody that comprises any antigen-binding fragment of an antibody as described above.
In one aspect, the présent invention relates to an isolated nucleic acid molécule encoding an antibody or antigen-binding domain thereof according to any of daims 1-14.
In one aspect, the présent invention relates to an expression vector that comprises any of isolated nucleic acid molécules described herein.
In one aspect, the présent invention relates to a host cell that comprises any nucléotide sequence described herein.
In one aspect, the présent invention relates to a method for producing a host cell that comprises transfection of a suitable stem cell with an expression vector.
In one aspect, the présent invention relates to a method for the préparation of any antibody as described herein, comprising the production of a host cell, culturing of a host cell under conditions sufficient to produce said antibody or fragment thereof, followed by isolation and purification of the obtained antibody or active fragment thereof.
In some embodiments, the présent invention relates to a pharmaceutical composition comprising an antibody or fragment thereof as described above, in combination with one or more pharmaceutically acceptable excipients, diluents or carriers. In some embodiments, a pharmaceutical composition is intended to be used for the treatment of oncological and infectious diseases.
In one aspect, the présent invention relates to a method for inhibiting the biological activity of PD-l in a subject in need of such inhibition, which comprises administering an effective amount of any antibody as described above.
In one aspect, the présent invention relates to a method for treatment of a patient in need of such treatment, which comprises administering any antibody or antigen-binding fragment or pharmaceutical composition described herein.
Description of the drawings
Fig. 1 Scheme of synthesis of a human naïve combinatorial library.
Fig. 2. Phagemid for cloning of Fab phage display libraries (A) and expression plasmid for production of Fab (B).
Fig. 3. BCD-100 electrophoregram under reducing conditions (3A, 12% SDS-PAGE), under non-reducing conditions (3B, 8% SDS-PAGE).
Fig. 4. Immunoenzymatic assay of interaction of BCD-100 with PDI and other antigens.
Fig. 5. Réactivation of NFAT-signaling by anti-PDI antibodies in JurkatNFAT-PD1 reporter cell line.
Fig. 6. Stimulation of production of IL-2 by anti-PDI antibodies in human whole blood in the presence of staphylococcal enterotoxin.
Fig. 7. Analysis of antibody-dependent cellmediated cytotoxicity (ADCC) of anti-PDI antibody on Jurkat-PDl cell line.
Fig. 8. Analysis of interactions of BCD-100 candidates with FcRn and Fcy receptors on Octet RED 96.
Fig. 9. Immunoenzymatic assay of interactions of BCD-100 with PDI receptors of different organisms.
Fig. 10. Immunoenzymatic assay of interactions of BCD-100 with CD28 family receptors.
Fig. 11. Analysis of interactions of BCD-100 candidates with human and cynomolgus monkey PDI receptors on Octet RED 96.
Fig. 12. Results of thermal stress (50°C, 12 h) of BCD-100 molécule.
Disclosure of the invention
Définitions and general methods
Unless defined otherwise, ail technical and scientific tenus used herein hâve the same meaning as is commonly understood by one of ordinary skill in the art. Although methods and materials similar or équivalent to those described herein can be used in the practice or testing of embodiments of the invention, exemplary methods and/or materials are described below. Ail publications and other references mentioned herein are incorporated by reference in their entirety. In case of contradiction, this description, including définitions, shall prevail. Although a number of prior art publications are referred to herein, such references do not constitute an admission that any of these documents form part of the common general knowledge in the art.
Further, unless otherwise required by context, singular terms shall include pluralities and plural terms shall include the singular. Typically, the classification and methods of cell and tissue culture, molecular biology, immunology, microbiology, genetics, analytical chemistry, organic synthesis chemistry, medical and pharmaceutical chemistry, as well as hybridization and chemistry of protein and nucleic acids described herein are well known and widely used by those skilled in the art. Enzyme reactions and purification methods are performed according to the manufacturées instructions, as is known in the art, or as described herein.
Throughout this disclosure and embodiments, the word consist and comprise or variations thereof, such as consists or consisting, comprises or comprising shall be understood to imply the inclusion of a stated integer or group of integers but not the exclusion of any other integer or group of integers.
Définitions related to antibodies
As used herein, the terms programmed death 1, programmable cell death 1, PD-1 protein, PD-1, CD279, PDCD1, hPD-1 and hPD-I are interchangeable and refer to any variants, isoforms, species homologs of human PD-1 and analogs thereof comprising at least one common epitope with PD-1.
The terms immune response, autoimmune response and autoimmune inflammation refer, for example, to the action of lymphocytes, antigenpresenting cells, phagocytic cells, granulocytes and soluble macromolecules produced by said cells or liver cells (including antibodies, cytokines and complément produced in the resuit of sélective damage, destruction or élimination of invasive pathogens, cells or tissues infected with pathogens, cancer cells or, in cases of autoimmunity or pathological inflammation, normal cells or tissues from the human body).
The terni binding molécule as used herein includes antibodies, immunoglobulins and antigen-binding fragments of an antibody. The term antibody (Ab) or immunoglobulin (Ig) as used herein is intended to refer to a tetramer comprising two heavy (H) chains (about 50-70 kDa) and two light (L) chains (about 25 kDa), which are linked by disulfide bridges. Each heavy chain consists of a heavy chain variable domain (VH) and a heavy chain constant région (CH). Each light chain consists of a light chain variable domain (VL) and a light chain constant région (CL). VH and VL domains can be further subdivided into régions of hypervariability, termed complementarity determining régions (CDR), interspersed with régions that are more conserved, termed framework régions (FR). Each VH and VL is composed of three CDRs (H-CDR as used herein dénotés a heavy chain CDR and L-CDR as used herein dénotés a light chain CDR) and four FRs, arranged from aminoterminus to carboxy-terminus in the following order: FRI, CDR1, FR2, CDR2, FR3, CDR3, FR4. Attribution of amino acids to each région can be made in accordance with the définitions by IMGT® (Lefranc et al., Dev Comp Immunol 27(1):55-77 (2003); or the définitions by Kabat, Sequences of Proteins of Immunological Interest (National Institutes of Health, Bethesda, MD (1987 and 1991)); Chothia & Lesk, J. Mol. Biol. 196:901-917 (1987); or Chothia et al., Nature 342:878-883 (1989).
As used herein, the ternis antibody and immunoglobulin are interchangeable.
The term antigen-binding portion of an antibody (or antigen portion, fragment) as used herein is intended to refer to one or more portions or fragments of an antibody, that retain the ability to specifically bind to an antigen (e.g., PDI). It has been shown that the antigen-binding function of an antibody can be performed by some fragments of a full-length antibody. Examples of binding fragments encompassed within the term antigen-binding portion include (i) a Fab fragment, a monovalent fragment consisting of VL, VH, CL and CH1 domains; (ii) a F(ab)2 fragment, a bivalent fragment comprising two Fab fragments linked by a disulfide bridge at the hinge région; (iii) a Fd fragment consisting of VH and CH1 domains; (iv) a Fv fragment consisting of VL and VH domains of a single arm of an antibody; (v) a single-domain antibody *dAb) fragment, which consists of a VH domain; and (vi) an isolated complementarity determining région (CDR) able to specifically bind to an antigen. Furthermore, although the two domains of the Fv fragment, VL and VH, are encoded by individual genes, they can be joined, using recombinant methods, by a synthetic linker that enables them to be made as a single chain protein in which the VL and VH régions pair to form monovalent molécules (known as single chain Fv (scFv)). The présent invention also provides antigen-binding molécules comprising V H and/or VL. In the case of VH, a molécule may also comprise one or more of CH1, hinge, CH2 or CH3 régions. Such single chain antibodies are also intended to be encompassed within the term antigen-binding portion of an antibody. Other forms of single-chain antibodies, such as diabodies, are also encompassed. Diabodies are small bivalent and bispecific antibodies, in which domain VH and domain VL are expressed on the same polypeptide chain (VHVL). By using a linker that is too short to allow pairing between the two domains on the same chain, the domains are forced to pair with the complementary domains of another chain and create two antigen-binding sites. Antibody régions, such as Fab- and F(ab') 2 fragments can be prepared from whole antibodies using conventional techniques, e.g., papain or pepsin hydrolyses of whole antibodies. Moreover, antibodies, portions thereof and immunoadhesion molécules can be prepared using standard recombinant DNA techniques, for example, as described herein.
The term recombinant antibody is intended to refer to an antibody that is expressed from a cell or cell line comprising nucléotide sequence(s) encoding antibodies, wherein said nucléotide sequence(s) is not naturally associated with the cell.
As used herein, the term variant antibody is intended to refer to an antibody, which has an amino acid sequence that differs from the amino acid sequence of a parental antibody thereof by virtue of adding, deleting and/or substituting one or more amino acid residues as compared to the sequence of a parental antibody. In a preferred embodiment, a variant antibody comprises at least one or more (e.g., one to twelve, e.g., two, three, four, five, six, seven, eight or nine, ten, eleven or twelve; in some embodiments, a variant antibody comprises from one to about ten) additions, délétions, and/or substitutions of amino acids as compared to a parental antibody. In some embodiments, such additions, délétions and/or substitutions are made in the CDRs of a variant antibody. Identity or homology with respect to the sequence of a variant antibody is defined herein as the percentage of amino acid residues in the variant antibody sequence that are identical to the parental antibody residues, after aligning the sequences and introducing gaps, if necessary, to achieve the maximum percent of sequence identity. A variant antibody retains the ability to bind to the same antigen, and preferably to an epitope, to which the parental antibody binds; and in some embodiments, at least one property or biological activity is superior to that of a parental antibody. For example, a variant antibody may hâve, e.g., a stronger binding affmity, longer half-life, lower IC50, or enhanced ability to inhibit antigen biological activity as compared to a parental antibody. The variant antibody of particular interest herein is one which displays at least 2-fold, (preferably at least 5-fold, 10-fold or 20-fold) enhancement in biological activity as compared to a parental antibody.
In a broad sense, the terni chimeric antibody is intended to refer to an antibody that comprises one or more régions of one antibody, and one or more régions of one or several other antibodies, typically, a partially human and partially non-human antibody, i.e. derived partially from a non-human animal, such as mice, rats, or the like vermin, or the Camelidae such as llama and alpaca. Chimeric antibodies are generally preferred over nonhuman antibodies in order to reduce the risk of a human anti-antibody immune response, e.g. a human anti-mouse antibody immune response in the case of a il murine antibody. An example of a typical chimeric antibody is one in which the variable région sequences are murine sequences, while the constant région sequences are human. In the case of a chimeric antibody, the non-human parts may be subjected to further alteration in order to humanize the antibody.
The term humanization is intended to refer to the fact that when an antibody has a fully or partially non-human origin, for example, a mouse or llama antibody obtained by immunizing mice or Hamas, respectively, with an antigen of interest, or is a chimeric antibody based on such an antibody of a mouse or llama, it is possible to substitute certain amino acids, in particular in the framework régions and constant domains of heavy and light chains, in order to avoid or minimize the immune response in humans. Antibodies interact with target antigens predominantly through amino acid residues that are located in the six heavy and light chain CDRs. For this reason, amino acid sequences within CDRs are far more variable between individual antibodies than those outside of CDRs. Because CDR sequences are responsible for most antibody-antigen interactions, it is possible to express recombinant antibodies that mimic the properties of a spécifie naturally occurring antibody, or more generally, of any spécifie antibody with said amino acid sequence, e.g., by constructing expression vectors that express CDR sequences from the spécifie antibody grafted onto framework sequences from a different antibody. As a resuit, it is possible to humanize a non-human antibody and, to a large extent, préservé binding specificity and affinity of the initial antibody. Although it is not possible to precisely predict the immunogenicity and thereby the human anti-antibody response of a particular antibody, non-human antibodies are typically more immunogenic than human antibodies. Chimeric antibodies, where the foreign (e.g. vermin or Camelidae) constant régions hâve been substituted with sequences of human origin, hâve shown to be generally less immunogenic than those of fùlly foreign origin, and the trend in therapeutic antibodies is towards humanized or fully human antibodies. Therefore, chimeric antibodies or other antibodies of non-human origin can be humanized to reduce the risk of a human anti-antibody response.
For chimeric antibodies, humanization typically involves modification of the framework régions of variable région sequences. Amino acid residues that are part of complementarity determining régions (CDRs) will be most often not modified by virtue of humanization, although in some cases it may be désirable in order to modify individual amino acid residues of a CDR, for example, in order to delete a glycosylation site, deamidation site, aspartate isomerization site, or undesired cysteine or méthionine residues. N-linked glycosylation is made by virtue of attaching an oligosaccharide chain to an asparagine residue in a tripeptide sequence Asn-X-Ser or Asn-X-Thr, where X can be any amino acid except Pro. Removal of an N-glycosylation site may be achieved by mutating either the Asn or Ser/Thr residue by a different residue, preferably by way of conservative substitution. Deamidation of asparagine and glutamine residues can occur depending on such factors as pH and surface exposure. Asparagine residues are especially susceptible to deamidation, primarily when présent in sequence Asn Gly, and in a lesser degree in other dipeptide sequences such as Asn-Ala. Provided a CDR sequence comprises such a deamidation site, in particular Asn-Gly, it may be désirable to remove this site, typically by virtue of conservative substitution to delete one of the implicated residues.
Numerous methods for humanizing an antibody sequence are known in the art; see, for example, a review by Almagro & Fransson, Front Biosci. 13:1619-1633 (2008). One commonly used method is CDR grafting, when, e.g., murine chimeric antibodies involve identification of human germ-line gene counterparts to the murine variable région genes and grafting of the murine CDR sequences into this framework. CDR grafting may be based on the CDR définitions by Kabat, although the last édition (Magdelaine-Beuzelin et al., Crit Rev.Oncol Hematol. 64:210 225 (2007)) suggests that the IMGT® (the international ImMunoGeneTics information System®, www.imgt.org) définition may improve humanization results (see Lefranc et al., Dev. Comp Immunol.
27:55-77 (2003)). In some cases, CDR grafting may reduce the binding specificity and affinity, and thus the biological activity, of a CDR grafted nonhuman antibody, as compared to a parental antibody from which the CDRs were obtained. Back mutations (which are sometimes referred to as framework région repair may be introduced at selected positions of a CDR grafted antibody, typically in framework régions, in order to restore the binding specificity and affinity of a parental antibody). Identification of positions for possible back mutations can be performed using information available in the literature and in antibody databases. Amino acid residues that are candidates for back mutations are typically those that are located at the surface of an antibody molécule, whereas residues that are buried or that hâve a low degree of surface exposure will not normally be altered. An alternative humanization technique to CDR grafting and back mutation is resurfacing, in which non-surface exposed residues of non-human origin are retained, whereas surface residues are altered to human residues.
In certain cases, it may also be désirable to alter one or more CDR amino acid residues in order to improve binding affinity to the target epitope. This is known as affinity maturation and may optionally be performed in connection with humanization, for example in situations where humanization of an antibody leads to reduced binding specificity or affinity and it is not possible to sufficiently improve the binding specificity or affinity by back mutations alone. Various affinity maturation methods are known in the art, for example the in vitro scanning saturation mutagenesis method described by Burks et al., Proc Natl Acad S ci USA, 94:412-417 (1997) and the stepwise in vitro affinity maturation method by Wu et al., Proc Natl Acad Sci USA 95:6037 6042 (1998).
The term isolated protein, isolated polypeptide or isolated antibody is intended to refer to a protein, polypeptide or antibody, that by virtue of origin or source of dérivation thereof (1) is not associated with naturally associated components that accompany them in a native State thereof, (2) is free of other proteins from the same species, (3) is expressed by a cell from a different species, or (4) does not occur in nature. Thus, a polypeptide that is chemically synthesized or synthesized in a cellular System different from the cell from which it naturally originates will be isolated from its naturally associated components. A protein may also be substantially free of naturally associated components by virtue of isolation, using protein purification techniques well known in the art.
As used herein, the term germinal is intended to refer to the nucléotide and amino acid sequences of antibody genes and gene segments and how they are transmitted from parents to progeny via germinal cells. Germ-line sequences differ from the nucléotide sequences encoding antibodies in mature B cells that hâve been altered as a resuit of recombination and supermutation during the maturation of B cells. An antibody that utilizes a particular germ-line sequence has nucléotide and amino acid sequences that are aligned to a germline nucléotide sequence or amino acid sequence, to which it corresponds more fully than to any other germ-line nucléotide or amino acid sequences.
The term affinity is intended to refer to measuring the attraction between an antigen and binding molécule, e.g., an antibody. The intrinsic ability to attract a binding molécule for an antigen is typically expressed as the binding affinity equilibrium constant (KD) of a particular binding molecule-antigen interaction. A binding molécule is said to specifically bind to an antigen when KD is < l mM, preferably <100 nM. A KD binding affmity constant can be measured, e.g., by surface plasmon résonance (BIAcoreTM) or bio-layer interferometry, for example using ProteOnTM XPR36 SPR (Bio-Rad) or OctetTM Systems.
The term Ka as used herein is intended to refer to the association rate of a particular antibody-antigen interaction, whereas the term Kd is intended to refer to the dissociation rate of a particular antibody-antigen interaction. The term Kd as used herein is intended to refer to the dissociation constant, which is obtained from the ratio of Kd to Ka (i.e. Kd/Ka) and is expressed as a molar concentration (M). Kd values for antibodies can be determined using methods well established in the art.
A preferred method for determining the Kd of an antibody is surface plasmon résonance using a biosensor system such as a BIAcoreTM system.
As used herein, the term high affinity for an IgG antibody is intended to refer to an antibody having Kd 10' M, more preferably 10' M or less and even more preferably 10'10 M or less for a target antigen. However, high affinity binding can vary for other antigen isotypes. For example, high affinity binding for an IgM isotype is intended to refer to an antibody having KD 10' M or less, more preferably 10' M or less, even more preferably 10' M or less.
The term koff as used herein is intended to refer to the dissociation rate constant of a particular binding molecule-antigen interaction. The dissociation rate constant (koff +) can be measured using bio-layer interferometry, for example, using Octet™ system.
The term epitope as used herein is intended to refer to a portion (déterminant) of an antigen that specifically binds to a binding molécule (for example, an antibody or a related molécule, such as a bispecific binding molécule). Epitope déterminants usually consist of chemically active surface groupings of molécules such as amino acids or carbohydrates or sugar side chains and typically comprise spécifie three-dimensional structural characteristics, as well as spécifie charge characteristics. Epitopes can be either linear or conformational. In a linear epitope, ail of the points of interaction between a protein (e.g., an antigen) and an interacting molécule (such as an antibody) occur linearly along the primary amino acid sequence of the protein. In a conformational epitope, the points of interaction occur across amino acid residues on the protein that are separated from one another in the primary amino acid sequence. Once a desired epitope of an antigen is determined, it is possible to generate antibodies to that epitope using techniques well known in the art. In addition, génération and characterization of antibodies or other binding molécules may elucidate information about désirable epitopes.
From this information, it is then possible to competitively screen antibodies for binding to the same or identical epitopes, e.g., by conducting compétition studies to find binding molécules that compete with one another for binding to the antigen. As used herein, the term epitope, inter alia, refers to a polypeptide fragment, having antigenic and/or immunogenic activity in animais, preferably in mammals such as mice and humans. The term antigenic epitope as used herein is a polypeptide fragment which can specifically bind to the antibody and can be detected by any technique well known from the prior art, for example, by a standard immunoassay. Antigen epitopes are not necessarily immunogenic; however, they can be immunogenic. Immunogenic epitope as used herein is defmed as a polypeptide fragment that evokes an antibody response in animais, as determined by any method known from the prior art. Nonlinear epitope or conformational epitope comprises nonadjacent polypeptides (or amino acids) within an antigen protein that binds to epitope-specific antibody.
One can détermine whether an antibody or other binding molécule binds to the same epitope or cross-competes for binding with a PD-1 binding molécule of the présent invention by using methods known in the art. In one embodiment, one allows a molécule of the invention to bind to PD-1 under saturating conditions and then measures the ability of the test antibody to bind to said target antigen. If the test antibody is able to bind to the target antigen at the same time as a référencé binding molécule, then the test antibody binds to a different epitope than that of the référencé binding molécule. However, if the test antibody is not able to bind to the target antigen at the same time, then the test antibody binds to the same epitope, an overlapping epitope, or an epitope that is in close proximity to the epitope bound to the binding molécule. This experiment can be performed using ELISA, RIA, BIACORETM, bio-layer interferometry or flow cytometry. To test whether a binding molécule of the invention cross-competes with another binding molécule, one may use the compétition method described above in two directions, i.e. determining if the known binding molécule blocks the test binding molécule and vice versa. Such cross-competition experiments may be performed, e.g., using IBIS MX96 SPR or OctetTM system.
In one embodiment, a binding molécule of the invention is a monoclonal antibody. As used herein, the acronym mAb is intended to refer to a monoclonal antibody, i.e. an antibody synthesized and isolated by a separate clonal population of cells. A clonal population can be a clonal population of immortalized cells. In some embodiments, the immortalized cells in a clonal population are hybrid cells -hybridomas - typically produced by the fusion of individual B lymphocytes from immunized animais with individual cells from a lymphocytic tumour. Hybridomas are a type of constructed cells and do not exist in nature.
The class (isotype) and subclass of antibodies can be determined by any method known in the art. In general, the class and subclass of an antibody can be determined by antibodies spécifie to a certain class and subclass of antibodies. Such antibodies are commercially available. The class and subclass can be determined using ELISA, western blot analysis, and other methods. In another embodiment, the class and subclass can be determined by virtue of sequencing ail or part of the heavy and/or light chain constant domains of antibodies, comparing amino acid sequences thereof with known amino acid sequences of various classes and subclasses of immunoglobulins, and determining the class and subclass of antibodies.
The ternis monoclonal antibody or monoclonal antibody composition as used herein refer to a préparation of antibody molécules of a single molecular composition. A monoclonal antibody composition displays single binding specificity and affinity with respect to a particular antigen epitope.
The terni human antibody as used herein is intended to include antibodies comprising variable régions in which both framework and CDRs are derived from human germ-line immunoglobulin sequences. Furthermore, if said antibody contains a constant région, the constant région is also derived from human germ-line immunoglobulin sequences.
The human antibodies of the invention may include amino acid residues not encoded by human germ-line immunoglobulin sequences (e.g., mutations introduced by random or site-specific in vitro mutagenesis, or an in vivo somatic mutation). However, the term human antibody as used herein is not intended to include antibodies in which CDR sequences derived from the germ-line of another mammalian species, such as a mouse, hâve been grafted onto human framework sequences.
The term human monoclonal antibody is intended to refer to antibodies displaying a single binding specificity which hâve variable régions in which both the framework and CDRs are derived from human germ-line immunoglobulin sequences. In one embodiment, the human monoclonal antibodies are produced by a hybridoma which includes a B cell obtained from a transgenic nonhuman animal, e.g., a transgenic mouse, having a genome comprising a human heavy chain transgene and human light chain transgene fused to an immortalized cell.
The term recombinant human antibody as used herein includes ail human antibodies that are prepared, expressed, engineered or isolated by recombinant means, such as (a) antibodies isolated from an animal (e.g., a mouse) that is transgenic or transchromosomal for human immunoglobulin genes or a hybridoma prepared therefrom (described further below), (b) antibodies isolated from a host cell transformed to express a human antibody, e.g., from a transfectoma, (c) antibodies isolated from a recombinant, combinatorial human antibody library, and (d) antibodies prepared, expressed, engineered or isolated by any other means that involve splicing of human immunoglobulin gene sequences to other DNA sequences. Such recombinant human antibodies hâve variable régions in which framework régions and CDRs are derived from human germ-line immunoglobulin sequences. In certain embodiments, however, such recombinant human antibodies can be subjected to in vitro mutagenesis (or, when an animal transgenic for Ig sequences is used, somatic mutagenesis) and thus the amino acid sequences of the VH and
VL régions of recombinant antibodies are sequences that, while derived from or related to human germ-line VH and VL sequences, may not naturally exist within the human antibody germ-line répertoire in vivo.
The phrases an antibody recognizing an antigen and an antibody spécifie for an antigen are used interchangeably herein with the term an antibody which binds specifically to an antigen.
The term antibody variant as used herein is intended to refer to a molécule the amino acid sequence of which differs from a parental sequence by virtue of addition, délétion and/or, substitution of one or more amino acid residues in the sequence of a parental antibody. In a preferred embodiment, a variant antibody comprises at least one (for example, from one to about ten preferably 2, 3, 4, 5, 6, 7 or 8) amino acid addition, délétion and/or substitution in the CDRs of a parental antibody. This application defines identity or homology regarding the sequence of a variant antibody as the percentage of amino acid residues in a variant antibody sequence that are identical to residues in a parental antibody after aligning the sequences and, if needed, cutting in order to achieve a maximum percentage identical sequence. A variant antibody retains the ability to bind the same antigen or, preferably, epitope as that with which a parental antibody binds, or, preferably, exhibits at least one property or biological activity exceeding that of a parental antibody. For example, an antibody preferably has stronger affmity, longer half-life, lower IC50 or enhanced ability to inhibit antigen biological activity, as compared to a parental antibody. The variant antibody of particular interest herein is one which displays at least about 2-fold, preferably at least about 5-fold, I0-fold or 20-fold enhancement in biological activity as compared to a parental antibody.
The term identity or homology in the context of nucleic acid sequences is intended to refer to the residues in two sequences that are the same when aligned for maximum correspondence. Comparison of sequence identity may extend over a length of at least about nine nucléotides, commonly at least about 18 nucléotides, more commonly at least about 24 nucléotides, typically at least about 28 nucléotides, more typically at least about 32 nucléotides, and preferably at least about 36, 48 or more nucléotides. There are a number of various algorithms known in the art which can be used to measure nucléotide sequence identity. For example, polynucleotide sequences can be compared using FASTA, Gap or BESTFIT, which are programs in Wisconsin Package Version 10.0, Genetics Computer Group (GCG), Madison, Wisconsin. FASTA, which includes, e.g., FASTA2 and FASTA3 programs, provides alignments and percent sequence identity of the régions of the best overlap between the query and search sequences (Pearson, Methods Enzymol. 183:63 98 (1990); Pearson, Methods Mol. Biol. 132: 185-219 (2000); Pearson, Methods Enzymol. 266: 227258 (1996); Pearson, J. Mol. Biol. 276: 71-84 (1998)). Unless otherwise specified, default parameters for a particular program or algorithm are used. For instance, percent sequence identity between nucleic acid sequences can be determined using FASTA with default parameters (word size of 6 and NOPAM factor for the scoring matrix) or using Gap with default parameters as provided in GCG Version 6.1.
The term homologous with regard to a polypeptide sequence of an antibody should be construed as an antibody exhibiting at least 70%, preferably 80%, more preferably 90% and most preferably 95% sequence identity relative to a polypeptide sequence. The term in relation to a nucleic acid sequence should be construed as a sequence of nucléotides exhibiting at least 85%, preferably 90%, more preferably 95% and most preferably 97% sequence identity relative to a nucleic acid sequence.
As used herein, a parental antibody is an antibody encoded by an amino acid sequence, which is used for obtaining a variant.
The antibodies of the invention can be prepared by various design techniques, including using recombinant methods, including the shuffling of DNA obtained from various sources.
The term humanized antibody is intended to refer to antibodies in which CDR sequences derived from the germ-line of another mammalian species, such as a mouse, hâve been grafted onto human framework sequences. Additional framework région modifications may be made within such human framework sequences.
The term chimeric antibody is intended to refer to antibodies in which the variable région sequences are derived from one species and the constant région sequences are derived from another species, such as an antibody in which the variable région sequences are derived from a murine antibody and the constant région sequences are derived from a human antibody.
The term specifically binds as used herein is intended to refer to the situation in which one member of a spécifie binding pair does not significantly bind to molécules other than spécifie binding partner(s) thereof. The term is also applicable where e.g. an antigen-binding domain of an antibody of the invention is spécifie for a particular epitope that is carried by a number of antigens; in this case, the spécifie antibody comprising the antigen-binding domain will be able to specifically bind to various antigens carrying the epitope.
As used herein, an antibody that specifically binds to human PD-1 is intended to refer to an antibody that binds to human PD-1 with KD of 1x 10' M
S or less, more preferably 5x10' M or less, more preferably 1x10' M or less, more preferably 5X 10‘9M or less.
The term bispecific antibody or multispecific antibody includes an antibody capable of selectively binding two or more epitopes. Bispecific antibodies, e.g., may comprise two different antigen-binding portions, wherein said antigen-binding portions specifically bind different epitopes either on different molécules (e.g., antigens), or on the same molécule (e.g., on the same antigen). If a bispecific antibody is able to selectively bind two different epitopes (a first epitope and second epitope), the affinity of the first antigenbinding portion for the first epitope will typically be at least one to two, or three, or four orders of magnitude lower than that of the first antigen-binding portion for the second epitope, and vice versa. Epitopes recognized by a bispecific antibody may be the same or different targets (e.g., on the same or a different protein). Bispecific antibodies can be prepared, for example, by combining heavy chains that recognize different epitopes on the same antigen. For example, nucleic acid sequences encoding variable heavy chain sequences that recognize different epitopes may be fused to nucleic acid sequences encoding various heavy chain constant régions, and such sequences may be expressed in a cell which expresses an immunoglobulin light chain. A typical bispecific antibody comprises two heavy chains, each comprising three heavy chain CDRs followed (from N-terminus to C-terminus) by a CH1 domain, hinge région, CH2 domain and CH3 domain, and immunoglobulin light chain which either does not hâve antigen-binding specificity but is able to combine with each of the heavy chains, or is able to combine with each of the heavy chains and bind one or more epitopes restricted by antigen-binding heavy chain régions, or is able to combine with each of the heavy chains and promûtes binding of one or the both heavy chains to one or the both epitopes.
The phrases biological property or bioactivity, activity or biological activity, in reference to an antibody of the présent invention, are used interchangeably herein and include, but are not limited to, epitope/antigen affinity and specificity, ability to neutralize or antagonize an activity of PD-1 in vivo or in vitro, IC50, the stability of an antibody and immunogenic properties of an antibody in vivo. Other identifiable biological properties of an antibody include, for example, cross-reactivity, (i.e., with non-human homologs of a target peptide, or with other proteins or tissues, generally), and an ability to preserve high levels of expression of protein in mammalian cells. Said properties or characteristics can be observed, measured or assessed using techniques recognized in the art, including, but not limited to, ELISA, compétitive ELISA, antigen-antibody interactions by surface plasmon résonance using BIACORE or KINEXA, or bio-layer interferometry using ForteBio, in vitro or in vivo neutralization assays without limitation, receptor binding, production and/or sécrétion of a cytokine or growth factor, signal transduction and immunohistochemistry of tissue sections from various sources including human, primate, or any other source.
The term inhibit or neutralize as used herein with respect to the activity of an antibody of the invention is intended to refer to the ability to substantially antagonize, prohibit, prevent, restrain, slow, disrupt, eliminate, stop, reduce or reverse, e.g., progression or severity of that which is being inhibited including, but not limited to, a biological activity (e.g., the activity of PD-1) or property, disease or condition. The inhibition or neutralization of activity of PD-1 resulted from binding an antibody of the invention to PD-1 is preferably at least about 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95% or higher.
PD-1 binding molécules
The présent invention relates to a binding molécule that has the ability to bind to a human PD-1 receptor that contains an amino acid sequence that is at least 75% homologous to the sequence of SEQ ID NO:3, for example, at least 91%, 92%, 93% %, 94%, 95%, 96%, 97%, 98% or 99% identical to the sequence of SEQ ID NO:3.
In some embodiments, the heavy chain (HC) of an anti-PD 1 antibody is at least 60% identical to the sequence of SEQ ID NO:1, for example, at least 60%, 70% or 80% identical to the sequence of SEQ ID NO:2. In some embodiments, the heavy chain (HC) of the anti-PD 1 antibody is at least 90% identical to the sequence of SEQ ID NO: 7, for example, at least 91%, 92%, 93%, 94%, 95%, 96 %, 97%, 98%, or 99% identical to the sequence of SEQ ID NO: 7. In a particular embodiment, an HC comprises or consists of the amino acid sequence ofSEQ ID NO: 5.
In some embodiments, the light chain of an anti-PD 1 antibody comprises a light chain CDR1 (L-CDR1) amino acid sequence of SEQ ID NO:4, light chain CDR2 (L-CDR2) amino acid sequence of SEQ ID NO:5, light chain CDR3 (L-CDR3) amino acid sequence of SEQ ID NO:6, or any combination thereof. In some embodiments, the light chain of PDI antibody comprises amino acid sequences L-CDR1, L-CDR2 and L-CDR3 shown in SEQ ID NOs: 4, SEQ ID NO: 5 and SEQ ID NO: 6. In some embodiments, the light chain of an antiPD1 antibody comprises a light chain variable domain (VL) that is at least 60% identical to the sequence of SEQ ID NO:8, for example, at least 60%, 70%, or 80% identical to the sequence SEQ ID NO:8. In some embodiments, the light chain of an anti-PDl antibody comprises a light chain variable domain (VL) that is at least 90% identical to the sequence of SEQ ID NO:8, for example, at least 91%, 92%, 93%, 94 %, 95%, 96%, 97%, 98% or 99% identical to the sequence of SEQ ID NO:8. In a particular embodiment, a VL domain comprises or consists of the amino acid sequence of SEQ ID NO:8.
In some embodiments, the light chain (LC) of an anti-PDl antibody is at least 60% identical to the sequence of SEQ ID NO: 10, for example, at least 60%, 70%, or 80% identical to the sequence of SEQ ID NO: 10. In some embodiments, the light chain (LC) of a PDI antibody is at least 90% identical to the sequence of SEQ ID NO: 10, for example, at least 91%, 92%, 93%, 94%, 95%, 96% 97%, 98%, or 99% identical to the sequence of SEQ ID NO: 10. In a particular embodiment, a VL domain comprises or consists of the amino acid sequence of SEQ ID NO: 10.
The class of a binding molécule obtained using techniques described herein may be switched with another class or subclass. In one aspect of the invention, a nucleic acid molécule encoding a VL or VH is isolated using methods well-known in the art such that it does not include nucleic acid sequences encoding a CL or CH. The nucleic acid molécules encoding VL or VH were operatively linked to a nucleic acid sequence encoding a CL or CH, respectively, from a different class of immunoglobulin molécule. This may be achieved using a vector or nucleic acid molécule that comprises a CL or CH chain, as described above. For example, a binding molécule that was originally IgM may be class-switched to IgG. Further, class-switching may be used to convert one IgG subclass to another, e.g., from IgGl to IgG2. An exemplary method for producing a binding molécule of the invention with a desired isotype comprises the steps of isolating a nucleic acid molécule encoding the heavy chain of a binding molécule and a nucleic acid molécule encoding the light chain of a binding molécule, obtaining the variable domain of the heavy chain, ligating the variable domain of the heavy chain with the constant domain of a heavy chain of the desired isotype, expressing the light chain and the ligated heavy chain in a cell, and obtaining the binding molécule with the desired isotype.
A binding molécule of the invention can be an IgG, IgM, IgE, IgA, or IgD molécule, but is typically of the IgG isotype, e.g., IgGl, lgG2a or b, IgG3, or IgG4 of the IgG subclass. In one embodiment, a binding molécule is an IgGl antibody of the IgG subclass.
In one embodiment, a binding molécule may comprise at least one mutation in the Fc région. A number of various Fc mutations are known, where these mutations provide altered effector fonction. For example, in many cases it will be désirable to reduce or eliminate the effector fonction, e.g., where ligandreceptor interaction is undesired or in the case of antibody-drug conjugates. Amino acid Fc région positions, which can be advantageously mutated to reduce the effector fonction, include one or more of positions 228, 233, 234 and 235, wherein amino acid positions are numbered according to the Kabat numbering scheme. In some embodiments, the binding molécule comprises an Fc région of at least one mutation that reduces ADCC and/or CDC, compared with the same binding molécule without mutations.
In some embodiments, a binding molécule of the invention may be part of a larger immunoadhesion molécule formed by covalent or noncovalent association of an antibody or antibody portion with one or more other proteins or peptides. Examples of such immunoadhesion molécules include use of a streptavidin core région to make a tetrameric scFv molécule (Kipriyanov et al., Human Antibodies and Hybridomas 6:93-101 (1995)) and use of a cysteine residue, a marker peptide and a C-terminal polyhistidine tag to make bivalent and biotinylated scFv molécules (Kipriyanov et al., Mol. Immunol. 31:10471058 (1994)). Other examples include where one or more CDRs from an antibody are incorporated into a molécule either covalently or noncovalently to produce an immunoadhesin that specifically binds to the antigen of interest. In such embodiments, CDRs may be incorporated as part of a larger polypeptide chain, may be covalently linked to another polypeptide chain, or may be incorporated noncovalently.
In a further embodiment, a fusion antibody or immunoadhesin may be produced which comprises ail or a portion of a binding molécule of the invention linked to another polypeptide. In some embodiments, only the variable régions of a binding molécule are linked to a polypeptide. In some embodiments, the VH domain of a binding molécule is linked to a first polypeptide, while the VL domain of a binding molécule is linked to a second polypeptide that associâtes with the first polypeptide in a manner in which the VH and VL domains can interact with one another to form an antigen-binding site. In another preferred embodiment, the VH domain is separated from the VL domain by a linker such that the VH and VL domains can interact with one another (e.g., single chain antibodies). The VH-linker-VL antibody is then linked to the polypeptide of interest. Furthermore, fusion antibodies can be created in which two (or more) single chain antibodies are linked to one another. This is useful if one wants to engineer a bivalent or polyvalent antibody on a single polypeptide chain, or if one wants to engineer multispecific antibodies.
To engineer a single chain antibody (scFv), VH- and VL-encoding DNA fragments are operatively linked to another fragment encoding a flexible linker, e.g., encoding an amino acid sequence (Gly4 -Ser)3 such that the VH and VL sequences can be expressed as a contiguous single chain protein with the VL and VH domains joined by a flexible linker. See, e.g., Bird et al., Science 242:423 426 (1988); Huston et al., Proc. Natl. Acad. Sci. USA 85:5879 5883 (1988); and McCafferty et al., Nature 348:552 554 (1990). The single chain antibody may be monovalent, if only a single VH and VL domain are used, bivalent, if two VH and VL domains are used, or polyvalent, if more than two VH and VL domains are used.
A binding molécule of the invention can be derivatized or linked to another molécule (e.g., another peptide or protein). In general, binding molécules (e.g., antibodies or antigen-binding portions thereof) are derivatized such that the PD-1 binding is not affected adversely by derivatization or labeling. Thus, the binding molécules of the invention can include both intact and modified forms of binding molécules described herein. For example, a binding molécule of the invention can be functionally linked (by virtue of Chemical coupling, genetic fusion, noncovalent association or otherwise) to one or more molecular entities, such as another antibody, a détection agent, a pharmaceutical agent, and/or a protein or peptide that can médiate association of the binding molécule with another molécule (such as a streptavidin core région or polyhistidine tag).
One type of a derivatized binding molécule is produced by crosslinking two or more antibodies (of the same type or of different types, e.g., to engineer bispecific antibodies). Suitable crosslinkers include those that are heterobifunctional, having two different reactive groups separated by a suitable spacer (e.g., m-maleimidobenzoyl-N-hydroxysuccinimide ester) or homobifunctional (e.g., disuccinimidyl suberate).
A binding molécule of the invention may also be derivatized with a Chemical group such as polyethylene glycol (PEG), a methyl or ethyl group, or a carbohydrate group. These groups may be useful to improve the biological characteristics of a binding molécule, e.g., to increase sérum half-life.
A binding molécule of the invention can also be labeled. As used herein, the ternis label or labeled refer to incorporation of another molécule in a binding molécule. In one embodiment, a label is a détectable marker, e.g., incorporation of a radioactive amino acid or attachment of biotinyl fragments to a polypeptide, and such fragments can be detected by labeled avidin (e.g., streptavidin comprising a fluorescent marker or enzymatic activity that can be detected by optical or colorimétrie techniques). In a further embodiment, a label or marker can be therapeutic, e.g., a drug conjugate or toxin. Various methods of labeling polypeptides and glycoproteins are known in the art and may be used. Examples of labels for polypeptides include, inter alia, the following: radioisotopes or radionuclides (e.g., 3H, 14C, 15N, 35S, 90Y, 99Tc, 11 lin, 1251, 1311), fluorescent labels (e.g., FITC, rhodamine, lanthanide phosphors), enzymatic labels (e.g., horseradish peroxidase, β-galactosidase, luciferase, alkaline phosphatase), chemiluminescent markers, biotinyl groups, predetermined polypeptide epitopes recognized by a secondary reporter (e.g., leucine zipper pair sequences, binding sites for secondary antibodies, métal binding domains, epitope tags), magnetic agents, such as gadolinium chelates, toxins such as pertussis toxin, taxol, cytochalasin, gramicidin D, ethidium bromide, emetine, mitomycin, etoposide, tenoposide, vincristine, vinblastine, colchicine, doxorubicin, daunorubicin, dihydroxy anthracin dione, mitoxantrone, mithramycin, actinomycin D, 1-dehydrotestosterone, glucocorticoids, procaine, tetracaine, lidocaine, propranolol, and puromycin and analogs or homologs thereof. In some embodiments, labels are attached by spacer arms of various lengths to reduce potential steric hindrance.
In certain embodiments, the binding molécules of the invention may be présent in a neutral form (including zwitterionic forms) or as a positively or negatively charged species. In some embodiments, the antibodies may be complexed with a counterion to form a pharmaceutically acceptable sait.
The term pharmaceutically acceptable sait is intended to refer to a complex comprising one or more binding molécules and one or more counterions, where the counterions are derived from pharmaceutically acceptable inorganic and organic acids and bases.
Pharmaceutically acceptable inorganic bases include metallic ions including, inter alia, suitable alkali métal salts, alkaline earth métal salts and other physiological ions of acceptable metals. Salts derived from inorganic bases include aluminum, ammonium, calcium, cobalt, nickel, molybdenum, vanadium, manganèse, chromium, sélénium, tin, copper, ferrie, lithium, magnésium, manganic or manganous, potassium, rubidium, sodium, and zinc salts, and in their typical valences.
Pharmaceutically acceptable acid addition salts of the binding molécules of the invention can be prepared from the following acids, including, inter alia, formic, acetic, acetamidobenzoic, adipic, ascorbic, boric, propionic, benzoic, camphoric, carbonic, cyclamic, dehydrocholic, malonic, edetic (ethylenediaminetetraacetic), ethylsulfuric, fendizoic, metaphosphoric, succinic, glycolic, gluconic, lactic, malic, tartaric, tannic, citric, nitric, glucuronic, maleic, folie, fumaric, pyruvic, aspartic, glutamic, hydrochloric, hydrobromic, hydroiodic, lysine, isocitric, trifluoroacetic, pamoic, anthranilic, mesylic, orotic, oxalic, oxalacetic, oleic, stearic, salicylic, aminosalicylic, silicate, phydroxybenzoic, nicotinic, phenylacetic, mandelic, embonic, sulfonic, methanesulfonic, phosphoric, phosphonic, ethanesulfonic, ethanedisulfonic, ammonium, benzenesulfonic, pantothenic, naphthalenesulfonic, toluenesulfonic, 2-hydroxyethanesulfonic, sulfanilic, sulfuric, nitric, nitrous, sulfuric acid monomethyl ester, cyclohexylaminosulfonic, β-hydroxybutyric, glycine, glycylglycine, cacodylate, diaminohexanoic, camphorsulfonic, thiocyanic, oxoglutaric, pyridoxal 5-phosphate, chlorophenoxyacetic, undecanoic, N-acetylL-aspartic, galactaric and galacturonic acids.
Pharmaceutically acceptable organic bases include trimethylamine, diethylamine, Ν,Ν'-dibenzylethylenediamine, chloroprocaine, choline, dibenzylamine, diethanolamine, ethylenediamine, meglumine (Nmethylglucamine), procaine, cyclic amines, quatemary ammonium cations, arginine, betaine, caffeine, clemizole, 2-ethylaminoethanol, 2diethylaminoethanol, 2-dimethylaminoethanol, ethanediamine, butylamine, ethanolamine, ethylenediamine, N-ethylmorpholine, N-ethylpiperidine, ethylglucamine, glucamine, glucosamine, histidine, hydrabamine, imidazole, isopropylamine, methylglucamine, morpholine, piperazine, pyridine, pyridoxine, neodymium, piperidine, polyamine resins, procaine, purines, theobromine, triethylamine, tripropylamine, triethanolamine, tromethamine, methylamine, taurine, cholate, 6-amino-2-methyl-2-heptanol, 2-amino-2-methyl-l,3propanediol, 2-amino-2-methyl-l-propanol, aliphatic mono- and dicarboxylic acids, phenyl-substituted alkanoic acids, hydroxy alkanoic acids, aromatic acids, aliphatic and aromatic sulfonic acids, strontium, tricine, hydrazine, phenylcyclohexylamine, 2-(N-morpholino)ethanesulfonic acid, bis(2hydroxyethyl)amino-tris(hydroxymethyl)methane, N-(2-acetamido)-2aminoethanesulfonic acid, 1,4-piperazinediethanesulfonic acid, 3-morpholino-2hydroxypropanesulfonic acid, 1,3bis[tris(hydroxymethyl)methylamino]propane, 4-morpholinepropanesulfonic acid, 4-(2-hydroxyethyl)piperazine-1-ethanesulfonic acid, 2-[(2-hydroxy-l,lbis(hydroxymethyl)ethyl)amino] ethanesulfonic acid, N,N-bis(2-hydroxyethyl)2-aminoethanesulfonic acid, 4-(N-morpholino)butanesulfonic acid, 3-(N,Nbis[2-hydroxyethyl]amino)-2-hydroxy propanesulfonic acid, 2-hydroxy-3-[tris (hydroxymethyl)methylamino]-l-propanesulfonic acid, 4-(2hydroxyethyl)piperazine-1 -(2-hydroxypropane sulfonic acid), piperazine-1,4bis(2-hydroxypropanesulfonic acid) dihydrate, 4-(2-hydroxyethyl)-lpiperazinepropanesulfonic acid, N,N-bis(2-hydroxyethyl) glycine, N-(2hydroxyethyppiperazine-N'-(4-butanesulfonic acid), N[tris(hydroxymethyl)methyl]-3-aminopropanesulfonic acid, Ntris(hydroxymethyl)methyl-4-aminobutanesulfonic acid, N-( 1,1 -dimethyl-2hydroxyethyl)-3-amino-2-hydroxy propanesulfonic acid, 2-(cyclohexylamino) ethanesulfonic acid, 3-(cyclohexylamino)-2-hydroxy-1 -propanesulfonic acid, 3(cyclohexylamino)-l-propanesulfonic acid, N-(2-acetamido)iminodiacetic acid, 4-(cyclohexylamino)-l-butanesulfonic acid, N-[tris(hydroxymethyl)methyl] glycine, 2-amino-2-(hydroxymethyl)-l,3-propanediol, and trometamol.
Nucleic acid molécules and vectors
The présent invention also relates to nucleic acid molécules, and sequences encoding the binding molécules of the invention described herein. In some embodiments, various nucleic acid molécules encode the first domain and second domain of the amino acid sequence of a binding molécule. In some embodiments, wherein a first domain and/or second domain comprises a heavy chain and light chain, various nucleic acids encode a heavy chain and light chain amino acid sequences. In other embodiments, the same nucleic acid molécule encodes a heavy chain and light chain amino acid sequences. In certain embodiments, a nucleic acid molécule can encode any combination of amino acid sequences (e.g., heavy and light chain sequences) of fîrst and second domains. In a particular embodiment, a nucleic acid molécule can encode the amino acid sequence of a fîrst binding domain and the light chain amino acid sequence of a second binding domain, optionally including any sequence of a peptide linker connecting them.
A reference to a nucléotide sequence encompasses the complément thereof unless otherwise specified. Thus, a reference to a nucleic acid having a particular sequence should be understood as one which encompasses the complementary strand thereof with the complementary sequence thereof. The terni polynucleotide as used herein means a polymeric form of either nucléotides that are at least 10 bases in length, or ribonucleotides, or deoxyribonucleotides or a modified form of either type of nucléotide. The term includes single and double stranded forms.
The présent invention also relates to nucléotide sequences that are at least 70%, 75%, 80%, 85%, 90%, 95%, 97%, 98%, or 99% identical to one or more of said nucléotide sequences or a nucléotide sequence encoding an amino acid sequence selected from a group consisting of SEQ ID NO: 1-10. In certain embodiments, nucléotide sequences are at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% identical to a nucléotide sequence encoding an amino acid sequence selected from a group consisting of SEQ ID NO: 4-9. The term percent sequence identity in the context of nucleic acid sequences is intended to refer to the residues in the two sequences which are the same when aligned for maximum correspondence. Comparison of sequence identity may extend over a length of at least about nine nucléotides, commonly at least about 18 nucléotides, more commonly at least about 24 nucléotides, typically at least about 28 nucléotides, more typically at least about 32 nucléotides, and preferably at least about 36, 48 or more nucléotides. There are a number of various algorithms known in the art which can be used to measure nucléotide sequence identity. For example, polynucleotide sequences can be compared using FASTA, Gap or BESTFIT, which are programs in Wisconsin Package Version 10.0, Genetics Computer Group (GCG), Madison, Wisconsin. FASTA, which includes, e.g., FASTA2 and FASTA3 programs, provides alignments and percent sequence identity of the régions of the best overlap between the query and search sequences (Pearson, Methods Enzymol. 183:63 98 (1990); Pearson, Methods Mol. Biol. 132: 185-219 (2000); Pearson, Methods Enzymol. 266: 227258 (1996); Pearson, J. Mol. Biol. 276: 71-84 (1998); incorporated herein by reference). Unless otherwise specified, default parameters for a particular program or algorithm are used. For example, percent sequence identity between nucleic acid sequences can be determined using FASTA with default parameters (word size of 6 and NOPAM factor for the scoring matrix) or using Gap with default parameters as provided in GCG Version 6.1, herein incorporated by reference.
In one aspect, the présent invention relates to a nucleic acid molécule comprising a nucléotide sequence encoding an amino acid sequence selected from SEQ ID NO: 1-10. A nucleic acid molécule can also comprise any combination of said nucléotide sequences. In one embodiment, a nucleic acid molécule comprises a nucléotide sequence encoding SEQ ID NO: 4. In a further embodiment, a nucleic acid molécule comprises a nucléotide sequence encoding SEQ ID NO: 4 and 6. In a further embodiment, a nucleic acid molécule comprises a nucléotide sequence encoding SEQ ID NO: 4, 6 and 16. In a further embodiment, a nucleic acid molécule comprises a nucléotide sequence encoding SEQ ID NO: 7. In a further embodiment, a nucleic acid molécule comprises a nucléotide sequence encoding SEQ ID NO: 5.
In any of the above embodiments, nucleic acid molécules can be isolated.
In another aspect, the présent invention relates to a vector suitable for the expression of any of nucléotide sequences described herein. The term vector as used herein means a nucleic acid molécule capable of transporting another nucleic acid to which it has been linked. In some embodiments, a vector is a plasmid, i.e., a circular double stranded piece of DNA into which additional DNA segments may be ligated. In some embodiments, a vector is a viral vector, wherein additional DNA segments may be ligated into the viral genome. In some embodiments, vectors are capable of autonomous réplication in a host cell into which they are introduced (e.g., bacterial vectors having a bacterial origin site of réplication and episomal mammalian vectors). In further embodiments, vectors (e.g., non-episomal mammalian vectors) can be integrated into the genome of a host cell upon introduction into a host cell, and thereby are replicated along with the host gene. Moreover, certain 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).
The présent invention relates to vectors comprising nucleic acid molécules that encode any of the amino acid sequences of binding molécules or portions thereof (e.g., heavy and/or light chain sequences of the first and/or second binding domains) as described herein. The invention further provides vectors comprising nucleic acid molécules encoding fusion proteins, modified antibodies, antibody fragments, and probes thereof.
A nucleic acid molécule of the invention can be isolated from any source that produces binding molécule or a portion thereof. In certain embodiments, a nucleic acid molécule of the invention can be synthesized, rather than isolated.
In some embodiments, a nucleic acid molécule of the invention can comprise a nucléotide sequence encoding a VH domain from the first or second domain of a binding molécule of the invention, joined in-frame to a nucléotide sequence encoding a heavy chain constant domain from any source. Similarly, a nucleic acid molécule of the invention can comprise a nucléotide sequence encoding a VL domain from the first or second région of a binding molécule of the invention, joined in-frame to a nucléotide sequence encoding a light chain constant domain from any source.
In a further aspect of the invention, nucleic acid molécules encoding the variable domain of heavy (VH) and/or light (VL) chains of a first or second binding domain may be converted throughout the length of antibody genes. In one embodiment, nucleic acid molécules encoding VH or VL domains are converted to antibody genes along the entire the length by virtue of insertion into an expression vector already encoding heavy chain constant (CH) or light chain constant (CL) domains, respectively, such that the VH segment is operatively linked to the CH segment(s) within the vector, and/or the VL segment is operatively linked to the CL segment within the vector. In another embodiment, nucleic acid molécules encoding the VH and/or VL domains are converted into antibody genes throughout the length by virtue of linking, e.g., ligating, a nucleic acid molécule encoding VH and/or VL domains to a nucleic acid molécule encoding CH and/or CL domains using standard molecular biological techniques. Nucleic acid molécules encoding heavy and/or light chains throughout the length may then be expressed from a cell into which they hâve been introduced.
Nucleic acid molécules may be used to express large quantities of recombinant binding molécules. Nucleic acid molécules may also be used to produce human antibodies, humanized antibodies, chimeric antibodies, bispecific antibodies, single chain antibodies, immunoadhesins, diabodies, mutated antibodies and antibody dérivatives, as described herein.
In another embodiment, a nucleic acid molécule of the invention is used as a probe or PCR primer for a spécifie antibody sequence. For example, a nucleic acid can be used as a probe in diagnostic techniques or as a PCR primer to amplify régions of DNA that could be used, e.g., to isolate additional nucleic acid molécules encoding binding molécule régions (e.g., variable domains). In some embodiments, nucleic acid molécules are oligonucleotides. In some embodiments, oligonucleotides are from highly variable domains of a binding molécule. In some embodiments, oligonucleotides encode ail or a part of one or more of the CDRs of a binding molécule of the invention as described herein.
In another embodiment, nucleic acid molécules and vectors may be used to make mutated binding molécules. Antibodies may be mutated in the variable domains of the heavy and/or light chains of a first and/or second binding domain, e.g., to alter a binding property of a binding molécule. For example, a mutation may be made in one or more of CDRs to increase or decrease the KD of a binding molécule, to increase or decrease koff, or to alter the binding specificity of an antibody with respect to PD-1. In another embodiment, one or more mutations are made at an amino acid residue that is known to be changed compared to the germ-line in an antibody corresponding to the first or second binding domain of a binding molécule of the invention. Such mutations may be made in the CDR or framework région of a variable domain, or in a constant domain. In a preferred embodiment, mutations are made in a variable domain. In another embodiment, one or more mutations are made at an amino acid residue that is known to be changed compared to the germ-line in the CDR or framework région of a variable domain of a binding molécule of the invention.
In another embodiment, a framework region(s) is mutated so that the resulting framework region(s) has the amino acid sequence of the corresponding germ-line gene. Such mutations may be made in a framework région or constant domain to increase the half-life of a binding molécule. See, e.g., WO 00/09560. A mutation in a framework région or constant domain can also be made to alter the immunogenicity of a binding molécule and/or to provide a site for covalent or non-covalent binding to another molécule. According to the invention, a binding molécule may hâve mutations in any one or more of the CDRs or framework régions of a variable domain or in a constant domain.
In some embodiments, the binding molécules of the invention are expressed by inserting a DNA partially or fully encoding the sequence of a first or second binding domain (e.g., light and heavy chain sequences where a binding domain comprises light and heavy chain sequences), obtained as described above, in expression vectors such that the genes are operatively linked to necessary expression control sequences, such as transcriptional and translational control sequences. Expression vectors include plasmids, retroviruses, adenoviruses, adeno-associated viruses (AAV), plant viruses, such as cauliflower mosaic virus, tobacco mosaic virus, cosmids, YACs, EBV derived episomes, and the like. DNA molécules may be ligated into a vector such that transcriptional and translational control sequences within the vector serve their intended fonction of regulating the transcription and translation of the DNA. An expression vector and expression control sequences may be chosen to be compatible with the expression host cell used. DNA molécules partially or folly encoding the sequences of first and second binding domains (for example, heavy and light chain sequences where a binding domain comprises a heavy and light chain sequence) can be introduced into individual vectors. In one embodiment, any combination of said DNA molécules is introduced into the same expression vector. DNA molécules can be introduced into an expression vector by standard methods (e.g., ligation of complementary restriction sites on an antibody gene fragment and vector, or blunt end ligation if no restriction sites are présent).
A suitable vector is one that encodes fonctionally complété human CH or CL immunoglobulin sequences, with appropriate restriction site engineering so that any VH or VL sequence can easily be inserted and expressed, as described above. HC- and LC-encoding genes in such vectors may contain intron sequences that results in enhanced overall antibody protein yields by stabilizing the corresponding mRNA. The intron sequences are flanked by splice donor and splice accepter sites, which déterminé where RNA splicing will occur. Location of intron sequences can be either in variable or constant régions of antibody chains, or in both variable and constant régions when multiple introns are used. Polyadenylation and transcription termination may occur at a native chromosomal site downstream of coding régions. A recombinant expression vector can also encode a signal peptide that facilitâtes sécrétion of an antibody chain from a host cell. An antibody chain gene may be cloned into a vector such that the signal peptide is linked in-frame to the amino terminus of an immunoglobulin chain. The signal peptide can be an immunoglobulin signal peptide or a heterologous signal peptide (i.e., a signal peptide from a nonimmunoglobulin protein).
In addition to antibody chain genes, the recombinant vector expression of the invention can carry regulatory sequences that control the expression of antibody chain genes in a host cell. It will be understood by those skilled in the art that the design of an expression vector, including the sélection of regulatory sequences, may dépend on such factors as the choice of a host cell to be transformed, the level of expression of a desired protein, and so forth. Preferred control sequences for an expression host cell in mammals include viral éléments that ensure high levels of protein expression in mammalian cells, such as promoters and/or enhancers derived from a retroviral LTR, cytomégalovirus (CMV) (such as a CMV promoter/enhancer), simian virus 40 (SV40) (such as a SV40 promoter/enhancer), adenovirus, (e.g., the adenovirus major late promoter (AdMLP)), polyomavirus and strong mammalian promoters such as native immunoglobulin and actin promoters. For further description of viral control éléments and sequences thereof, see, e.g., US patents Nos. 5,168,062, 4,510,245 and 4,968,615. Methods for expressing binding molécules, such as antibodies in plants, including a description of promoters and vectors, as well as transformation of plants is known in the art. See, e.g., U. S. Patent No. 6,517,529. Methods for expressing polypeptides in bacterial cells or fungal cells, e.g., yeast cells, are also well known in the art.
In addition to antibody chain genes and regulatory sequences, the recombinant expression vectors of the invention may carry additional sequences, such as sequences that regulate réplication of a vector in host cells (e.g., origins of réplication) and selectable marker genes. The selectable marker gene facilitâtes the sélection of host cells into which a vector has been introduced (see e.g., U.S. Patent Nos. 4,399,216, 4,634,665 and 5,179,017). For example, typically the selectable marker gene confers résistance to médicinal agents, such as G418, hygromycin or methotrexate, to a host cell into which a vector has been introduced. For example, selectable marker genes include a dihydrofolate reductase (DHFR) gene (for use in dhfr-host cells during methotrexate selection/amplification), a neo gene (for G418 sélection), and a glutamate synthetase gene.
The term expression control sequence as used herein is intended to refer to polynucleotide sequences that are necessary to affect the expression and Processing of coding sequences to which they are ligated. Expression control sequences include appropriate transcription initiation, termination, promoter and enhancer sequences; efficient RNA processing signais such as splicing and polyadenylation signais; sequences that stabilize cytoplasmic mRNA; sequences that enhance translation efficiency (i.e., Kozak consensus sequence); sequences that enhance protein stability; and when desired, sequences that enhance protein sécrétion. The nature of such control sequences differs depending upon the host organism; in prokaryotes, such control sequences generally include the promoter of a ribosomal binding site, and transcription termination sequences; in eukaryotes, typically, such control sequences include promoters and transcription termination sequences. The term control sequences is intended to include at least ail components, the presence of which is essential for expression and processing, and can also include additional components, the presence of which is advantageous, for example, leader sequences and fusion partner sequences.
Host cells and a method for producing a binding molécule
A further aspect of the invention relates to methods for producing binding molécules of the invention. One embodiment of the invention relates to a method for producing binding molécules as defmed herein, comprising introducing a recombinant host cell capable of expressing a binding molécule, cultivating said host cells under conditions suitable for expression of the binding molécule, and isolating the obtained binding molécule. Binding molécules produced by such expression in such recombinant host cells are referred to herein as recombinant binding molécules. Where the binding molécules are antibodies, they are called recombinant antibodies. The invention also relates to the progeny of cells from such host cells and binding molécules obtained analogously.
The term recombinant host cell (or simply host cell) as used herein is intended to refer to a cell into which a recombinant expression vector has been introduced. The présent invention relates to host cells, which may include, for example, a vector according to the invention described above. The présent invention also relates to host cells that comprise, for example, a nucléotide sequence encoding a heavy chain or antigen-binding portions thereof, a light chain-encoding nucléotide sequence or antigen-binding portions thereof, or both, of the first binding domain and/or second binding domain of a binding molécule of the invention. It should be understood that recombinant host cell and host cell are intended to refer not only to a particular subject cell but to the progeny of such a cell as well. Since modifications may occur in succeeding générations due to either mutation or environmental influences, such progeny may not, in fact, be identical to a parental cell, however, such cells are still included within the scope of the term host cell as used herein.
Nucleic acid molécules encoding the binding molécules of the invention and vectors comprising these nucleic acid molécules can be used for transfection of a suitable mammalian, plant, bacterial or yeast host cell. Transformation can be by any known technique for introducing polynucleotides into a host -cell. Methods for introduction of heterologous polynucleotides into mammalian cells are well known in the art and include dextran-mediated transfection, calcium phosphate précipitation, polybrene-mediated transfection, protoplast fusion, encapsulation of the polynucleotide(s) in liposomes, and direct microinjection of
DNA into nuclei. In addition, nucleic acid molécules may be introduced into mammalian cells by viral vectors. Methods for transforming cells are well known in the art. See, e.g., U.S. Pat. Nos. 4,399,216, 4,912,040, 4,740,461 and 4,959,455. Methods for transforming plant cells are well known in the art, including, e.g., Agrobacterium-mediated transformation, biolistic transformation, direct injection, electroporation and viral transformation. Methods of transforming bacterial and yeast cells are also well known in the art.
Mammalian cell Unes available as hosts for expression are well known in the art and include a plurality of immortalized cell lines available from the American Type Culture Collection (ATCC). These include, inter alia, Chinese hamster ovary (CHO) cells, NSO cells, SP2 cells, HEK-293T cells, FreeStyle 293 cells (Invitrogen), NIH-3T3 cells, HeLa cells, baby hamster kidney (BHK) cells, African green monkey kidney cells (COS), human hepatocellular carcinoma cells (e.g., Hep G2), A549 cells, and a number of other cell lines. Cell lines of particular preference are selected through determining which cell lines hâve high expression levels. Other cell lines that may be used are insect cell lines, such as Sf9 or Sf21 cells. When recombinant expression vectors encoding binding molécules are introduced into mammalian host cells, the binding molécules are produced by culturing the host cells for a period of time sufficient to allow for expression of binding molécules in host cells or, more preferably, sécrétion of a binding molécule into the culture medium in which the host cells are grown. Binding molécules can be reconstituted from the culture medium using standard protein purification techniques. Plant host cells include, e.g., Nicotiana, Arabidopsis, duckweed, corn, wheat, potato, etc. Bacterial host cells include K coli and Streptomyces species. Yeast host cells include Schizosaccharomyces pombe, Saccharomyces cerevisiae and Pichia pastoris.
Furthermore, expression of the binding molécules of the invention from production cell lines can be enhanced using a number of known techniques. For example, the glutamine synthetase gene expression system (the GS system) is a common approach for enhancing expression under certain conditions. The GS System is discussed in whole or part in connection with European Patent Nos. 0 216 846, 0 256 055, 0 323 997 and 0 338 841.
It is likely that binding molécules expressed by different cell lines or in transgenic animais will hâve a different glycosylation profile as compared to each other. However, ail binding molécules encoded by the nucleic acid molécules described herein, or comprising the amino acid sequences provided herein are part of the présent invention, regardless of the glycosylation of the binding molécules, and, in general, regardless of the presence or absence of post-translational modifications.
The binding molécules can be produced using a plurality of methods. For example, PDI-binding domains can be prepared separately (e.g., using Chemical protein synthesis, recombinant expression techniques, hybridoma technology, etc.) and then chemically attached to each other, directly or via a linker. Means for Chemical conjugation of molécules (e.g., antibodies or antigen-binding portions thereof) are well known in the art. Polypeptides typically contain a variety of functional groups, such as carboxylic acid (COOH) or free amine (NH2) groups, which are capable of reacting with a suitable functional group of a corresponding polypeptide or linker. An antibody can also be derivatized to expose or attach additional reactive functional groups and may include attachment of any of a number of linker molécules such as those available from Pierce Chemical Company, Rockford, 111. The linkers used in the binding molécules of the invention can be any of suitable linkers known in the art.
In some embodiments, the binding of domains to PDI is produced by the expression of recombinant antibodies or antigen-binding portions thereof in host cells. Sequences encoding any combination of binding domains may be linked (directly or through a linker). The obtained nucleic acid molécules encoding the domains which bind to PD-1 are inserted into expression vectors and introduced into host cells. The obtained binding molécules then are expressed, isolated and purified from the expression System.
In some embodiments, binding domains of a binding molécule may be coupled together by an innovative linker molécule intended for protection against the proteolytic dégradation of a binding molécule. Such a linker is typically devoid of a proteolytic cleavage site.
As used herein, the expressions cell, host cell, cell line, and cell culture, cell line as a producer are used interchangeably and include an individual cell or cell culture that is a récipient of any isolated polynucleotide of the invention or any recombinant vector (any recombinant vectors) that comprise a sequence encoding an HCVR, LCVR or monoclonal antibody of the invention. Host cells include the progeny of a single host cell, and the progeny may not necessarily be completely identical (in morphology or in total DNA complément) to the original parental cell due to natural, accidentai, or deliberate mutation and/or alterations. A host cell includes cells transformed, transduced or infected with a recombinant vector, or a monoclonal antibody that expresses a polynucleotide of the invention or a light or heavy chain thereof. A host cell which comprises a recombinant vector of the invention (either stably incorporated into host chromosomes or not) may also be referred to as a recombinant host cell. Preferred host cells for use in the invention are CHO cells (e.g., ATCC CRL-9096), NSO cells, SP2/0 cells, COS cells (ATCC, e.g., CRL-1650, CRL-1651), andHeLa (ATCC CCL-2). Additional host cells for use in the invention include plant cells, yeast cells, other mammalian cells and prokaryotic cells.
Pharmaceutical composition
The term “pharmaceutical composition” is intended to refer to a formulation and/or composition containing a therapeutically effective amount of an antibody of the invention plus excipients (diluents, vehicles, solvents and other excipients).
The term excipient is used herein to describe any ingrédient other than the compound(s) of the invention. The choice of an excipient dépends largely on factors such as the particular technique of administration, the effect of an excipient on solubility and stability, and the nature of a dosage form. As used herein, pharmaceutically acceptable excipient includes any and ail solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonie and absorption delaying agents and similar physiologically compatible substances. Examples of said pharmaceutically acceptable excipients are water, physiologie saline, phosphate buffer, dextrose, glycerol, éthanol, etc., and combinations thereof. It is often préférable to add to the composition isotonie agents, e.g., sugars, polyalcohols, such as mannitol or sorbitol, or sodium chloride. The further examples of pharmaceutically acceptable excipients are wetting agents or a small amount of auxiliary substances, such as moisturizers and emulsifiers, preservatives or buffers that will increase the storage duration and efficiency of an antibody.
An antibody of the invention can be incorporated into a pharmaceutical composition suitable for administration to a patient (see Example 17). The antibodies of the invention may be administered alone or in combination with a pharmaceutically acceptable carrier, diluent, and/or excipient, in single or multiple doses. Pharmaceutical compositions for administration are designed to be appropriate for the selected mode of administration, and pharmaceutically acceptable diluents, carriers, and/or excipients, such as dispersing agents, buffers, surfactants, preservatives, solubilizing agents, isotonicity agents, stabilizing agents and the like be used as appropriate (see Example 17). Said compositions are designed in accordance with conventional techniques as in e.g., Remington, The Science and Practice of Pharmacy, 19th Edition, Gennaro, Ed., Mack Publishing Co., Easton, PA 1995, which provides various techniques for obtaining the compositions as are generally known to a skilled person.
A composition comprising an anti-PD-1 monoclonal antibody of the invention may be administered to a patient exhibiting the risk of adverse development or pathologies as described herein using standard administration techniques, including pérorai, intravenous, intraperitoneal, subeutaneous, pulmonary, transdermal, intramuscular, intranasal, buccal, sublingual, or suppository administration.
A pharmaceutical composition of the invention preferably comprises or is a therapeutically effective amount of an antibody of the invention. The term therapeutically effective amount is intended to refer to an amount that is effective, at dosages and for periods of time necessary, to achieve the desired therapeutic resuit. A therapeutically effective amount of an antibody may vary according to factors such as disease State, âge, sex and weight of a subject, and the ability of an antibody or part thereof to elicit a desired response in a subject. A therapeutically effective amount is also one in which any toxic or detrimental effects of the antibody are outweighed by the therapeutically bénéficiai effects. Prophylactically effective amount is intended to refer to the amount that is effective, at dosages and for periods of time necessary to achieve the desired prophylactic resuit. Since a prophylactic dose is prescribed for individuals before or at an early stage of disease, typically a prophylactically effective amount may be less than a therapeutically effective amount.
A therapeutically effective or prophylactically effective amount is at least a minimal therapeutically bénéficiai dose that is less than the toxic dose of an active agent. On the other hand, a therapeutically effective amount of an antibody of the invention is an amount that reduces the biological activity of PD-1 in mammals, preferably humans.
The route of administration of an antibody of the invention can be oral, parentéral, inhalation or local. Preferably antibodies of the invention can be involved in a pharmaceutical composition acceptable for parentéral administration. The term parentéral as used herein includes intravenous, intramuscular, subcutaneous, rectal, vaginal or intraperitoneal administration. Intravenous, intraperitoneal or subcutaneous injections are preferred routes of administration. Acceptable pharmaceutical carriers for such injections are well known from the prior art.
As described in appropriate guidelines, pharmaceutical compositions shall be stérile and stable under the conditions of production and storage in a container, which is provided by, for example, hermetically sealed vials (ampoules) or syringes. Thus, pharmaceutical compositions can be subjected to filtration sterilization after preparing the composition, or can be made microbiologically suitable by any other technique. A typical composition for an intravenous infusion can include 250-1000 ml of fluid such as stérile Ringer's solution, physiologie saline, dextrose solution or Hank's sait solution, and a therapeutically effective dose (for example, 1-100 mg/ml or more) of an antibody concentrate. Doses may vary depending on disease type and severity. It is well known from the State of the medical art that doses for a patient dépends on multiple factors including patient's sizes, body surface area, âge, spécifie compound to be administered, gender, duration and route of administration, general health State and other simultaneously administered médications. A typical dose can be, for example, in a range of 0.001-1000 pg; however, doses lower and higher than this illustrative range are anticipated, especially given the above-mentioned parameters. The daily parentéral dosing regimen may be from 0.1 pg/kg to 100 pg/kg of overall body weight, preferably from 0.3 pg/kg to 10 pg/kg, and more preferably from 1 pg/kg to 1 pg/kg, even more preferably from 0.5 to 10 pg/kg of body weight per day. The treatment process can be monitored by periodical assessment of patient's health State. For repeated administration for several days or longer, depending on patient's condition, the treatment is repeated until the desired response or suppression of symptoms of a disease. However, another dosing regimens not described herein can also be applied. The desired dose may be administered by single bolus or multiple bolus dosing, or by means of a continuous infusion of an antibody depending on a pharmacokinetic breakdown desired by a practitioner.
Said assumed properties of an antibody largely dépend on a physician's decision. The intended effect is the key factor for choosing a proper dose and regimen. Factors considered herein include a certain disease to be treated, a certain mammal to receive the treatment, clinical condition of a certain patient, disorder cause, antibody administration site, spécifie antibody type, route of administration, administration regimen and other factors well known in the medical arts.
Therapeutic agents of the invention can be frozen or lyophilized and reconstituted in an appropriate stérile carrier prior to administration. Freezedrying and reconstitution can resuit in some loss of antibody's activity. Doses can be adjusted to compensate this loss. In general, pharmaceutical composition pH values from 6 to 8 are préférable.
Therapeutic use of a binding molécule of the invention
In one aspect, a binding molécule of the invention is useful in the treatment of disorders that are associated with PD 1 activity.
Treat, treating and treatment refer to a method of alleviating or abrogating a biological disorder and/or at least one of attendant symptoms thereof. As used herein, to alleviate a disease, disorder or condition means reducing the severity and/or occurrence frequency of the symptoms of a disease, disorder, or condition. Further, references herein to treatment include references to curative, palliative and prophylactic treatment.
In one aspect, the subject is a mammal, preferably a human subject. Said subject may be either male or female, of any âge.
Therapeutically effective amount is intended to refer to that amount of the therapeutic agent being administered which will relieve to some extent one or more of the symptoms of the disorder being treated.
The binding molécules of the invention may be administered alone or in combination with one or more other préparations or antibodies (or any combination thereof). Thus, the pharmaceutical compositions, methods and uses of the invention also encompass embodiments of combinations (coadministration) with other active agents, as described below.
As used herein, the ternis co-administration, co-administered and in combination with, referring to binding molécules and one or more other therapeutic agents, are expected to mean, refer to or include the following:
simultaneous administration of such combination of a binding molécule of the invention and a therapeutic agent to a patient in need of treatment, when such components are formulated together into a single dosage form which releases said components at substantially the same time to said patient, substantially simultaneous administration of such combination of a binding molécule of the invention and a therapeutic agent to a patient in need of treatment, when such components are formulated apart from each other into separate dosage forms which are taken at substantially the same time by said patient, whereupon said components are released at substantially the same time to said patient, sequential administration of such combination of a binding molécule of the invention and a therapeutic agent to a patient in need of treatment, when such components are formulated apart from each other into separate dosage forms which are taken at consecutive times by said patient with a significant time interval between each administration, whereupon said components are released at substantially different times to said patient; and sequential administration of such combination of a binding molécule of the invention and a therapeutic agent to a patient in need of treatment, when such components are formulated together into a single dosage form which releases said components in a controlled manner, whereupon they are concurrently, consecutively, or jointly released at the same and/or different times to said patient, where each portion may be administered by either the same or different routes.
The binding molécules of the invention can be administered without further therapeutic treatment, i.e., as an independent therapy. Furthermore, treatment by the binding molécules of the invention may comprise at least one additional therapeutic treatment (combination therapy). In some embodiments, the binding molécule may be administered jointly or formulated with another medication/preparation for the treatment of an autoimmune or inflammatory disease.
Pharmaceutical préparations comprising a binding molécule of the présent invention and at least one other agent (e.g., an immunosuppressive or antiinflammatory agent) may be used as a combination therapy for simultaneous, independent or consecutive administration in the treatment of inflammatory and autoimmune disorders.
It is meant that the binding molécules of the invention may be used in the methods of treatment as described above, may be used in the treatment as described above, and/or may be used in the manufacture of a médication for treatment as described above.
Doses and routes of administration
Any method for administering peptides, proteins or antibodies accepted in the art may be suitably employed for a binding molécule of the invention.
The pharmaceutical compositions of the invention are typically suitable for parentéral administration. As used herein, parentéral administration of a pharmaceutical composition includes any route of administration characterized by physical breaching of the tissue of a subject and administration of a pharmaceutical composition through the breach in the tissue, thus generally resulting in the direct administration into the blood stream, into the muscle, or into an internai organ. Thus, parentéral administration includes, inter alia, administration of a pharmaceutical composition by virtue of injection of the composition, by administration of the composition through a surgical incision, by application of the composition through a tissue-penetrating non-surgical wound, and the like. In particular, parentéral administration is meant to include, inter alia, subcutaneous, intraperitoneal, intramuscular, intrastemal, intravenous, intraarterial, intrathecal, intraventricular, intraurethral, intracranial, intrasynovial injection or infusions; and kidney dialytic infusion techniques. Intra-tumor delivery, for example, intra-tumor injection, can also be useful. Régional perfusion is also provided. Preferred embodiments include intravenous and subcutaneous routes.
Dosage forms of pharmaceutical compositions suitable for parentéral administration typically comprise an active ingrédient combined with a pharmaceutically acceptable carrier, such as stérile water or stérile isotonie saline. Such dosage forms may be prepared, packaged, or sold in a form suitable for bolus administration or for continuous administration. Injectable formulations may be prepared, packaged, or sold in a common dosage form, e.g., in ampoules or in multi-dose containers containing a preservative. Formulations for parentéral administration include, inter alia, suspensions, solutions, émulsions in oily or aqueous bases, pastes, and the like. Such dosage forms may further comprise one or more additional ingrédients including, inter alia, suspending, stabilizing, or dispersing agents. In one embodiment, a composition for parentéral administration comprises an active ingrédient which is provided in dry (i.e. powder or granular) form for reconstitution with a suitable base (e.g., stérile pyrogen-free water) prior to parentéral administration of the reconstituted composition. Parentéral dosage forms also include aqueous solutions which may comprise excipients such as salts, carbohydrates and buffering agents (preferably to pH 3 to 9), but, for some applications, they may be more suitably formulated as a stérile non-aqueous solution or as a dried form to be used in conjunction with a suitable base such as stérile pyrogen-free water. Exemplary forms for parentéral administration include solutions or suspensions in stérile aqueous solutions, for example, aqueous propylene glycol or dextrose solutions. Such dosage forms can be buffered, if necessary. Other suitable dosage forms for parentéral administration may include those which comprise an active ingrédient in a microcrystalline form, or in a liposomal préparation. Dosage forms for parentéral administration may be formulated to hâve an immédiate and/or modified release. Modified release dosage forms include a delayed, sustained, pulsed, controlled, targeted and programmed release.
For example, in one aspect, stérile injectable solutions can be prepared by incorporating at least one binding molécule in the required amount in an appropriate solvent with one ingrédient or a combination of ingrédients enumerated above, as required, followed by filtered sterilization. Dispersions are typically prepared by incorporating the active compound into a stérile solvent that contains a basic dispersion medium and the other required ingrédients from those enumerated above. In the case of stérile powders for the préparation of stérile injectable solutions, the methods for préparation are freeze-drying (lyophilization) that yields the powder of an active ingrédient plus any additional desired ingrédient from a previously sterile-filtered solution thereof. The proper fluidity of a solution can be maintained, for example, by the use of a coating such as lecithin, by the maintenance of a required particle size in the case of dispersions and by the use of surfactants. Prolonged absorption of injectable compositions can be brought about by virtue of incorporating in the composition an agent that delays absorption, for example, monostearates and gelatin, and/or by virtue of modified release coatings (e.g., slow release coatings).
The binding molécules of the invention can also be administered intranasally or by inhalation, typically in the form of a dry powder (either alone, as a mixture, or as particles comprising mixed components, for example, mixed with a suitable pharmaceutically acceptable excipient) from a dry powder inhaler, such as a pressurized aérosol container, pump, spray, atomizer (preferably an atomizer using electrohydrodynamics to produce fine mist), or nebulizer, wherein a suitable propellant is used or not used, or as nasal drops.
A pressurized container, pump, spray, atomizer, or nebulizer typically contains a solution or suspension of a binding molécule of the invention comprising, for example, a suitable agent for dispersing, reconstituting, or extending release of the active substance, a propellant as solvent.
Prior to use as a dry powder or suspension, the médicinal préparation is typically micronized to a size suitable for delivery by virtue of inhalation (typically less than 5 microns). This may be achieved by virtue of any suitable comminuting technique, such as spiral jet milling, fluid bed jet milling, supercritical fluid processing to form nanoparticles, high-pressure homogenization, or spray drying.
Capsules, blisters and cartridges for use in an inhaler or insufflator may be formulated to contain a powder mix of a compound of the invention, a suitable powder base and performance modifier.
A suitable solution formulation for use in an atomizer using electrohydrodynamics to produce a fine mist may contain a suitable dose of a binding molécule of the invention per actuation and the actuation volume may vary from, e.g., 1 μΐ to 100 μΐ.
Suitable flavours, such as menthol and levomenthol, or sweeteners, such as saccharin or saccharin sodium, may be added to those dosage forms of the invention intended for inhalation/intranasal administration.
Dosage forms for parentéral administration may be formulated for an immédiate and/or modified release. Modified release dosage forms include a delayed, sustained, pulsed, controlled, targeted and programmed release.
In the case of dry powder inhalers and aérosols, the dosage unit is determined by means of a valve which delivers a metered amount. Units in accordance with the invention are typically arranged to administer a metered dose or pufC of a binding molécule of the invention. The overall daily dose will typically be administered in a single dose or, more frequently, as divided doses throughout the day.
The binding molécules of the invention may also be formulated as dosage forms for pérorai administration. Pérorai administration may include swallowing, so that the compound enters the gastrointestinal tract, and/or buccal, lingual, or sublingual administration by which the compound enters the blood stream directly from the mouth.
Dosage forms suitable for pérorai administration include solid, semi-solid and liquid Systems such as tablets; soft or hard capsules comprising multi- or nano-particulates, liquids, or powders; lozenges (including liquid-filled); chews; gels; fast dispersing dosage forms; films; ovules; sprays; and buccal/mucoadhesive patches.
Liquid dosage forms include suspensions, solutions, syrups and élixirs. Such dosage forms may be employed as excipients in soft or hard capsules (made, for example, of gelatin or hydroxypropylmethylcellulose) and typically comprise a carrier, for example, water, éthanol, polyethylene glycol, propylene glycol, methylcellulose, or a suitable oil, and one or more emulsifying agents and/or suspending agents. Liquid dosage forms may also be prepared by reconstituting a solid substance, for example, from a sachet.
The binding molécules of the invention will be administered in an amount that is effective in treatment of the condition in question, i.e. in doses and during the periods of time required to achieve the desired resuit. A therapeutically effective amount may vary according to factors such as the spécifie condition to be treated, âge, sex, and weight of a patient, and whether the binding molécules are administered alone or in combination with one or more additional antiautoimmune or anti-inflammatory treatment techniques.
Dosage regimens may be adjusted to provide the optimum response. For example, a single bolus may be administered, several divided doses may be administered over time or the dose may be proportionally reduced or increased as indicated by the exigencies of the therapeutic situation. It is especially advantageous to formulate parentéral compositions in a unit dosage form for ease of administration and uniformity of dosage. A unit dosage form as used herein is intended to refer to physically discrète units suited as unitary dosages for patients/subjects to be treated; each unit contains a predetermined quantity of active compound calculated to produce the desired therapeutic effect in association with the desired pharmaceutical carrier. Spécification for the unit dosage forms of the invention is typically dictated by and directly dépendent on (a) the unique characteristics of a chemotherapeutic agent and particular therapeutic or prophylactic effect to be achieved, and (b) the limitations inhérent in the art of compounding such an active compound for the treatment of sensitivity in the subjects.
Thus, a skilled artisan would appreciate, based upon the disclosure provided herein, that the doses and dosage regimen are adjusted in accordance with methods well known in the therapeutic arts. That is, the maximum tolerable dose can be readily established, and the effective amount providing a détectable therapeutic effect to a patient may also be determined, as can the temporal requirements for administering each agent to provide a détectable therapeutic effect to a patient. Thus, while certain dose and administration regimens are exemplified herein, these examples in no way limit the doses and administration regimen that may be provided to a patient in practicing the embodiments of the invention.
It is to be noted that dosage values may vary with the type and severity of the condition to be alleviated and may include single or multiple doses. Furthermore, it is to be understood that for any particular subject, spécifie dosage regimens should be adjusted over time according to the individual need and the judgment of a medical professional administering or supervising the administration of the compositions, and that dosage ranges set forth herein are exemplary only and are not intended to limit the scope or practice of the claimed compositions. Furthermore, the dosage regimen with the compositions of the présent invention can be based on various factors, including the type of a disease, âge, weight, gender, patient's health condition, severity of a condition, route of administration and a particular binding molécule used. Thus, the dosage regimen may widely vary, but can be determined regularly using standard techniques. For example, doses may be adjusted based on pharmacokinetic or pharmacodynamie parameters, which may include clinical effects such as toxic effects and/or laboratory values. Thus, the présent invention encompasses individual dose escalation as determined by a skilled artisan. Methods for determining appropriate dosages and regimens are well known in the art and would be understood by a skilled artisan once provided the ideas disclosed herein.
Examples of suitable administration methods are provided above.
It is believed that a suitable dose of a binding molécule of the invention will be in the range of 0.1-100 mg/kg, including about 0.5-50 mg/kg, for example about 1-20 mg/kg. A binding molécule may be administered, e.g., in a dose of at least 0.25 mg/kg, such as at least 0.5 mg/kg, including at least 1 mg/kg, e.g., at least 1, 5 mg/kg, such as at least 2 mg/kg, e.g., at least 3 mg/kg, including at least 4 mg/kg, e.g., at least 5 mg/kg; and for example up to a maximum of 50 mg/kg, including up to a maximum of 30 mg/kg, e.g., up to a maximum of 20 mg/kg, including up to a maximum of 15 mg/kg. The administration will typically be repeated in appropriate time intervals, such as once a week, once every two weeks, once every three weeks or once every four weeks, and for as long as deemed appropriate by a responsible physician, who may, in some cases, increase or reduce the dose if necessary.
Effective amount for the treatment of autoimmune or inflammatory disorders can be measured by the ability thereof to stabilize the progression of disease and/or to improve the symptoms in a patient, and preferably to reverse disease manifestations. The ability of a binding molécule of the invention to suppress autoimmune or inflammatory disorders can be evaluated in in vitro assays, for example as described in the given examples, as well as in suitable animal models that predict efficacy in such disorders. A suitable dosage regimen will be selected to provide an optimal therapeutic response in a certain situation, e.g., single bolus administration or continuous infusion with the possible adjustment of a dose as indicated by the exigencies of each case.
Diagnostic use and compositions
The binding molécules of the invention are also used in diagnostic processes (e.g., in vitro, ex vivo). For example, they can be used for detecting or measuring the level of PD-1 in samples obtained from a patient (e.g., tissue sample or a sample of body fluid, such as an inflammatory exudate, blood, sérum, intestinal fluid, saliva or urine). Suitable methods for détection and measurement include immunoassays, such as flow cytometry, enzyme-linked immunosorbent assay (ELISA), chemiluminescent assay, radioimmunoassay, and immunohistology.
Articles of manufacture
In another embodiment, an article of manufacture is provided containing materials useful for the treatment or prévention of disorders and conditions described above.
The article of manufacture comprises a container with an antibodycontaining pharmaceutical composition with a label, and possibly a package insert. Suitable containers include, e.g., vials, ampoules, syringes and analytical tubes. The containers may be made of a plurality of materials such as glass or polymer material. The container comprises a composition of the invention which is effective for treating a PD-1-mediated disease or disorder and can hâve a stérile access port (e.g., the container may be an intravenous solution bag or a vial having a stopper pierceable by a hypodermic injection needle). An anti-PD1-antibody of the invention is an active agent of the composition. A label located on the container or a package insert attached thereto indicates that the composition is used for treating the desired disease. The article of manufacture may further comprise a second container with a pharmaceutically acceptable buffer such as phosphate-buffered saline, Ringer's solution and dextrose solution. It can further include other materials désirable from a commercial and consumer standpoint, including other buffers, diluents, filters, needles, syringes and package inserts.
The following examples are provided to better understand the invention. These examples are presented for illustrative purposes only and are not intended to limit the scope of the présent invention in any way.
Examples
Example 1
Engineering of a naive human antibody Fab-library MeganLibTM
Total RNA of B lymphocytes from blood samples of more than one thousand individual human donors was isolated using RNeasy Mini Kit according to the suggested protocol (QIAGEN). An RNA concentration assay was performed using Nanovue kit (GE Healthcare); the quality of isolated RNA was tested by means of 1.5% agarose gel electrophoresis.
A reverse transcription reaction was conducted using MMLV RT kit (Evrogen) according to the recommended protocol with MMuLV reverse transcriptase and random hexamer oligonucleotides as primers.
Reverse transcription products were used as a matrix in a two-stage polymerase chain reaction to obtain the genes of variable domains flanked with restriction sites; the reaction was performed using oligonucleotide kit and protocols by [J Biol Chem. 1999 Jun 25; 274(26): 18218-30].
The obtained DNA préparation VL-CK-VH (Fig. 1) was treated with NheI/Eco91I restriction endonucleases and ligated into original phagemid pH5 (Fig. 2A). Ligation products were transformed into SS320 electrocompetent cells prepared in accordance with protocols [Methods Enzymol. 2000;328: 33363.]. The répertoire of combinatorial phage Fab-display library MeganLibTM was 1011 transformants. The préparations of phage-Fab-libraries were prepared in accordance with the earlier described procedure [J Mol Biol. 1991 Dec 5;222(3): 581-97].
Example 2
Sélection of fab-libraries of phage antibodies
Spécifie anti-PD-1 phage Fab-antibodies were isolated from a combinatorial library phage Fab-display library of MeganLibTM. Sélection was performed using a recombinant human PD-1, with panning under conditions as described earlier (J Biol Chem. 1999 Jun 25; 274(26): 18218-30; Nat Biotechnol. 1996 Mar;14(3):309-14; J Mol Biol. 1991 Dec 5;222(3): 581-97). To perform the sélection process by the panning method, human PD-1 in 50 mM carbonate buffer (pH 9.5) was adsorbed ovemight at 4°C on the surface of HighSorb tubes (Nunc). Further, tubes were washed with PBS (pH 7.4) and then blocked with a solution containing PBS (pH 7.4) - fat-free milk (0.5% weight/volume) for 1 hour. Then, 2-4 ml of phage solution (1013 phage particles per ml) in PBS (pH 7.4) - fat free milk (0.5% w/vol) were transferred to the tube with the antigen, and the system was incubated for 1 hour under stirring. Unbound phages were removed by a sériés of washing cycles with PBS (pH 7.4) - Tween 20 (0.1% vol./vol.). The number of washing cycles was increased from the first round to the third one - 20-30-40 times, respectively. Phage particles that remained bound were eluted with 100 mM Gly-HCl solution (pH 2.5) during 15 min under stirring, and then neutralized with 1 M TRIS-HC1 (pH 7.6). E. coli TGI bacteria were infected with phages obtained; further, phages were isolated and used in the next sélection cycle. After the second and third round of sélection, DNA (phagemids) were isolated and genes of antibody variable domains were cloned to expression vectors (Fig. 2B) for production of Fab in E.coli. cells.
Example 3
Analysis of Fab spécifie binding to human PD-1
ELIS A was used to measure the binding of test Fab-fragments to human PD-1. Fab with a published Nivolumab sequence (Bristol-Myers Squibb) was used as a positive control. In order to analyze the spécifie binding, ELISA plate wells (medium binding from Greiner bio one) were coated with 50 μΐ (0.5 pg/ml in lx coating carbonate buffer, pH 9.5) PD1-H6F, hermetically closed and incubated ovemight at 4°C. Ail further stages were conducted in accordance with the standard ELISA protocol with a high-performance automated platform based on robotic Systems such as Genetix Qpix2xt (Molecular Devices) and
Tecan Freedom EVO 200 (Tecan). Non-specific binding was blocked by adding a blocking buffer BB (200 μΐ 0.5% fat-free milk in PBS). Plates were incubated on a shaker for 1 h at room température. After washing with PBS-Tween, each cell was coated with 50 μΐ of test Fab-containing cell supernatant mixed with the equal volume of BB. Plates were incubated on a shaker for 1 hour at room température; further, each plate well was 3 times washed with PBS-Tween buffer. After washing, each well was coated (50 μΐ/well) with anti-human Fab HRP-conjugated secondary antibody (Pierce-ThermoScientific) in PBS-Tween (1:5000). Plates were transferred to rotation shaker (50 min at room température) and then 3 times washed with PBS-Tween buffer as described above. Colorimétrie signal was obtained by adding TMB (50 μΐ/well) until saturated (average of 3-5 min); further color development was blocked by adding the stop solution (30 μΐ/well, 10% sulfuric acid). Absorbance was measured at 450 nm using an appropriate Tecan-Sunrise plate reader (Tecan). Antibody binding was proportional to the signal produced.
Example 4
Compétitive ELISA of blocking the interaction of PDL1 ligand and PD-1 receptor
A compétitive ELISA technique was used to test the antagonistic capacity of previously selected anti-PD-1 spécifie Fab. Fab with published Nivolumab sequence (Bristol-Myers Squibb) was used as a positive antagonist control. ELISA well plates (medium binding, Greiner bio one) were covered with 50 μΐ/well PD1-H6F receptor (1 pg/ml solution in IX coating carbonate buffer pH 9.5) and incubated ovemight at 4°C. Ail further stages were performed in accordance with standard ELISA protocols with a highperformance automated platform based on robotic Systems such as Genetix Qpix2xt (Molecular Devices) and Tecan Freedom EVO 200 (Tecan). Nonspecific binding was blocked by adding a blocking buffer BB (200 μΐ 0.5% fat free milk in PBS). Plates were incubated on a shaker for 1 h at room température.
After the plate containing PD 1 receptor was washed of BB solution, it was coated with the test Fab-containing cell supematant, incubated under 500 rpm shaking for 45 min at room température. Then it was mixed with 50 μΐ PDLl-Fc at a final concentration of 1 pg/ml, and incubated for 45 min under the same conditions, further, each plate well was washed 3 times with PBS-Tween buffer. Further, 50 μΐ /well of goat anti-human IgG (Fc) HRP-conjugated secondary antibody (Sigma) were added in PBS-Tween (1:5000). Plates were incubated on rotation shaker for 45 min at room température and 5 times washed with PBS-Tween, as mentioned above. A colorimétrie signal was obtained by adding TMB (50 μΐ/well) until saturated (average of 3-5 min); further color development was blocked by adding the stop solution (30 μΐ/well, 10% sulfuric acid). Absorbance was measured at 450 nm using an appropriate Tecan-Sunrise plate reader (Tecan). Fab binding was inversely proportional to the colour signal produced.
Example 5
Comparative kofi-screening for anti-PDl Fab human candidates koff screening was performed using Pall Forte Bio Octet Red 96. AntiFABCH1 biosensors were rehydrated for 30 min in a working buffer comprising 10 mM PBS (pH 7.2-7.4), 0.1% Tween-20 and 0.1% BSA. lOx working buffer was added to test samples of E.coli supernatants up to Ix final concentration. Anti-FABCHl biosensors were then steeped into E.coli supernatants containing Fab-fragments of candidate antibodies and incubated for 12 hours at a température of 4°C. Sensors were then transferred to wells with an analyte solution (PD-1, 30 pg/ml) to achieve antigen-antibody association (300 sec). After that, sensors were returned into wells with working buffer for further dissociation (300 sec). Used sensors were subject to régénération after each test: they were placed three times into regenerating buffer (Gly-HCl, pH 1.7) and then could be used in further experiments. The curves obtained were analyzed using Octet Data Analysis (version 7.0) according to the standard procedure with 1:1 interaction model.
Example 6
Producing recombinant antigens and antibodies in suspension mammal cell culture
Antibodies and antigens were generated in an established cell line obtained from Chinese hamster ovary cells (CHO-K1) according to published protocols [Biotechnol Bioeng. 2005 Sep 20; 91(6):670-677, Liao Métal., 2004; Biotechnol Lett. 2006 Jun;28(l l):843-848; Biotechnol Bioeng. 2003 Nov 5;84(3):332-342]. Cells constitutively expressing the gene of EBNA1 protein (Epstein-Barrvirus nuclear antigen 1) were used. A suspension culture was conducted in flasks on orbital shaker using serum-free media from Life Technologies Corporation and in accordance with manufacturer’s guidelines. For transient expression, cells in a concentration of 2*106/ml were transfected by means of linear polyethyleneimine (PEI MAX, Polysciences). DNA/PEI ratio was 1:3-1:10. In 5-7 days after transfection, cell culture was centrifuged under 2000 g for 20 min and filtered through a 0.22 pm filter. Target proteins were isolated from culture liquid by affine HPLC.
A recombinant PD-1 protein containing 6 His amino acids in C-terminal région was isolated and purified from culture liquid on Profmity IMAC Nicharged resin (Bio-Rad). Prior to purification procedures, NiCl2 was added to culture liquid to a concentration of ImM. Then 5 ml of Profmity IMAC Nicharged was added to culture liquid and mixed on a shaker for 1 h at room température. Sorbent was transferred to 5 ml Thermo scientific Polypropylene columns and washed with 5 column volumes of PBS to remove non-specifically bound components. Bound antigen was eluted with 0.3 M imidazole (pH 8) and 150mM NaCl. Then the protein was dialyzed into PBS (pH 7.4) by means of SnakeSkin Dialysis Tubing technique, filtered (0.22 μm), transferred into tubes and stored at -70°C.
Recombinant PDI-Fc and PDLl-Fc proteins were isolated and purified from cell culture on a Protein A column for affine HPLC. Cleared culture liquid was passed through 5 ml HiTrap rProtein A Sepharose FF column (GE Healthcare) equilibrated with phosphate buffered saline (PBS, pH 7.4). Then the column was washed with 5 volumes of PBS to remove non-specific bound components. Bound antigen was eluted with 0.1 M glycine buffer (pH 8). The principal protein elution peak was collected and brought to neutral pH with 1 M Tris-buffer (pH 8). Ail stages were conducted under 110 cm/h flow rate. Then the protein was dialyzed into PBS (pH 7.4) by means of SnakeSkin Dialysis Tubing technique, filtered (0.22 μm), transferred into tubes and stored at -70°C.
IgGl antibodies were purified on a 1 ml Hi Trap rProteinA FF column (GE Healthcare) in accordance with the aforementioned procedure for PDI-Fc antigen. Purity of the obtained protein was evaluated by SDS-PAGE (Fig. 3A and 3B).
Example 7
Reactivating NFAT-signaling by anti-PDI antibodies in JurkatNFAT-PD1 reporter cell line
Engineering of a human T-cell line of Jurkat origin was conducted by introducing two genetic constructs into the genome thereof. One construct encodes a human PDI receptor gene. The second construct encodes luc2P luciferase gene under control of NFAT-sensitive genetic element. The resuit was a reporter cell line Jurkat-PD1-NFAT-Luc2, which expresses PD1 receptor on the surface membrane and contains NFAT-dependent promoter that directs transcription of luc2P luciferase gene. Synthesis of the luciferase enzyme in cells of this line is proportional to the level of NFAT activity, which, in tum, reflects the overall level of cell activation.
Interaction of PDL1 with PDI inhibits signaling from TCR receptors to NFAT-promoter. When anti-PDI Antibodies uncouple PDL1-PD1 interaction, réactivation of intracellular signaling occurs.
A plate for the experiment was prepared one day prior to analysis as follows: solution of anti-CD3 antibodies in PBS (1 pg/ml with 5 pg/ml PDL1Fc) was introduced to 96-well opaque plate made of opaque plastic. Solution of anti-CD3 antibodies without PDL1 was used for a positive control of activation. Then, it was left overnight at +4°C.
Test antibodies were then diluted from 11 pg/ml in cell growth medium, using three-fold dilution, to fit ten-point curve, cells with antibody dilutions were incubated for 20 minutes at room température, then cells with antibodies were introduced to a prepared plate. A solution of anti-CD28 antibodies in growth medium was added to ail wells to a final concentration of 0.25 pg/ml. Then it was left in a CO2 incubator for 6 hours.
A luciferase substrate from Bio-Glo Luciferase assay System (Promega) pre-prepared according to protocol was thawed from -70°C and added at a rate of V cells/V substrate. Luminescence was measured using Fluoroscan Ascent (Fig. 5). The anti-PDl antibodies reactivate luminescence level in Jurkat-PDlNFAT reporter line, and therefore bind to the receptor and inhibit PDL1/PD1 interaction. BCD-100 showed the best results, the effective dose (ED50) was 114.6 ng/pL.
Example 8
Stimulation of production of IL-2 by anti-PDl antibodies in human whole blood in the presence of staphylococcal enterotoxin
Superantigens, such as SEB cytotoxin (staphylococcal enterotoxin), activate T cells by virtue of binding a class II MHC molécule on antigenpresenting cells to TCR receptor νβ element, resulting in production of cytokines, including IL2 autocrine growth factor.
The test is based on the description of an article (MK-3475; Anti-PD-1 Monoclonal Antibody) in Patients With Advanced Solid Tumors (Clin Cancer Res Published OnlineFirst May 14, 2015).
Briefly, whole blood was incubated with SEB or with SEB plus anti-PDl antibodies, then IL-2 concentration was measured.
Heparinized whole blood from donor was diluted 1:5 times in growth medium (RPMI with 10% fêtai bovine sérum). SEB was added to diluted blood to a final concentration of 1 pg/ml. Anti-PDl antibodies were diluted in growth medium from 50 pg/ml with an incrément of 3, totaling eight dilutions. A solution of blood and SEB in growth medium (volume/volume) was added to the diluted antibodies. Plates were incubated for 3 days in a CO2 incubator at 37°C. Further, IL-2 concentration was measured in selected samples by ELISA technique (Fig. 6), according to a commercially available kit (Human IL-2 Quantikine ELISA Kit from R & D Systems) protocol. BCD-100 showed the best results due to greater functional activity than control antibodies.
Example 9
Analysis of antibody-dependent cell-mediated cytotoxicity (ADCC) of anti-PDl antibodies on Jurkat-PDl cell line
Grown Jurkat-PDl cell culture intended for use in a quantitative test was collected from vials and centrifuged for 5 minutes at 200 g. Supernatant was drained and washed once again in a medium for a quantitative test.
The cell pellet was suspended in 5 ml of medium for the quantitative test; viability and number of cells were determined. Cell suspension was prepared for seeding white 96-well culture plates at 3xl05cells/ml.
Dilutions of the test sample were added to the wells of a 96-well plate. Jurkat-PDl cell suspension was added to wells comprising test samples, and the plate was incubated for 15-30 minutes in a CO2 incubator.
PBMCs cell suspension at a concentration of 7.5 * 106 cells/ml was prepared and added to wells comprising test samples. The plate was incubated at 37°C in a CO2 incubator for 4 hours.
Assay Buffer and AAF-Glo™Substrate from CytoTox-Glotm Cytotoxicity Assay kit were mixed and added to each well comprising test samples. A plate was incubated for 15 minutes at room température. Luminescence was measured using Fluoroscan Ascent FL (Fig. 7). A principle of the method is based on determining the activity of intracellular proteases, the resulting luminescence signal is proportional to the number of lysed cells. AntiPD1 antibodies do not hâve détectable antibody-dependent cellmediated cytotoxicity.
Example 10
Immunoenzymatic analysis of interaction of anti-PDl antibodies with PDI and other antigens
ELIS A was used to measure the relative affinity of antibodies to PDI and other antigens. ELIS A plate wells (medium binding from Greiner bio one) were used to measure the binding. ELIS A plate wells were coated with 50 μΐ of PDl-Fc, IL6R-Fc, CD38-Fc, HER3-Fc, IGFR-Fc, CD3-Fc, IL23-Fc (1 pg/ml for PDI and 5 pg/ml for the other antigens in lx coating carbonate buffer), hermetically closed and incubated overnight at 4°C. Ail further stages were conducted in accordance with the standard ELISA protocol. Non-specific binding was blocked by adding a blocking buffer BB (200 μΐ 0.5% fat-free milk in PBS). Plates were incubated on a shaker for 1 h at room température. After washing with PBS-Tween, 50 μΐ per well of test antibodies were added at a concentration of 5 pg/ml in PBS-Tween. Plates were once again incubated, while shaken, for one hour at room température, thereafter each plate well was washed three times with PBS-Twin buffer. After washing, each well was coated (50 μΙ/well) with anti-human Fab HRP-conjugated secondary antibody (PierceThermoScientific) in PBS-Tween (1:5000). Plates were transferred to a rotation shaker (50 min at room température) and then washed 3 times with PBS-Tween buffer as described above. A colorimétrie signal was obtained by adding TMB (50 μΙ/well) until saturated (average of 3-5 min); further color development was blocked by adding the stop solution (30 μΙ/well, 10% sulfuric acid). Absorbance was measured at 450 nm using an appropriate Tecan-Sunrise plate reader (Tecan). Antibody binding was proportional to the signal produced (Fig. 4). Anti-PD 1 antibody specifically binds to PD 1 and does not bind to other antigens in question.
Example 11
Immunoenzymatic analysis of interactions of anti-PDl antibodies with PDI receptors of different organisms
ELIS A was used to measure relative affinity of antibodies to PDI receptors of different organisms. ELISA plate wells (medium binding from Greiner bio one) were used to measure the binding. ELISA plate wells were coated with 50 μΐ of human and Javanese monkey PDI-Fc, PDI of mouse, rat, dog, rabbit, guinea pig (0.5 pg/ml in lx coating carbonate buffer, pH 9.5), hermetically closed and incubated overnight at 4°C. Ail further stages were conducted in accordance with the standard ELISA protocol. Anti-PDl antibody specifically binds to human and cynomolgus monkey PD 1 and does not bind to other test receptors (Fig. 9).
Example 12
Immunoenzymatic analysis of interactions of anti-PDl antibodies with CD28 family receptors
ELISA was used to measure relative affmity of antibodies to CD28 family receptors ELISA plate wells (medium binding from Greiner bio one) were used to measure the binding. ELISA plate wells were coated with 50 μΐ of human PDI-Fc, CD28, CTLA-4 and ICOS-Fc (0.5 μg/ml in lx coating carbonate buffer, pH 9.5), hermetically closed and incubated overnight at 4°C. Ail further stages were conducted in accordance with the standard ELISA protocol. Anti-PDl antibody only binds to PDI and does not bind to other CD28 family members (Fig. 10).
Example 13
Analysis of interactions of anti-PDl antibodies with human and cynomolgus monkey PDI receptors on Octet RED 96
Affmity constants of an antibody binding to human and cynomolgus monkey PD-1 was investigated on OctetRed 96 (ForteBio). BCD100 antibodies were non-specifically immobilized on the surface of amine reactive second-generation sensors (ForteBio, Pall) according to the standard protocol described in the manufacturées manual in regard to préparation and immobilization of AR2G sensors. An analysis was conducted at 30 C using PBS comprising 0.1% Tween-20 and 0.1% BSA as a working buffer. Human and cynomolgus monkey PD-1 was titrated with working buffer from a concentration of 126 nM to 2 nM with an incrément of 2.
Binding curves, after subtracting a reference signal, were analyzed using Octet Data Analysis software (Version 7.0) in accordance with the standard procedure and using 1:1 interaction model. Anti-PDl antibody specifically binds to human and cynomolgus monkey PDI antigen (Fig. 11).
Example 14
Analysis of interactions of anti-PDl antibodies with FcRn and Fc/ receptors using Octet RED 96
Forte bio Octet RED96 and streptavidin (SA) biosensors were used to analyze the interaction of antibodies with FcgRIIIaV, FcgRIIaH, FcgRIIb, FcgRIa, FcRn.
In the course of experiment, biotin-labeled receptors were orientally immobilized on the surface of streptavidin-coated sensors. Antibodies were diluted in sériés from a concentration of 500 pg/ml with an incrément of 2 by 7 points and placed in wells within a 96-well plate. Association stage was then carried out by virtue of immersing sensors in solutions of antibodies at various concentrations, further, dissociation stage was carried out by virtue of immersing sensors in a working buffer.
Working buffer PH7.4 was used to analyze antibody affinity to FcgRIIIaV, FcgRIIaH, FcgRIIb, FcgRIa, and working buffer PH6.0 was used to analyze affinity to FcRn.
The obtained curves, after subtracting a reference signal, were analyzed by SteadyState using 2:1 interaction model (heterogeneous ligand) in accordance with the standard procedure. The results are shown in Fig. 8. Analysis of affinity to Fcg receptors shows that effector functions of modified
IgGl are reduced and are approximately comparable to IgG4. Based on analysis of affinity to FcRn receptor, one can assume that pharmacokinetics of anti-PDI antibody is identical to that of Nivolumab antibody.
Example 15
Determining aggregation stability of anti-PDI antibody under thermal stress
BCD100 antibody préparation of 9 mg/ml in PBS buffer was heated for 12 hours at a température of 50°C. Aggregation after thermal stress was determined by high-performance gel-filtration chromatography. Chromatography was performed on a HPLC system (Agilent) on column Tosoh TSK-Gel G3000SWXL, 7.8 mm x 30 cm, order no. 08541 with precolumn Tosoh TSKgel Guard SWXL, 6.0 mm x 4.0 cm, with a particle diameter of 7 pm, order no. 08543. Elution was performed in isocratic mode, mobile phase: 50 mM NaFb, 0.3 M NaCl, pH 7.0 at a rate flow of 0.5 ml/min. Détection was performed at wavelengths of 214 and 280 nm. Samples of antibodies were diluted with FSB buffer, pH 7.5, to a concentration of ~1 mg/ml. Injection volume was 10 microliters. Calibration mixture Gel filtration standard (BioRad), order. no 151- 1901. was pre-chromatographed. Fig. 12 shows combined chromatograms: red - intact, blue - after 12 h incubation at 50°C. Anti-PD-1 antibody remains stable under thermal stress (différence in aggregate content in solution before/after thermal stress was less than 5%).
Example 16
Engineering of a stable cell line, production and purification of antiPDI antibody
A stable cell line producing BCD-100 monoclonal antibody was obtained by transfecting with electroporation using Néon Transfection System (Life Technologies) the parental suspension CHO-S cell line with vector constructs that comprised the optimum ratio of light and heavy antibody chains. High level clonal lineages (over 1 g/1) were obtained using ClonePix robotic platform (Molecular Devices) and preliminary minipool sélection stages using antibiotics in different cultivation formats. Productivity of selected clones was analyzed by Biomek FX robotics automated System (Beckman Coulter), and productivity was analyzed by Octet RED96 analytical System (Pall Life Sciences). DOE for selecting basic environment and cultivation scheme was carried out using Biomek FX robotics automated System (Beckman Coulter). Producer was cultured in serum-free media and feedings containing no animalderived proteins.
Culture liquid was filtered through a Zêta Plus Maximizer 60M02» (3M) depth filter. Primary purification of the antibody was performed on Protein A affinity sorbent. The target protein was specifically eluted with glycine buffer pH 3.3-3.8 under acidic conditions. The collected eluate was exposed to acidic pH for 30-60 min for the purpose of viral inactivation, and then neutralized with IM Tris-OH solution to pH 6.8-7.2. Final chromatographie purification to remove residual DNA, producer cell proteins, released affine sorbent's ligand, aggregates and antibody fragments was performed using CaptoAdhere sorbent (GE Healthcare LifeSciences) in a flow-through mode. Thus, the protein solution was flowed through prepared sorbent pH 6.8-7.2, under low conductivity (<2msec/cm2). The purified protein was then subject to virusremoving filtration using Viresolve PRO filter kit (Millipore), concentrating and diafiltration against the final buffer containing histidine buffer (pH 6.0-6.5), Tween 80 and trehalose. Protein concentration was 50 mg/ml and higher.
Example 17
Obtaining of a pharmaceutical composition comprising anti-PD-1 antibody of the invention
Antibody concentration to PDI (BCD-100) - 25 mg/ml, sodium acetate t/g - 0.436 mg, mannitol - 50 mg, Kolliphor (poloxamer) P188 - 0.2 mg, glacial acetic acid pH 5.15 Osm 300 ± 20 mOsm until pH 5.0
Example 18
Study of cross-reactivity of anti-PDl antibody in normal frozen human tissues
A study of cross-reactivity of anti-PDl préparation was performed on normal frozen human tissues (autopsy material). The following 33 human tissues were used in the analysis: hypophysis, retina, stomach, peripheral blood cells, cérébral cortex, skin, lung, lymph node, utérus, tonsils, cerebellum, mammary gland, bladder, ureter, adrenal gland, peripheral nerve, parotid gland, liver, pancréas, striated muscle, kidney, prostate, spleen, heart, spinal cord, large intestine, small intestine, fallopian tube, thyroid and parathyroid glands, vascular endothélium, testis, ovary. Frozen suspension of Jurkat-PDl cells comprising PDI membrane antigen was used as a positive control.
Manufactured frozen tissue blocks from pièces of organ tissues were embedded in Tissue-Tec (Sacura) tissue-filling and freezing medium, frozen in liquid nitrogen vapor and stored at -70°C. Precipitated suspension of Jurkat-PDl cells expressing PDI were used as a positive control, 1 * 10 6 cells/ml were resuspended in 1 ml of Tissue-Tec (Sacura) tissue-filling and freezing medium, resulting suspension was frozen in liquid nitrogen vapor, and stored at -70°C.
pm sections were prepared on Thermo HM525U cryostat. Further, sections were fixed with cold acetone for 10 minutes, dried in air at room température for 2-24 hours. Fixed sections were stored in the dark at -70°C.
Anti-PDl préparation (JSC Biokad, Russia) was labeled with FITC using FluoReporter FITC Protein Labeling Kit (Invitrogen) according to the manufacturées instructions.
Endogenous peroxidase was blocked. Sections were washed twice with PBS (0.05% Tween 20), endogenous peroxidase was blocked using Hydrogen Peroxide Block (Thermo) for 10 minutes at room température, and further washed twice with PBS.
Non-specific staining was blocked before the staining procedure as follows: sections were treated with Protein Block Serum-Free (Dako) for 10 minutes at room température. Primary antibodies were applied without washing.
Sections were coated with primary antibody at a working concentration of 0.2 pg/ml (anti-PDl antibody labeled with FITC, human IgGl isotype antibody at a concentration of 0.2 pg/ml) and incubated at room température for 1 hour, and then washed twice with PBS for 5 min each time.
A solution of murine monoclonal anti-FITC antibody conjugated to peroxidase (working dilution 1/1000) was incubated for 30 minutes at room température, and then washed twice with PBS for 5 min each time. Sections were treated with DAB staining solution for 10 minutes, and then washed twice with deionized water.
Nuclei were lightly stained with hematoxylin for 10 minutes, and then washed twice with deionized water. Sections were treated with 1% HCl solution for 1 second, and then three times washed with deionized water. Nuclei were stained with saturated lithium carbonate solution (blue) for 45 s. Once stained, sections were washed twice with deionized water, dehydrated in alcohol solutions with increasing concentration of alcohol (70%, 80%, 96%), clarified in a xylene replacer, embedded into mounting medium, and then cover-slipped using ClearVue (Thermo).
Semi-quantitative cytotoxic measurement of positive staining was performed. Screening of préparation was performed using Leica DM 6000B light microscope, and digital images were transmitted to a computer monitor via Leica DFC 420 video caméra and recorded on a hard disk using included Leica Application Suite (version 2.5.0.R1). For screening, 40x lens was used, with a spécifie adjusted (digital setting) standard level of micropreparation illumination. Staining intensity in an image is proportional to relative concentration of labeled substance, in this case it is anti-PD 1. In addition to visual assessment of immunopositive staining of cell types and structures, ImageJ digital image analysis software was used to measure the level of immunopositive staining in a selected area. Depending on staining intensity and total number of stained structures, évaluation was made using 0-3 point scale. Final immunopositive staining was evaluated taking into account intensity of staining of a tissue which was coated with control isotypie antibodies.
Example 19
Evaluation of in vivo efficacy of anti-PDI antibody
Efficacy was evaluated by humanized PBMC mice (The Jackson Laboratory) which were injected subcutaneously with human melanoma cell line A2058. Each animal in the group received 2.5x106 tumor cells. Cells were mixed with 5 Matrigel® (1:1) prior to administration. The obtained mixture was administered subcutaneously. Efficacy was evaluated using three doses of BCD-100 préparation, Keytruda® reference préparation (positive control) and normal human immunoglobulin préparation for intravenous administration (négative control) (Table 1).
Table 1. Efficacy évaluation scheme
Group Animal qty Préparation Days Method of administration Dose, mg/kg
1 6 U) BCD-100 7, 10, 13, 16, 19, 22, 25, 28 and 31 i.p. 5
2 6 (s) BCD-100 20
3 6 Ci) BCD-100 40
4 6 U) Keytruda® (Pembrolizumab) Positive control 40
5 6 U) IVIg Vehivle vontrol 40
In the course of the experiment, weight (before injection, and then twice a week) 15 and volume of the tumor node in animais were evaluated using the following formula:
W2 x L x 0.536, where W - width of tumor node, L - length of tumor node.
Animais were euthanized on day 37 of the experiment. Before euthanizing, the animal's blood was sampled to evaluate the level of circulating human blood lymphocytes of the following subpopulations:
o CD45+- CD20 + o CD45+ CD3 + o CD45+ CD3 + CD4+ û CD45T CD3+ CD8 + û CD45+ CD3- CD(16 + 56) +
B-lymphocytes T- lymphocytes T-helper cells Cytotoxic T cells NK-cells
Example 20
Anti-PDl antibody toxicokinetic évaluation
The below toxicokinetic study was carried out on cynomolgus monkeys using three BCD-100 dosage levels. A scheme for the experimental groups is shown in Table 2 below.
îo Table 2. Toxicokinetic study scheme
Group no. Animal qty Préparation Method of administratio n Dose, mg/kg
1 3(0) Anti-PD-1 monoclonal antibody intravenous 10
2 3(0) Anti-PD-1 monoclonal antibody 70
3 3(0) Anti-PD-1 monoclonal antibody 140
4 3(0) Placebo -
The following parameters were evaluated during the study:
- results of clinical examinations;
- animal weight (before administration and on day 4, 8, 22, 42 of the 15 experiment);
- body température (before administration and after 1, 2, 4, 6, 24 hours after administration, on day 4, 8, 22, 42 of the experiment);
- urinalysis (before administration and on day 4, 8, 22, 42 of the experiment);
- complété blood analysis on the following parameters: number of érythrocytes, number of leucocytes, hemoglobin concentration (before administration and on day 4, 8, 22, 42 of the experiment);
- biochemical analysis of sérum on the following parameters: lactate dehydrogenase, total bilirubin, total protein, glucose, aspartate aminotransferase, alanine aminotransferase (before administration and on day 4, 8, 22, 42 of the experiment);
- examination of concentration of préparation in the blood sérum of primates (5, 15 minutes and 0.5, 1, 3, 6, 24, 30, 48, 72, 96, 120, 144, 168, 192, 264, 336, 408, 576, 888, 984 hours after administration).
Example 21
Evaluation of toxicity in case of multiple subcutaneous administrations in cynomolgus monkeys for three months followed by a period without administrations for three months
Examination of toxicity in the case of multiple subcutaneous administrations for three months followed by a recovery period of three months was performed on relevant animais - cynomolgus monkeys. Three dosage levels were used in the experiment. Scheme for the experimental groups is shown in Table 3 below.
Table 3. Scheme for évaluation of toxicity in case of multiple subcutaneous administrations
Group no. Animal qty Préparation Method of administratio n Dose
1 3(ί) Anti-PDl monoclonal antibody intravenous 5.0 mg/kg
3(9)
2 3(0) Anti-PDl monoclonal antibody 30.0 mg/kg
3 (?)
3 3(0' Anti-PDl monoclonal antibody 70.0 mg/kg
3 (?) *
3(0)
3(9)
4 3(0) Placebo -
3(9)
The following parameters were evaluated during the study:
- results of clinical examinations;
- animal weight (before administration and on weeks 1, 3, 5, 7, 9, 11, 13 thereafter);
- body température (before administration and then weekly until termination of the experiment);
- effect on cardiovascular System based on bioelectric activity of heart evaluated by Poly-Spectrum cardiograph; évaluation was performed before administration and then on week 5, 9, 13, 18, 22, 26 of the experiment;
- urinalysis (before administration and on week 1, 3, 5, 7, 9, 11, 13 of the experiment);
- complété blood analysis on the following parameters: number of érythrocytes, number of leukocytes, hemoglobin concentration, number of lymphocytes, number of monocytes, number of neutrophils, number of eosinophils, number of basophils, number of platelets (before administration, and then once a week starting from the first week of the experiment);
- évaluation of effect on blood coagulation System on the following parameters: - activated partial thromboplastin time, fibrinogen concentration, prothrombin time (before administration, then once in two weeks for the period of administration, starting from the second week of the experiment, for the recovery period on week 15, 20 and 25 of the experiment);
- biochemical analysis of sérum on the following parameters: sodium, potassium, créatinine, urea, alkaline phosphatase, lactate dehydrogenase, total bilirubin, total protein, glucose, triglycérides, aspartate aminotransferase, alanine aminotransferase, total cholestérol (before administration and on week 4, 8, 12, 15, 20 of the experiment);
- at the end of the period of administration, animais of satellite group 3* were euthanized, followed by pathomorphological examination thereof; at the end of the study, animais of group 3 and control group were euthanized, followed by pathomorphological examination thereof;
- as part of the toxicity study, local irritant effects of préparations were also evaluated, and soft tissues located near the injection areas were therefore selected and histologically examined.
Example 22
Evaluation of immunotoxicity of anti-PDl antibody préparation
Examination of immunotoxicity in case of multiple subcutaneous administrations for three months followed by a recovery period for three months was performed on relevant animais - cynomolgus monkeys. Three dosage levels were used in the experiment. The scheme for the experimental groups is shown in Table 4 below.
Table 4. Scheme for évaluation of immunotoxicity in case of multiple subcutaneous administrations
Group no. Animal Qty Préparation Method of administration Dose
1 3(7) Anti-PD-1 monoclonal antibody intravenous 5.0 mg/kg
3 (?)
2 3(7) Anti-PD-1 monoclonal antibody 30.0 mg/kg
3 (?)
3 3(6) 3 (?) Anti-PD-1 monoclonal antibody 70.0 mg/kg
4 3(6) 3($) Placebo -
The following parameters were evaluated during the study:
- subpopulation composition of lymphocytes which was evaluated before préparation administration and then on week 3, 7, 13, 21 and 26 of the experiment;
- ratio of immunoglobulin classes were evaluated before administration and on week 4, 8, 12, 20, 25 of the experiment;
- effect on phagocytosis was evaluated before administration and on week 3, 7, 13, 21, 26 of the experiment;
- reaction of blast-transformation of lymphocytes was measured before administration of préparation and then on week 5, 13, 21, 26 of the experiment.
Example 23
Evaluation of pharmacokinetics and immunogenicity in case of multiple subcutaneous administrations of anti-PD-1 antibodies
Examination of pharmacokinetics and immunotoxicity in case of multiple subcutaneous administrations for three months, followed by a recovery period for three months was performed on relevant animais - cynomolgus monkeys. Three dosage levels were used in the experiment. Scheme for the experimental groups is shown in Table 5 below.
Table 5. Scheme for évaluation of toxicity in case of multiple subcutaneous administrations
Group no. Animal qty Préparation Method of administratio n Dose
1 3 (3) Anti-PD-1 monoclonal antibody intravenous 5.0 mg/kg
3 (?)
2 Anti-PD-1 monoclonal antibody 30.0 mg/kg
3 (?)
3 3 0) Anti-PD-1 monoclonal antibody 70.0 mg/kg
3 (9)
To evaluate dynamics of préparation concentration and to subsequently calculate pharmacokinetic parameters, blood sérum of animais was taken before administration of préparation and then on day l, 2, 8, 9, 15, 16, 22, 23, 29, 30, 5 36, 37, 43, 44, 50, 51, 57, 58, 64, 65, 71, 72, 78, 79, 85, 86, 92, 99, 106, 113, 120, 127, 134, 148, 162, 176 ofthe experiment.
Immunogenicity was evaluated by the level of binding antibodies, and for this purpose, blood was taken and sérum was separated before administration of préparation and then on week 4, 8, 12, 20, 26 of the experiment.

Claims (27)

  1. Claims
    1. An antibody or an antigen binding fragment thereof having the ability to bind to a human PD-1 receptor comprising an amino acid sequence of SEQ ID NO: 3.
  2. 2. The antibody or fragment thereof according to claim 1, characterized in that the antibody or fragment thereof contains:
    - a sequence of a heavy chain variable domain that is at least 75% identical to SEQ ID NO:7, and
    - a sequence of a light chain variable domain that is at least 75% identical to SEQ ID NO:8.
  3. 3. The antibody or fragment thereof according to claim 1, characterized in that the binding domain comprises the amino acid sequences of SEQ ID NO: 1-3.
  4. 4. The antibody or fragment thereof according to claim 1, characterized in that the binding domain competes for binding or binds to the same epitope as the binding domain comprising the amino acid sequence of SEQ ID NO: 7.
  5. 5. The antibody or fragment thereof according to claim 1, characterized in that the binding domain is at least 90% identical to SEQ ID NO: 7.
  6. 6. The antibody or fragment thereof according to claim 1, characterized in that a heavy chain variable domain comprises the amino acid sequence ofSEQ ID NO: 7.
  7. 7. The antibody or fragment thereof according to claim 1, characterized in that a light chain variable domain comprises the amino acid sequence of SEQ ID NO: 8.
  8. 8. The antibody or fragment thereof according to claim 1, characterized in that the binding domain is humanized.
  9. 9. The antibody or fragment thereof according to any one of claims 1-8, characterized in that it relates to one of the following human isotypes IgGl, IgG2, IgG3, IgG4.
  10. 10. The antibody or fragment thereof according to claim 1, which binds to human PD-1 and has a heavy chain sequence that is at least 90% identical to SEQ ID NO 9.
  11. 11. The antibody or fragment thereof according to claim 1, which binds to human PD-1 and has a light chain sequence that is at least 90% identical to SEQ IDNO 10.
  12. 12. The antibody or fragment thereof according to claim 1, which binds to human PD-1 and is characterized in that a heavy chain comprises the amino acid sequence of SEQ ID NO 9.
  13. 13. The antibody or fragment thereof according to claim 1, which binds to human PD-1 and is characterized in that a light chain comprises the amino acid sequence of SEQ ID NO 10.
  14. 14. The antibody or fragment thereof according to claim 1, wherein the Fc constant portion comprises any mutations that reduce or abolish any of effector functions ADCC, ADCP or CDC, as compared to the natural sequence.
  15. 15. The antibody or fragment thereof according to claim 1, wherein the Fc constant portion comprises mutations that increase animal or human pharmacokinetic parameters, such as ti/2p (hour) or Cmax (pg/ml).
  16. 16. The antibody or fragment thereof according to claim 1, characterized in that it has at least one of the following properties:
    a) aggregation stability: the aggregate content does not increase by more than 5% of the initial content in solution at concentrations above 10 mg/ml and at a storage température of T = 4°C for more than 6 months;
    b) aggregation stability: the aggregate content does not increase by more than 5% of the initial content in solution at concentrations above 10 mg/ml and with an increase in température to 37 °C for more than 2 weeks;
    c) aggregation stability: the aggregate content does not increase by more than 5% of the initial content in solution at concentrations above 10 mg/ml and with an increase in température to 50 °C for more than 6 hours;
    d) a dissociation constant KD of not more than ΙΟ'9 (M) when binding to human PD-1;
    e) a kinetic association constant kon (1 / Ms) of at least 105 (1/Ms) when binding to human PD-1 ;
    f) a kinetic dissociation constant dis (1/s) of not more than 10'4 (1/s) when binding to human PD-1.
  17. 17. A bispecific antibody comprising an antigen-binding fragment of an antibody according to any one of daims 1-16.
  18. 18. An isolated nucleic acid molécule encoding an antibody or an antigenbinding fragment thereof according to any one of daims 1-16.
  19. 19. An expression vector comprising any of the isolated nucleic acid molécule according to claim 18.
  20. 20. A host cell comprising the isolated nucleic acid molécule according to claim 18.
  21. 21. A method for producing a host cell according to claim 20, comprising transfecting a suitable stem cell with an expression vector according to claim 19.
  22. 22. A method for preparing an antibody according to any one of daims 1-16, comprising: producing a host cell according to claim 20, culturing a host cell under conditions sufficient to produce said antibody or fragment thereof, and isolating and purifying the obtained antibody or active fragment thereof.
  23. 23. A pharmaceutical composition comprising an antibody or fragment thereof according to any one of daims 1-17, in combination with one or more pharmaceutically acceptable excipients, diluents or vehicles.
  24. 24. The pharmaceutical composition according to claim 23, intended to be used for the treatment of oncological and infectious diseases.
  25. 25. An antibody or fragment thereof according to any one of daims 1-16, or a pharmaceutical composition according to claim 23 or claim 24, for use in a method of inhibiting PD-1 activity in a subject, the method comprising administering an effective amount of the antibody or fragment thereof, or the pharmaceutical composition, to the subject.
  26. 26. An antibody or fragment thereof according to any one of daims 1 -16, or 5 a pharmaceutical composition according to claim 23 or claim 24, for use in a method of treatment of oncological and infectious diseases, the method comprising administering an effective amount of the antibody or fragment thereof, or the pharmaceutical composition, to the subject.
  27. 27. The antibody or fragment thereof, or pharmaceutical composition, for io use according to claim 26, wherein the oncological disease is melanoma.
OA1201800540 2016-07-13 2017-07-04 Anti-Pd-1 antibodies, method for producing same and method for using same. OA19651A (en)

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
RU2016128487 2016-07-13

Publications (1)

Publication Number Publication Date
OA19651A true OA19651A (en) 2020-12-31

Family

ID=

Similar Documents

Publication Publication Date Title
US11136408B2 (en) Anti-PD-1 antibodies, method for producing same and method for using same
TWI784976B (en) Anti-lag-3 antibodies and compositions
TWI751981B (en) Anti-pd-1 antibodies and compositions
WO2018113258A1 (en) Anti-pd-1 antibody and use thereof
ES2616355T3 (en) Antibodies for the human programmed death receptor PD-1
US20160194402A1 (en) Cd70-binding peptides and method, process and use relating thereto
PT2426150T (en) Anti-nkg2a antibodies and uses thereof
JP7145895B2 (en) recombinant bispecific antibody
WO2021217893A1 (en) Antibody binding to tigit antigen, preparation method therefor and use thereof
US20210301022A1 (en) Antibody molecules
WO2021063352A1 (en) Anti-pd-l1 antigen binding protein, and application thereof
CA3156983A1 (en) Antibodies against the poliovirus receptor (pvr) and uses thereof
US20230303711A1 (en) Anti-cd47 antibody and use thereof
WO2018034597A1 (en) Antibody or an antigen-binding fragment thereof capable of binding to a human receptor of interleukin-6
EP4292611A1 (en) Anti-cd112r antibody and use thereof
KR20230057428A (en) Development of drug therapeutics containing adapters and their uses
OA19651A (en) Anti-Pd-1 antibodies, method for producing same and method for using same.
CN110023335B (en) Anti-PD-1 antibodies, methods of producing and methods of using the same
WO2023138579A1 (en) Anti-b7-h7 antibody or antigen-binding fragment thereof, and preparation method therefor and use thereof
WO2022171108A1 (en) Anti-pd-l1 antibody and use thereof
JP6529602B2 (en) Anti-CD20 / anti-BAFF bispecific antibody
OA19502A (en) Antibody Or An Antigen-Binding Fragment Thereof Capable Of Binding To A Human Receptor Of Interleukin-6.
NZ793343A (en) Anti- LAG-3 antibodies and compositions