NZ615477B2 - Tumor necrosis factor-? humanized antibody - Google Patents

Tumor necrosis factor-? humanized antibody Download PDF

Info

Publication number
NZ615477B2
NZ615477B2 NZ615477A NZ61547712A NZ615477B2 NZ 615477 B2 NZ615477 B2 NZ 615477B2 NZ 615477 A NZ615477 A NZ 615477A NZ 61547712 A NZ61547712 A NZ 61547712A NZ 615477 B2 NZ615477 B2 NZ 615477B2
Authority
NZ
New Zealand
Prior art keywords
seq
antibody
substituted
monoclonal antibody
cdr
Prior art date
Application number
NZ615477A
Other versions
NZ615477A (en
Inventor
Zhenxiang Hu
Qiang Li
Ruey Shyan Liou
Yucai Peng
Bill Naichau Sun
Cecily Rouyun Sun
Leehwei King Sun
Desheng Tao
Jingwei Zhang
Baoguo Zhu
Original Assignee
Livzon Mabpharm Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from CN201210026698.5A external-priority patent/CN102675460B/en
Application filed by Livzon Mabpharm Inc filed Critical Livzon Mabpharm Inc
Publication of NZ615477A publication Critical patent/NZ615477A/en
Publication of NZ615477B2 publication Critical patent/NZ615477B2/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • A61P31/18Antivirals for RNA viruses for HIV
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/20Antivirals for DNA viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/20Antivirals for DNA viruses
    • A61P31/22Antivirals for DNA viruses for herpes viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P33/00Antiparasitic agents
    • A61P33/02Antiprotozoals, e.g. for leishmaniasis, trichomoniasis, toxoplasmosis
    • A61P33/06Antimalarials
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/24Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against cytokines, lymphokines or interferons
    • C07K16/241Tumor Necrosis Factors
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/24Immunoglobulins specific features characterized by taxonomic origin containing regions, domains or residues from different species, e.g. chimeric, humanized or veneered
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/30Immunoglobulins specific features characterized by aspects of specificity or valency
    • C07K2317/33Crossreactivity, e.g. for species or epitope, or lack of said crossreactivity
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/76Antagonist effect on antigen, e.g. neutralization or inhibition of binding
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/90Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/90Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
    • C07K2317/92Affinity (KD), association rate (Ka), dissociation rate (Kd) or EC50 value
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Abstract

Disclosed is a humanised anti-TNF monoclonal antibody comprising the CDRs and FRs of the sequences as defined in the specification. Also disclosed is its use in treatment.

Description

TUMOR NECROSIS FACTOR - ! HUMANIZED ANTIBODY TECHNICAL FIELD The present invention relates to at least a humanized anti tumor necrosis factor-! (TNF!) or fragments thereof, including specific parts or variants, and nucleic acid encoding the humanized anti-TNF! antibody and its complementary nucleic acid, vectors, host cells, and the preparation method thereof, and compositions and kits comprising the humanized anti-TNF! antibody, and the use thereof.
BACKGROUND Human tumor necrosis factor-! (TNF!) is a proinflammatory cytokine produced by monocytes and macrophages, which is a 26 kDA precursor protein when initially produced with N terminal inside the cells and C terminal outside the cells, named transmembrane type TNF!. Pennica et al. cloned TNF! gene cDNA for the first time in 1984, and deduced that human TNF! molecule is composed of 157 amino acid residues, and weights about 17 KD (Pennica D, et al, Nature 1984!312:724)"Huamn TNF has two molecular forms, TNF! and TNF". TNF! is produced by activated macrophages or monocytes, and causes neoplastic tissues hengrrhagie necrosis, thus it is also called Cachectin. TNF" is mainly secreted by active T lymphocytes. Both have similar pyretogenesis. TNF! acts on receptors on the surface of oncocytes, and breaks into lysosome by identifying the cell, binding and endocytosing, and then activates lysosomes and proteases to cause cell death. TNF! plays an important role in immune response, inflammation, and response to injury, majorly affects the regulation of cell proliferation and cell apoptosis. Besides the effects on tumor cell such as cytotoxicity, cytolysis, induction of apoptosis and cell proliferation suppression, TNF! also can facilitate cell differentiation of myeloid leukemia cell to macrophage, and improve the phagocytic activity of neutrophile granulocyte.
An appropriate amount of TNF! can activate immune system to enhance immunity of the body, and play an important role in defense system of host resisting microbial invasion and tumor inhibition. But when over expressed, TNF! may cause several pathologic damages with other inflammatory factors. Therefore, the activities of TNF! may be suppressed or neutralized at different levels to block it from approaching receptors, in turn avoid the consequence of signal transduction.
For the purpose of overcoming relevant problems caused by using non- human antibodies, it is a relatively effective strategy of treatment to construct human- murine chimeric antibody to decrease organism immunogenicity initiated by HAMA.
Such kind of chimeric antibody is made by incorporating non-human antibody variable region into human antibody constant region while retaining amino acid sequences of original heavy chain, light chain variable regions of non-human antibody (see, Daddona, P.E et al. PCT publication WO92/16553, Le, J. et al., U.S. Patent NO. 5,919,452, Kang, Heui H et al. PCT publication WO2005/047329, Jin BOquan et al. Chinese patent publication CN1544466A), Jin Yihui et al. Chinese patent publication CN101177453 provides a chimeric antibody which can bind to human tumor necrosis factor.
Compared to non-human antibody, the immunogenicity of the chimeric antibody decreases, however, it may cause HAMA response in varied degrees since the murine derived portion in the chimeric antibody is still high, specifically including skin mucosa reaction, allergic reaction, arrhythmia and stenocardia, renal insufficiency, even coma when severe. Therefore, the clinical applications of this kind of chimeric antibodies are greatly limited.
Clinical trials demonstrate that this kind of chimeric antibodies as heterogeneous protein may cause immunological rejecting response of the heterogeneous protein by organism immune system (i.e., Human anti mouse antibody, HAMA response) when administrated to human. The response leads to rapid clearance of the murine antibody in human bodies, and short half life. Repeated administration may even result in severe anaphylactic shock. Moreover, the “foreign” antibody may be attacked by immune antibody, so that they may be neutralized before presenting pharmaceutical effects.
The inventors develop a new technique to prepare humanized antibody by utilizing genetic technology for the purpose of reducing the murine derived portion to minimum in the chimeric antibody on the basis of aforesaid patents. The technique comprises separately incorporating complementary determining regions (CDRs) of murine antibody heavy chain variable region and light chain variable region into human antibody framework region (FR). The obtained humanized antibody is similar to human sequence in structure as possible, meantime, it also can maintain CDR conformation similar to parent non-human antibody. Compared to parent non-human antibody and chimeric antibody, the portion of parent non-human amino acid sequence in the engineered humanized antibody decreases, one hand, the ability of antibody recognizing antigen is remained; the other hand, the immunogenicity of murine antibody has been greatly decreased. Therefore, safety of the antibody in clinical applications has been improved.
Consequently, the present invention provides a humanized antibody, which is safer, has longer half life more significant effects in human body, compared to murine chimeric antibody in prior arts.
SUMMARY OF THE INVENTION In one aspect, the present invention provides at least one humanized anti tumor necrosis factor monoclonal antibody, or specified complementary determining regions, heavy chain or light chain variable region, heavy chain or light chain constant region, framework region, or any other arbitrary parts, thereof. The antibody of the present invention may be derived from any mammals, for example, but not limited to human, mouse, rat, rodent, primate or any combination thereof.
The antibody of the present invention specifically binds to at least one epitope of TNF proteins, subunit, fragment, part thereof, or any combination thereof.
The said at least one epitope may comprise at least on antibody binding area. The said at least one epitope may optionally comprise at least one complementary determining area (CDR) (for example, heavy chain variable region or light chain variable region) and/or at least constant or variable framework region (FR) or any arbitrary part. The amino acid sequence of the said at least antibody may further optionally comprise insertion, deletion or conservatively substitution of at least one amino acid residue.
In another aspect, the present invention provides at least one nucleic acid molecules, the nucleic acid molecule comprises polynucleotide encoding at least one humanized anti tumor necrosis factor antibody of the present invention, or is complementary to or hybridized to the polynucleotide encoding at least one humanized anti tumor necrosis factor antibody of the present invention, wherein the antibody comprises at least one specific sequence, domain, part or variant thereof.
In another aspect, the present invention provides a recombinant vector comprising the nucleic acid molecule encoding the humanized anti tumor necrosis factor antibody, a host cell comprising the nucleic acid and/or recombinant vector, and the preparation method and/or use of the nucleic acid, vector and/or host cell.
At least one antibody of the present invention has at least one activity, for example, but not limited to neutralizing the toxicity of rhTNF! to L929 target cell, Suppressing and/or competing the binding of TNF with receptor and/or other monoclonal antibody for example, but not limited to Humira.
In another aspect, the present invention provide the use of antibody and/or composition of the present invention in inhibiting hTNF! activities, wherein the hTNF! related disease is selected for the group consisting of pyaemia, autoimmune diseases, malignant tumor, lung function disorder, transplant rejection, bacterial meningitis, cerebral malaria, AIDS and AIDS related complex (ARC), secondary cytomegalovirus infection after transplantation.
In another aspect, the present invention provides the use of in preparation of a medicament in diagnostic analysis of hTNF!, wherein the humanized anti tumor necrosis factor antibody also may be labeled by detecting molecule and/or may not be labeled, and the label comprises radioactive isotope; fluorescence label; various kinds of enzyme substrate mark.
In another aspect, the present provides the use of antibody and/or composition of the present invention in analysis method, wherein the analysis method includes competitive binding analysis, direct or indirect sandwich analysis, or immunoprecipitation analysis.
In another aspect, the present invention provide at least a composition, which comprises the humanized anti tumor necrosis antibody and/or its encoding nucleic acid, one or more auxiliaries selected from but not limited to pharmaceutically accepted vector, excipients, diluent, and additive. The composition may further optionally comprise at least one other antibody, nucleic acid, auxiliaries or any combination.
In another aspect, the present invention provides a kit comprising predetermined amount of reagents and instruction, the reagents comprise the antibody, nucleic acid and/or composition of the present invention. The kit further comprises other additives for example, but not limited to stabilizer, buffers.
BRIEF DESCRIPTION OF FIGURES Persons skilled in the art may understand that the following figures are for the purpose of illustrating the present invention, which do not limit the scope of the present invention in any manners.
Figure 1 shows the curve of the antibody neutralizing the effect of TNF! killing U937.
Figure 2 shows the score of degree of rat joint swelling induced by type II collagen.
Figure 3A shows the score of Tg197 mouse arthritis, figure 3B shows the histology evaluation of treating group of Tg197 mouse, figure 3C shows the effect of treating group on arthritis score (AS) and histology score (HS) of Tg197 mouse.
DETAILED DESCRIPTION OF THE INVENTION Anti tumor necrosis factor antibody The antibody of the present invention comprises antibody amino acid sequence encoded by any suitable polynucleotide, or any separated or prepared antibody. The humanized antibody or antigen binding fragment preferably binds to human tumor necrosis factor, and as result, partially, substantially or completely neutralizes at least one activity of human tumor necrosis factor, consequently inhibits the signal transduction process and physiological process mediated by the binding of TNF with TNF receptor.
The humanized antibody of the present invention may be any type (IgG, IgA, IgM, IgE, IgD, et al.) or isotype, and may comprise # or $ light chains, and!, µ, %, & or ' heavy chains.
At least one antibody of the present invention binds to at least one epitope of TNF protein, subunit, fragment, part thereof, or any combination thereof.
At least one anti tumor necrosis factor monoclonal antibody comprises following amino acid sequence, wherein the amino acid sequence of heavy variable region of the antibody is shown as SEQ ID NO: 1, and the amino acid sequence of light variable region of the antibody is shown as SEQ ID NO: 2.
SEQ ID NO: 1: QVQLVQSGPELKKPGASVKISCKASGYTFTHYGMHWVKQTPGRGLK WVGWINTYTGEPTYDADFQGRFTFSLETSVSTAFLQINSLKDEDLATYFCARYDFDGF DYWGQGTTLTVSS SEQ ID NO: 2: ENVLTQSPPILSASPGERVTMTCRASSSITFNYLHWYQQKSGDSPKVW IYSTSNLVSGVPSRFSGSGSGTSYSLTISSLEAEDAATYYCQQYSDYPYTFGGGTKLEI wherein complementary determining region CDR-H1 of the heavy chain variable region has a sequence of SEQ ID NO: 3; CDR-H2 has a sequence of SEQ ID NO: 4; CDR-H3 has a sequence of SEQ ID NO: 5; wherein, within framework region FR-H1, A can be substituted by E at amino acid residue position 16, S can be substituted by T at position 17, I can be substituted by V at position 20 (e.g., as shown in SEQ ID NO: 11); within FR-H2, K can be substituted by R at position 3, G can be substituted by S at position 9 (e.g., as shown in SEQ ID NO: 12); within FR-H3, T can be substituted by V at position 3, E can be substituted by D at position 7, V can be substituted by T at position 10, F can be substituted by Y at position 14, S can be substituted by T at position 19, T can be substituted by V at position 27 (e.g., as shown in SEQ ID NO: 13); and complementary determining region CDR-L1 has a sequence of SEQ ID NO: 6; CDR-L2 has a sequence of SEQ ID NO: 7; CDR-L3 has a sequence of SEQ ID NO: 8; wherein, within framework region FR-L1, L can be substituted by M at position 11, R can be substituted by E at position 18, M can be substituted by I at position 21 (e.g., as shown in SEQ ID NO: 14); within FR-L2, W can be by L at position 13 (e.g., as shown in SEQ ID NO: 15); within FR-L3, S can be substituted by A at position 4, L can be substituted by V at position 22, A can be substituted by F at position 27 (e.g., as shown in SEQ ID NO: 16).
SEQ ID NO.3: HYGMH SEQ ID NO: 4: WINTYTGEPTYDADFQG SEQ ID NO: 5: YDFDGFDY SEQ ID NO: 6: RASSSITFNYLH SEQ ID NO: 7: STSNLVS SEQ ID NO: 8: QQYSDYPYT SEQ ID NO: 9: QVQLVQSGPELKKPG(E/A)(T/S)VK(I/V)SCKASGYTFTHYGMHWV(K/R) QTPGR(S/G)LKWVGWINTYTGEPTYDADFQGRF(T/V)FSL(E/D)TS(T/V)STA(F/Y)LQIN (T/S)LKDEDLA(T/V)YFCARYDFDGFDYWGQGTTLTVSS SEQ ID NO: 10: ENVLTQSPPI(M/L)SASPGE(E/R)VT(M/I)TCRASSSITFNYLHWYQQKS GDSPKV(W/L)IYSTSNLVSGVP(A/S)RFSGSGSGTSYSLTISS(V/L)EAED(A/F)ATYYCQ QYSDYPYTFGGGTKLEIK SEQ ID NO: 11: QVQLVQSGPELKKPG(E/A)(T/S)VK(I/V)SCKASGYTFT SEQ ID NO: 12: WV(K/R)QTPGR(S/G)LKWVG SEQ ID NO: 13# RF(T/V)FSL(E/D)TS(T/V)STA(F/Y)LQIN(T/S)LKDEDLA(T/V)YFCAR SEQ ID NO: 14: ENVLTQSPPI(M/L)SASPGE(E/R)VT(M/I)TC SEQ ID NO: 15: WYQQKSGDSPKV(W/L)IY SEQ ID NO: 16: GVP(S/A)RFSGSGSGTSYSLTISS(V/L)EAED(A/F)ATYYC In an embodiment of the present invention, the heavy chain constant region sequence of the humanized anti tumor necrosis factor antibody is the heavy chain constant region of human IgG1.
In an embodiment of the present invention, the light chain constant region sequence of the humanized anti tumor necrosis factor antibody is the light chain constant region of human antibody.
In a preferred embodiment of the present invention, the amino acid sequence of humanized anti tumor necrosis factor monoclonal antibody may be modified by inserting, deleting or conservatively substituting one or more amino acid residues, preferably 1-5 amino acid residues.
The monoclonal antibody modified or mutated by one or more insertion, deletion or conservatively substitution in any combinational forms may have differences in the amino acid sequences. In the preferred variant, the modification is obtained by amino acid conservative substitution from the aforesaid monoclonal antibody of the present invention. The conservative substation means a specific amino acid is substituted by another amino acid with similar properties. The amino acids below listed in non-limited manners are considered as conservatively exchangeable (with similar properties): a) alanine, serine and threonine; b) glutamic acid and aspartic acid; c) asparagine and glutamine; d) arginine and lysine; e) isoleucine, leucylacid, methionine and valine; and f) phenylalanine, tyrosine and tryptophan.
The functionally equivalent monoclonal antibody of the present invention is a variant, wherein one or more amino acid residues, preferably 1-5 amino acid residues are conservatively substituted. The conservative substitution includes: any one of aromatic amino acids Ala, Val, Leu and Ile can be substituted by another; hydroxyl residues Ser and Thr are exchangeable; acid residues Asp and Glu are exchangeable; amide residues Asn and Glun are exchangeable; basic residues Lys and Arg are exchangeable; aromatic residues Phe and Tyr are exchangeable.
Furthermore, present invention discloses the amino acid sequences which have at least 50% identity with the amino acid sequences of the present invention, or fragment thereof, and amino acid sequences with equivalent functions. In one embodiment, the amino acid sequences have at least 75% identity with the amino acid sequence SEQ ID NO:1 or 2 according to present invention, more preferably at least 85% identity, even more preferably at least 90% identity, even more preferably at least 95% identity, even more preferably at least 97% identity, and most preferably at least 99% identity.
Several kinds of antibodies are encompassed by present invention. For example, anti hTNF! antibody may be full-length antibody (for example, comprising complete human Fc region); or antibody fragment (for example, Fv, scFv, Fab, Fab’ and (Fab’)2). Moreover, the antibody can be labeled with detectable labels, fixed on solid phase carrier, and/or coupled with heterologous compounds (such as cytotoxin materials).
Fab is produced by treating IgG antibody molecule by protease/papain. It is an antibody fragment with molecular weight of about 50,000 and have antigen binding activity, wherein, in the fragment obtained by papain treatment (cleaving H chain at amino acid residue 224 ), about half of H chain from the N-terminal and the whole L chain are bound together by disulfide bonds. The Fab of the present invention also may be produced by inserting DNA encoding Fab of the antibody into prokaryotes expression vectors and/or eukaryotes expression vectors.
Fab’ is an antibody fragment with antibody binding activity, produced by cleaving disulfide bonds in (Fab’)2 hinge region, with molecular weight of about 50,000.
The Fab’ of the present invention also may be produced by inserting DNA encoding Fab’ of the antibody into prokaryotes expression vectors and/or eukaryotes expression vectors, and subsequently introducing the vectors into prokaryote and/or eukaryote to express Fab’.
(Fab’)2 is an antibody fragment with antigen binding activity, with molecular weight of about 50,000, wherein, in in the fragment obtained by protease/pepsin treatment (cleaving H chain at amino acid residue 234) of IgG antibody, the antibody fragment of Fab by bound together with disulfide bonds in hinge region is slightly larger. The (Fab’)2 of the present invention may be produced by treating antibody with pepsin. Besides, the (Fab’)2 of the present invention also may be produced by linking Fab’ with thioether bonds or disulfide bonds.
ScFv is an antibody fragment with antigen binding activity, consisting of a chain V and a chain V which are connected by appropriate peptide joints. The scFv of the present invention may be produced by obtaining cDNAs encoding V and V of the antibody, constituting DNA encoding scFv, inserting the DNA encoding scFv into prokaryotes expression vectors and/or eukaryotes expression vectors, and subsequently introducing the vectors into prokaryote and/or eukaryote to express scFv.
Nucleic Acid The nucleic acids of the present invention are nucleotide sequences encoding at least one of SEQ ID NO: 1-16, specific fragments, variants thereof, or at least 70-100% continuous amino acid sequences of consensus sequences. The nucleic acid molecules encoding at least one anti TNF antibody may be obtained by the methods described in present invention or known in the art.
Nucleic acid molecules of the present invention can be in the form of RNA, such as mRNA, hnRNA, tRNA or any other form, or in the form of DNA, including, but not limited to, cDNA and genomic DNA obtained by cloning or produced synthetically, or any combinations thereof. The DNA can be triple-stranded, double- stranded or single-stranded, or any combination thereof. Any portion of at least one strand of the DNA or RNA can be the coding strand, also known as the sense strand, or it can be the non-coding strand, also referred to as the anti-sense strand.
As indicated herein, nucleic acid molecules of the present invention which comprise a nucleic acid encoding an anti-TNF antibody can include, but are not limited to, those encoding the amino acid sequence of an antibody fragment, by itself; the coding sequence for the entire antibody or a portion thereof; the coding sequence for an antibody, fragment or portion, as well as additional sequences, such as the coding sequence of at least one signal leader or fusion peptide, with or without the aforementioned additional coding sequences, such as at least one intron, together with additional, non-coding sequences, including but not limited to, non-coding 5' and 3' sequences, such as the transcribed, non-translated sequences that play a role in transcription, mRNA processing, including splicing and polyadenylation signals (for example - ribosome binding'and stability of mRNA); an additional coding sequence that codes for additional amino acids, such as those that provide additional functionalities.
Thus, the sequence encoding an antibody can be fused to a marker sequence, such as a sequence encoding a peptide that facilitates purification of the fused antibody comprising an antibody fragment or portion.
The nucleic acids of the present invention can be made using (a) recombinant methods, (b) synthetic techniques, (c) purification techniques, or combinations thereof, as well-known in the art.
The nucleic acid compositions of the present invention, such as RNA, cDNA, genomic DNA, or any combination thereof, can be obtained from biological sources using any number of cloning methodologies known to persons skilled in the art.
In some embodiments, oligonucleotide probes that selectively hybridize, under stringent conditions, to the polynucleotides of the present invention are used to identify the desired sequence in a cDNA or genomic DNA library. The isolation of RNA, and construction of cDNA and genomic libraries, is well known to persons skilled in the art.
A cDNA or genomic library can be screened using a probe based upon the sequence of a polynucleotide of the present invention, such as those disclosed herein. Probes can be used to hybridize with genomic DNA or cDNA sequences to isolate homologous genes in the same or different organisms. Persons skilled in the art will appreciate that various degrees of stringency of hybridization can be employed in the assay; and either the hybridization or the wash medium can be stringent. As the conditions for hybridization become more stringent, there must be a greater degree of complementarity between the probe and the target for duplex formation to occur. The degree of stringency can be controlled by one or more of temperature, ionic strength, pH and the presence of a partially denaturing solvent such as formamide. For example, the stringency of hybridization is conveniently varied by changing the polarity of the reactant solution through, for example, manipulation of the concentration of formamide within the range of 0% to 50%. The degree of complementarity (sequence identity) required for detectable binding will vary in accordance with the stringency of the hybridization medium and/or wash medium. The degree of complementarity will optimally be 100%, or 70-100%, or any range or value therein. However, it should be understood that minor sequence variations in the probes and primers can be compensated for by reducing the stringency of the hybridization and/or wash medium.
Methods of amplification of RNA or DNA are well known in the art and can be used according to the present invention without undue experimentation, based on the teaching and guidance presented herein.
The isolated nucleic acids of the present invention can also be prepared by direct chemical synthesis by known methods. Chemical synthesis generally produces a single-stranded oligonucleotide, which can be converted into double- stranded DNA by hybridization with a complementary sequence, or by polymerization with a DNA polymerase using the single strand as a template.
Construction of Humanized Monoclonal Antibody Expression Vector 5’ fragment is obtained by using a plasmid (pHu-V ) comprising humanized antibody heavy chain variable region V gene fragment as template, 5' primer FVHX (5’-CGCGCAAG-CTTCCTCGAG-3’ SEQ ID NO: 17) and 3' primer RVCG (5’-CGATGGGCCCTTGGTGGA-3’ SEQ ID NO: 18), which comprises the gene for humanized antibody heavy chain variable region (V ) and 7 amino acid at 5'-teminal of human IgG heavy chain constant region (C ). Meanwhile, A gene comprising human 1 %1 IgG heavy chain constant region (C ) encoding sequence is obtained from RNA 1 %1 prepared from human leucocyte by using 5' primer HuCGF (5’- ACCAAGGGCCCATCGGTCTTC-3’; SEQ ID NO: 19) and 3' primer HUCGE (5’- CGGAATTCTCATTTACCCGGAGACAGGGA 3’, SEQ ID NO: 20) through reverse transcription and PCR. Finally, the fragment of humanized antibody heavy chain variable region and human C gene are linked by PCR using 5' primer (FVHX, SEQ ID NO: 17) and 3' primer (HUCGE, SEQ ID NO: 20) to obtain a gene fragment of length about 1400 bp comprising heavy chain encoding sequence. The gene fragment is treated with endonuclease Hind III and EcoR1, and then inserted into vectors such as PUC19 (ref: Yanisch-Perron, C., Vieira, J. and Messing, J. (1985) Gene, 33, 103-119). 5’ fragment is obtained by using a plasmid (pHu-V ) comprising humanized antibody light chain variable region V gene fragment as template, 5' primer FVHX (SEQ ID NO: 17) and 3' primer VKCKO (5’ -AGA TGG TGC AGC CAC AGT TCG CTT GAT CTC CAG CTT GGT GCC -3’ SEQ ID NO: 21), which comprises the gene for humanized antibody light chain variable region (V ) and 7 amino acid at 5'-teminal of human # light chain constant region (C#). Meanwhile, A gene comprising human # light chain constant region (C#) encoding sequence is obtained from RNA prepared from human leucocyte by using 5' primer HuCKF (5’ -GTG GCT GCA CCA TCT GTC TTC -3’ SEQ ID NO: 22) and 3' primer HUCKB (5’ -TGC GGA TCC CTA ACA CTC TCC CCT GTT GAA -3’, SEQ ID NO: 23) through reverse transcription and PCR. Finally, the fragment of humanized antibody light chain variable region and human C# gene are linked by PCR using 5' primer (FVHX, SEQ ID NO: 17) and 3' primer (HUCKB, SEQ ID NO: 23) to obtain a gene fragment of length about 700 bp comprising light chain encoding sequence. The gene fragment is treated with endonuclease Hind III and Bam H1, and then inserted into vectors such as PUC19 (ref: Yanisch-Perron, C., Vieira, J. and Messing, J. (1985) Gene, 33, 103-119.).
The cDNA encoding the heavy chain or light chain or the cDNA encoding their modified products which are obtained by aforesaid methods are inserted into pcDNA3 (Invitrogen USA, Carlsbad, CA, U.S.A.) vector to construct Hu_anti-TNF! humanized expression vector. The expression vector plasmid comprises cytomegalovirus early gene promoter-enhancer required for high level expression in mammal cells. Meanwhile, the vector plasmid also comprises optional maker gene, so as to have amicillin resistance in bacteria, have G418 resistance in mammal cells.
Furthermore, the vector plasmid comprises DHFR gene. In suitable host cells, chimeric antibody gene and DHFR gene can be co-amplified by Methotrexate (MTX, Sigma) (see, for example, Axel, R., et al. U.S Patent No. 5,179,017; Kaufman,R. and Sharp,P., J.Mol. Biol. 159:601-621,1982).
Antibody Host Cell The present invention also relates to produce at least one anti TNF antibody by using recombinant vector gene engineered host cell, and recombinant technique in the art.
Polynucleotides may be optionally linked to vector comprising optional labels, for amplification in the host. generally, the plasmid vector is introduced into a precipitate, such as calcium phosphate, or introduced into complex comprising charged lipid.
Appropriate culture mediums and conditions for the above-described host cells are known in the art. Suitable vectors will be readily apparent to persons skilled in the art. Introduction of a vector construct into a host cell can be effected by calcium phosphate transfection, DEAE-dextran mediated transfection, cationic lipid- mediated transfection, electroporation, transduction, infection or other known methods.
The host cells for the humanized anti tumor necrosis factor monoclonal antibody of the present invention are derived from Chinese hamster ovary cells, which are obtained by transfection with plasmid comprising anti-TNF! gene code, and followed by a series of stringent, specified screening, which including drug screening, gene amplification, and single cell cloning to produce the fanal cell strain. The host cell of the present invention, Chinese hamster ovary cell strain CHO HUAT 132 was deposited at CCTCC at March 7, 2011, with Deposit NO. C201117.
The cell of the cell strain can be bred when suspended in serum free medium; when cultivated in 2 L Fermenter, The level of secreted anti-TNF! in medium will not lower than 1 g/L after the cultivation cycle of 16-20 days ends. The anti-TNF! produced by the cell strains is humanized monoclonal antibody.
Activity of Binding to TNF! The humanized anti tumor necrosis factor monoclonal antibody of the present invention only has specific affinity with recombinant human tumor necrosis factor (rhTNF!, target molelules), but does not crosslinked to any other protein molecules. In competitive assay of affinity to their target molecules, the humanized anti tumor necrosis factor monoclonal antibody of the present invention has been determined to have a similar affinity as Humira.
The humanized anti tumor necrosis factor monoclonal antibody of the present invention also has the activity to neutralize the toxicity of rhTNF! to L929 target cell, and its EC50 is similar to that of Humira, which is between 20.4 ng/mL and 50 ng/mL.
Therapeutic Use Anti hTNF! antibody can be used to treat and/or prevent hTNF! related diseases, wherein the hTNF! related disease may be, for example, pyaemia, autoimmune diseases, malignant tumor, lung function disorder, transplant rejection, bacterial meningitis, cerebral malaria, AIDS and AIDS related complex (ARC), secondary cytomegalovirus infection after transplantation. The use of the antibody and antibody components of the present invention in treating hTNF! related diseases will be further discussed as follows: 1) Sepsis The role of tumor necrosis factor in sepsis pathology, and the biological effects thereof includes hypotension, myocardial depression, vascular leak syndrome, organ necrosis (see for example, U.S. patent 5,231,024). Therefore, the humanized antibodies of the present invention and antibody components can be used for the treatment of any sepsis with clinical background, including septic shock, endotoxic shock, gram-negative sepsis and toxic shock syndrome. 2) Autoimmune diseases It has been found that the tumor necrosis factor plays a role in the pathophysiology of a variety of autoimmune diseases, for example, it has been found that TNF! is involved in activating tissue inflammation and lead to joint destruction in rheumatoid arthritis (see, for example, U.S. Patent 5,231,024; Moeller, A. et al. (1990) Cytokine 2:162-169;). It also has been found that TNF! is involved in promoting islet cell death in diabetes and mediates the cytotoxicity to oligodendrocyte and induces inflammatory plaques.
Humanized antibodies and antibody components of the present invention can be used to treat autoimmune diseases, especially those associated with inflammation, including rheumatoid arthritis, rheumatoid myelitis, osteoarthritis and gout arthritis, allergy, multiple sclerosis, autoimmune diabetes, autoimmune eye uveitis, and nephrotic syndrome. 3) Malignant tumors It has been found that tumor necrosis factor has been found in malignant tumors is involved in inducing cachexia, stimulating tumor growth, enhancing metastatic potential and mediating cytotoxicity. Therefore, the antibodies and the antibody components of the present invention can be used to treat malignant tumors, inhibit tumor growth or metastasis and/or reduce the malignant secondary cachexia. The antibody or antibody component can be systemic or local administrated to the tumor site. 4) Pulmonary function disorders It has been known that the tumor necrosis factor is involved in pathophysiology of adult respiratory distress syndrome (ARDS), including stimulating white blood cell - endothelial cell activation, cytotoxicity-oriented to lung cells and inducing vascular leak syndrome. Thus, the antibody and the antibody components of the present invention can be used to treat lung function disorders, including adult respiratory distress syndrome, chronic pneumonia, pulmonary sarcoidosis, pulmonary fibrosis and silicosis. 5) Intestinal dysfunction Human antibodies and antibody components of the present invention can be used to treat intestinal disorders such as idiopathic inflammatory bowel disease, which includes two syndrome, Crohn and ulcerative colitis. 6) Transplantation It has been found that tumor necrosis factor may be the key mediators of allograft rejection and transplant plants versus-host disease (GVHD) , and mediates the side effects observed in the inhibition of renal transplant rejection by rat antibody OKT3 whhich is targeted to T-cell receptor CD3 complex (see for example, Suthanthiran, M., and Strom, TB (1994) New Engl J.Med.331 :365-375) Therefore, the antibody and the antibody components of the present invention can be used to suppress transplant rejection, including allograft and xenograft rejection, and suppress GVHD. 7) Infectious diseases The antibodies and the antibody components of the present invetion can be used to treat infectious diseases, including bacterial meningitis, cerebral malaria, AIDS and AIDS-related syndrome (ARC), and secondary cytomegalovirus infection after transplantation. They can also be used to reduce infectious diseases related symptoms, including fever and muscle pain caused by infection (eg influenza), and infection secondary cachexia (such as AIDS or secondary ARC).
Analysis and Diagnostic Purposes The antibody of the present invention can be used in any known analysis method, such as competitive binding assays, direct or indirect sandwich analysis and immunoprecipitation analysis. Zola, Monoclonal Antibodies: Technical Manual "(Monoclone Antibodies; A Manual of Techniques), pp. 147-158 (CRC Press, Inc., 1987).
Competitive binding analysis depends on the ability of marked standard material competing with analyte in the measured sample to bind limit amount of antibody. Amount of the standard material is inversely proportional to the amount of antibody bound with hTNF! in the measured sample. In order to facilitate the determination of the amount of the bound standard material, the antibodies usually are insoluble before or after the competition, so that it will be convenient to separate the bound standard material and the analyte from unbound standard material and analyte separation.
Sandwich analysis involves the use of two antibodies, each bind to different immunogenicity site or epitope on target proteins. In sandwich analysis, the measured sample analyte is bound to the first antibody fixed on the solid phase carrier, and then the second antibody binds to the analyte, thus forming insoluble three- component complex. See U.S. Patent 4,376,110. The second antibody itself can marked with detectable part (direct sandwich assay), or can be detected by using anti- immunoglobulin antibodies labeled with detectable part (indirect sandwich assay). For example, one of the sandwich analysis is ELISA, in which the detectable part is enzyme.
Anti-hTNF! antibodies can also be used in the diagnostic analysis of hTNF!, for example, to detect its expression in specific cells, tissues or serum. This diagnostic method can be used for the diagnosis of causes for autoimmune diseases The antibody usually can be labeled with detectable molecular. Many Markers can be used, and generally they can be classified as follows: (a) Radioisotopes, such as 111In, 99Tc, 14C, 131I, 125I, 3H, 32P or 35S. Antibodies can be labeled with radioactive isotopes in accordance with the methods decribed in Current Protocols in Immunology, Volume 1 and 2, Coligen eds, Wiley-Interscience, New York, New York, the Pubs (1991), in which the radioactivity may be determined by scintillation counting method, and the diseased sites can be located by using immune-flash photography. (b) Fluorescent marker, such as rare earth chelating agent (europium chelator), or luciferase and its derivatives, rhodamine and its derivatives, dansyl, lissamine, phycoerythrin and Texas Red. Fluorescent marker may be coupled with antibody using the methods described in for example, Current Protocols in Immunology mentioned above. Fluorescence can be quantified by fluorometer. (c) A variety of substrate markers are available, and U.S. Patent 4,275,149 disclosed some of them. The enzymes usually catalyze chemical changes of a variety of chromogenic substrates which can be detected by many techniques. For example, the enzymes catalyze the color changes of the substrates, which can be measured by spectrophotometer, or the enzymes change the fluorescence or chemiluminescence of the substrate. Previously, the technology of quantitative determination of the fluorescence changes has been described. Chemiluminescent substrates are electrically excited due to chemical reactions, and then illuminate. The light emitted can be determined (such as the use of chemical photometer) or provides energy to fluorescent receptors. The enzyme markers include such as luciferase (for example, firefly luciferase and bacterial fluorescence luciferase; U.S. Patent 4,737,456), luciferin, 2,3-dihydrophthalazinediones, malate dehydrogenase, urease, peroxidase such as horseradish peroxidase (HRPO), alkaline phosphatase, b-galactosidase, glucoamylase, lysozyme, sugar oxidase (eg glucose oxidase enzyme, galactose oxidase and glucosephosphate dehydrogenase), heterocyclic oxidases (such as uricase and xanthine oxidase), lactoperoxidase, micro-peroxidase. O’Sullivan describes the technology of conjugating enzymes to antibodies in Methods for the Preparation of Enzyme-Antibody Conjugates for use in Enzyme Immunoassay (Methods In Enzym.) (by J. Langone, and H. the Van Vunakis ed.), Academic press, New York, ,73:147-166 (1981).
The compositions of enzyme - substrate include, for example: (i) Horseradish peroxidase (HRP) and hydrogen peroxidase as substrate, wherein, the hydrogen peroxidase oxidize the dye precursor (eg, o-phenylenediamine (OPD) or hydrochloric acid to 3,3',5,5'- tetramethylbenzidine (TMB)); (ii) Alkaline phosphatase (AP) and p-nitrophenyl phosphate as chromogenic substrate; (iii) b-D-galactosidase anhydrase (b-D-Gal) and chromogenic substrate (for example, nitrophenyl-b-D-galactosidase) or fluorogenic substrate and 4-methyl umbelliferone-b-D-galactosidase.
Persons skilled in the art may know many other enzyme-substrate combinations. Review of these combinations can be found in U.S. patents 4,275,149 and 4,318,980. Sometimes, markers and antibodies are indirectly coupled. Persons skilled in the art also know all kinds of methods to obtain the said compositions. For example, the antibodies may coupled with biotin, and any one of the above three categories of markers can coupled with avidin, or vice versa. Biotin selectively binds to avidin, and the markers can be coupled with the antibody indirectly. Or, to coupled the marker with antibody indirectly, the antibody can be coupled with a small hapten (eg digoxin), while one of the markers of different types can be couple with anti-hapten antibody (eg anti-digoxin antibody). Therefore, the indirect coupling of markers with the antibody is obtained.
In another embodiment of the present invention, it is not necessary to mark the anti-hTNF! antibodies, and its existence can be detected by marked antibody which binds to the hTNF! antibody.
Affinity Purification Kit The antibodies of the present invention can be used as affinity purification reagents. In this method, the antibody is fixed on solid-phase, for example, Sephadex resin or filter paper by methods known in the art. The fixed antibody contact with hTNF!-containing samples to be purified, and then the carrier is washed with suitable solvents, and the solvent can substantially remove all the other materials expect hTNF! bound with immobilized antibody.
Pharmaceutical composition and mode of administration The antibody and antibody components of the present invention may be added into pharmaceutical composition suitable for administration to subjects, wherein the pharmaceutical compositions comprise the antibodies of the present invention and pharmaceutically acceptable excipients, and the pharmaceutical excipients include any physiological applicable solvents, dispersion media, antibacterial agents, antifungal agents, isotonic agents, coating, absorption delay agent. The pharmaceutical compositions of the present invention can take various forms, such as liquid semi-solid and solid dosage forms.
The anti-hTNF antibody of the present invention in a pharmaceutically acceptable dosage form can be administrated to human using known methods. The method includes intravenous (for example, intravenous injection of concentrated drug (bolus) or infusion within a period of time), intramuscular, intraperitoneal, cerebrospinal cavity, subcutaneous, intra-arterial, synovial cavity, intrathecal injection, oral, local injections, or inhalation.. The antibody can also be appropriately administrated via intratumor, around tumor, inside injury sites, around injury side to obtain local and systemic treatment. Intraperitoneal injection is expected to be particularly useful, for example, for the treatment of ovarian cancer.
In order to prevent or treat diseases, the appropriate dose of the antibody will depend on the type of the disease to be treated as defined above, disease severity and duration of disease, antibody given for prevention or for treatment, previous treatment, and patient history and antibody response, as well as the attending physician's independent judgment. The antibody is suitable for one-off or series doses to the patients.
According to the type and severity of the disease, no matter one dose or several separated doses, or continuous infusion, 1 µg/kg to 50 mg/kg (eg 0.1-20 mg/kg) of the antibody is the initial candidate dose to patients. The typical daily dose or weekly dose is about 1 mg/kg -20 mg/kg or more, depending on the factors mentioned above.
As to repeated dose within a few days or more (depending on condition), the treatment should be continued until the disease symptoms have been inhibited as desired.
However, the other regimen can also be used. The progress of treatment can easily monitored using conventional techniques and analysis methods.
Product 1) Injection Another embodiment of the present invention provides a product containing the material used for the treatment of these diseases. The product includes a container and a label. The suitable containers include such as ordinary bottles, medicine bottles, syringes and test tubes. The container can be made of various materials, such as glass or plastic. The container contains the effective composition for the treatment of diseases, and with a sterile entrance (for example, the container can be a plugged intravenous infusion bag or bottle, the stopper can be penetrated using a hypodermic needle). The active ingredient in the composition is anti-hTNF! antibody.
The label on the container or associated with the container associated demonstrates the specific conditions treated by the composition. The products can also have another container, which contains a pharmaceutically acceptable buffer, such as phosphate buffer, Ringer’s solution and glucose solution. According to business needs or the needs of users, it can include other materials, such as other buffers, diluents, filters, needles, syringes, and instructions. 2) Sustained Release Formulations The humanized anti-tumor necrosis factor monoclonal antibodies of the present invention can be used for the preparation of sustained-release formulations.
The suitable sustained-release formulations include such as semi-permeable matrix body comprising solid hydrophobic polymers of the antibodies, and the matrix body is a tangible object, such as film or microcapsules. The suitable sustained-release matrix body include such as polyesters, hydrogels (for example, poly(methacrylic acid 2- hydroxy ethyl) or polyvinyl alcohol), polylactide (U.S. Patent 3,773,919), L-glutamic acid and L-glutamic acid ethyl ester copolymer, non-degradable ethylene vinyl acetate, degradable lactic acid-ethanol acid copolymer such as Lupron DepotTM (injectable microspheres composed of lactic acid-glycolic acid copolymer and leuprolide acetate), and poly-D-(-)hydroxybutyric acid. Molecular polymers such as ethylene vinyl acetate and lactic acid-glycolic acid enable the release of molecules lasting for 100 days or more, while some hydrogels release proteins in a short period of time. When encapsulated antibodies are retained in the body for a long time, they may become denaturation or cohesion for contacting with water at 37°C, resulting in biological activity decrease and may lead to changes in immunogenicity. Reasonably stable strategy can be designed according to the mechanism. For example, if the condensation mechanism is the formation of intermolecular S-S bond by sulfur-disulfide interchange reaction, the stabilization can be achieved by modifying thiol residues, lyophilizing acidic solutions, controlling moisture content, using suitable additives and designing special polymer matrix composition.
For convenience, the antibody of the present invention can be provided in the form of kits, that is to say, predetermined amounts of reagents and the instruction for diagnostic analysis are packed together. If the antibody is enzymatic marked, the kit will contain the substrate and cofactors (such as the substrate precursors providing detectable chromophores and fluorophores) required for the enzyme. Besides, it may also include other additives such as stabilizers, buffers (such as blocking buffer or lysis buffer). In order that the provided reagent concentration can achieve the highest sensitivity of the analysis, the relative amounts of various reagents significantly change.
Specifically, the reagent can be dry powder, usually in lyophilized powder form, and excipients may be included. They will form a reagent solution with appropriate concentration upon dissolution.
The present invention is further set forth, in connection with following specific embodiments. It should be understood that these embodiments are for illustrating the present invention only, but not be used to limit the scope of the present invention. In the following embodiment, the experimental methods without indicating the specific conditions are usually performed in conventional conditions, for example, the conditions described in Sambrook et al, Molecular Cloning: Laboratory Manual (New York; the Cold Spring Harbor Laboratory Press, 1989), or the conditions recommended by the manufacturer.
Examples Example 1: production of anti-hTNF! mouse monoclonal antibody 1) Immune Some 7-11 weeks old female Balb/c mice were IP or ID immunized with the recombinant hTNF (rhTNF!, purchased from Pepro Tech Inc.), and the recombinant human TNF! was emulsified using an equal volume of TITERMAX or Freund's complete adjuvant with final volume of 100-400 µL. At 1-7, 5-12, 10-18, 17-25 and/or 21-34 days thereafter, each mouse were IP (1-400 µg) and SC (1400 µg(2) with TNF which was emusified with equal volume of TITERMAX or incomplete Freund adjuvant..
At 12-25 and 25-40 days thereafter, blood samples were collected from the mouse by hip puncture under non-anticoagulant conditions. And the blood coagulates in RT for 1 hour to collect the serum. Titers were determined using TNF! ELA according to known methods. When repeated injections do not lead to increase in titers, fusion was conducted. At this point, 1-400 µg of of TNF! diluted in 100 µL of saline can be injected to the mice for last booster shots. Three days later, the mice were sacrificed by breaking cervical vertebrae, and spleens were removed under sterile conditions, and immersed into 10 mL of cold hydrochloric acid buffered saline (PBS) containing 1000 U/mL penicillin, 100 µg/mL streptomycin, and 0.25 µg/mL amphotericin B (PSA).
Spleen cells were harvested by sterile perfusing the spleen with PSA-PBS, and counted by trypan blue dye exclusion method, and resuspended in RPMI 1640 medium containing 25 mM Hepes. 2) Serum Test A plate was coated with 2 µg/mL of TNF! in PBS overnight. After washed with 0.15 M saline containing 0.02% (v / v) Tween 20, each well was blocked for 1 hour using 1% (w / v) BSA in PBS, 200 µL/well. at RT. The plate was immediately used or frozen at -20 for future use. 50 µL/well of mouse serum dilutions were incubated at RT for 1 hour on the TNF! coated plate. The plate was washed, and then monitored at RT for 1 hour with of 1:30000 specific diluted 50 µL/well of HRP labeled IgG Fc probe in 1% BSA-PBS, washed again, added 100 µL/well of citrate-phosphate substrate solution (0.1 M citric acid and 0.2 M sodium sulfate, 0.01% H O and 1 mg/mL OPD) at RT for 15 minutes, then added 25 µL/well termination solution (4N sulfuric acid). And OD values were read at 490 nm with automatic plate spectrophotometric photometer. 3) Cell Fusion The live spleen cells identified with a high level of anti-hTNF! antibody in serum in the serum test were fused with mouse myeloma cells at the ratio of 1:1 to 10:1.
As a non-restrictive example, the spleen cells and myeloma cells were co-precipitation, and resuspended for 30 seconds or more at 37 in 1 mL of 50% (w/v) of PEG/PBS solution (PEG molecular weight 1450, sigma). In order to terminate the fusion, 10.5 mL of RPMI 1640 medium containing 25 mM Hepes (37 °C) for 1 minutes or more. The fused cells were centrifuged at 500-1500 rpm for 5 minutes, and then were resuspended in HAT medium (RPMI 1640 medium containing 25 mM Hepes, 10% Fetal the Clone I serum (Hyclone), 1 mM sodium pyruvate, 4 mM L-glutamine, 10 µg/mL of celebration Trappe factors, 2.5% Origen culture supplement (Fisher), 10% of 653 adjusted RPMI 1640/Hepes medium, 50 µM 2-mercaptoethanol, 100 µM hypoxanthine and 16 µM thymidine), and were seeded on 15 pieces of 96-well flat-bottom plates with 200 µL/well. And then the plates were placed in a 37 °C incubator with 5% CO and 95% humility for 7-10 days.
Example 2: Qualitative Test of Anti-hTNF! Murine Antibodies Two kinds of qualitative tests are available for anti-hTNF! antibody. In one test, the antibody and Humira are competitive in binding hTNF!, and the competition is determined. In the other test, the ability of the antibody to neutralize hTNF! is determined in the assay of determining L929 cell toxicity. Following, these two methods and experimental results are described, respectively. 1) Competitive binding assay against humira The anti-hTNF! human antibody humira marked with horseradish peroxidase (HRP) was used as a reagent. Elisa plates were coated with rhTNF (50 µl of 0.05 g/mL), and left overnight at room temperature. The coating solution was discarded, and each well was blocked with 1% skim milk in phosphate buffered saline (PBS) for about 0.5 hours, and washed with PBS containing 0.05% Tween 20. And then a mixture of 50 µl of growth medium and 50 µl of HRP labeled humira was added to each well. Unlabeled humira and antibody-free medium were used as positive and negative controls. The method can screen out mouse monoclonal antibody that can highly inhibit the binding of HRP-labeled humira to rhTNF!. The wells in which the binding of HRP-labeled humira and rhTNF! was inhibited Kong were amplified and subcloned, followed by analysis of several mouse monoclonal antibodies which show the inhibition effects, and finally hybridoma cells were screened out. The hybridoma cells were intermediate cultured, and the supernatant was taken for purification, and the mouse monoclonal antibody TM212 and TM23. The purified mouse monoclonal antibodies TM212 and TM23 were tested in competition binding assays. Even at the concentration up to 1 µg/mL, the mouse antibody TM23 only competed out about 50% of the binding of humira to hTNF!. Another mouse antibodies TM212 showed as good competitiveness as unlabeled humira, since at the concentration of about 0.05 µg/mL (equivalent to 3 ( 10 M), it can compete out about 50% of the binding of humira to hTNF!. 2) Qualitative test of anti-hTNF! murine antibodies: determination of in vitro activity to neutralize hTNF! The biological activities of both anti-hTNF! mouse antibody and chimeric antibody to neutralize hTNF! biological activity can be measured using the L929 cytotoxicity assay as described below. Each well of a 96-well culture plate was injected 7.5(10 of L929 cells (ATCC) (10 /mL, 75 µl), and was placed in a 37 °C, 5% CO incubator for 24 hours. The growth medium for L929 cell was RPMI-1640 containing % fetal bovine serum (GIBCO). Using another 96-well culture plate, the solution containing anti-hTNF! antibody was 1/2 serially diluted with RPMI growth medium, and rhTNF! was added to a final concentration of rhTNF! of 5 ng/mL in each sample well.
After the plate containing mixtures was placed in a 37 °C, 5% CO incubator for 2 hours, the mixture of antibody and rhTNF! was added to the L929 cells well. In each line, the concentrations of antibody in each well were in the order of 0.001 to 2 µg/mL. The culture plate was placed in a 37 °C, 5% CO incubator. When surviving cells were measured 3 days later, 20 µl of PBS containing 2.5 mg/mL 3-(4,4-dimethyl-thiazole yl)-2,5-diphenyl-tetrazole bromide salt (MTT; purchased from Sigma Biochemicals) was added, and incubated at 37 °C for 4 hours, and then 100 µl of 0.01 N HCl containing % sodium dodecyl sulfate (SDS) was added overnight. Subsequently, 540/690 nm optical density was tested for each well. The curve for the degree of optical density and antibody concentration was plotted. IC were obtained by analyzing the binding curve, wherein, IC50 means a concentration at which 50% of the rhTNF! toxicity to L929 cells can be neutralized. Therefore, IC50 values can be used to compare the abilities of antibodies to inhibit hTNF! cell toxicity. The IC50 values of several anti-hTNF! murine antibody (TM212, TM220, TM22) and of humira were all in the range of 0.01 to 0.04 µg mL, which means they all have similar capabilities to neutralize L929 cytotoxic caused by rhTNF!. The anti-hTNF! mouse antibody TM212 was selected for further preparation of the chimeric antibody.
Example 3: Cloning of TM212 murine antibody heavy chain and light chain 1) Cloning of TM212 murine antibody heavy chain variable region To design the humanization of murine antibody, DNA fragments containing the murine antibody TM212 heavy chain and light chain variable region encoding sequences must be obtained at the beginning. RNA was isolated from TM2- 11-12 mouse hybridoma cells with RNA purification kit (Invitrogen Corp.), in order to prepare cDNA (GeneRacer kit, Invitrogen, Corp.). By polymerase chain reaction (PCR), using 5' primer (5'-CGACTGGAGCACGAGGACACTGA-3', SEQ ID NO: 24) and 3' primer (5'-TCCAGGGGCCAGTGGATAGAGAGA-3', SEQ ID NO: 25), heavy chain variable region DNA fragment was isolated from cDNA. 3' primer is homologous antisense to the mouse IgG1 heavy chain constant region. Those obtained DNA fragments were cloned into TOPO TA vector (Invitrogen) and sequenced. The heavy chain variable region amino acid sequence is SEQ ID NO: 34: QVQLVQSGPELKKPGETVKISCKASGYTFTHYGMHWVKQTPGRSLKWVGWINTYTG EPTYDADFQGRFTFSLETSTSTAFLQINTLKDEDLATYFCARYDFDGFDYWGQGTTLT VSS. The amino acid sequences in complementary determining regions are CDR-H1 (SEQ ID NO: 3), CDR-H2 (SEQ ID NO: 4) and CDR-H3 (SEQ ID NO: 5). The definition for complementary determining regions can be found in Kabat E. et al. Sequences of Proteins of Immunological Interest, 5th Edition U.S. Department of Health and Human Services, NIH Publication No.91-3242. 2) Cloning of TM212 murine antibody light chain variable region Similar to the PCR method of cloning the heavy chain variable region, using SEQ ID NO: 29 as 5' primer and another 3' primer which is homologous antisense to the mouse immunoglobulin # light chain constant region (5'- CACTGGATGGTGGGAAGATGGATA-3', SEQ ID NO: 26), light chain variable region DNA fragment was isolated from the cDNA. The obtained DNA fragments were cloned into TOPO TA vector and sequenced. Two types of clones were found. About 3/4 of the clones show that part of the nucleotide sequence cannot be translated into readable amino acid sequence (sequences not shown). Such clones were mutated light chain messenger RNA, which cannot encode functional antibody light chain protein. The remaining about 1/4 of clones shows the nucleotide sequence which can be completely translated into readable amino acid sequence. Such clones are derived from the functional light chain messenger RNA. Those obtained DNA fragments were cloned into the TOPO TA vector (Invitrogen) and sequenced. The light chain variable region is amino acid sequence SEQ ID NO: 35: ENVLTQSPPIMSASPGEEVTMTCRASSSITFNYLHWYQQKSGDSPKVWIYSTSNLVSG VPARFSGSGSGTSYSLTISSVEAEDAATYYCQQYSDYPYTFGGGTKLEIK. The amino acid sequences in complementary determining region are CDR-L1 (SEQ ID NO: 6), CDR-L2 (SEQ ID NO: 7) and CDR-L3 (SEQ ID NO: 8). The amino acid sequence was used for designing humanized light chain.
Example 4: humanization design of heavy chain and light chain variable region In order to retain the antigen binding activity, all the amino acid sequences within light chain and heavy chain hypervariable regions are still the same as those of TM212 murine antibody during the humanizing process. The humanization design includes changing amino acid residues within framework regions in accordance with the sequences of human antibody, designing heavy chain variable region and light chain variable region of humanized antibody with a variety of modification, and directed mutating oligonucleotides sites of the antibody heavy chain and light chain variable region sequence by computer simulation technology, so as to increase antibody binding affinity or reduce antibody immunogenicity.
For the humanized antibody heavy chain variable region (SEQ ID NO: 1), within framework region FR-H1, A can be substituted by E at amino acid residue position 16, S can be substituted by T at position 17, I can be substituted by V at position 20; within FR-H2, K can be substituted by R at position 3, G can be substituted by S at position 9; within FR-H3, T can be substituted by V at position 3, E can be substituted by D at position 7, V can be substituted by T at position 10, F can be substituted by Y at position 14, S can be substituted by T at position 19, T can be substituted by V at position 27.
For humanized antibody light chain variable region (SEQ ID NO: 2), within framework region FR-L1, L can be substituted by M at position 11, R can be substituted by E at position 18, M can be substituted by I at position 21; within FR-L2, W can be by L at position 13; within FR-L3, S can be substituted by A at position 4, L can be substituted by V at position 22, A can be substituted by F at position 27.
By introducing at least one of the aforesaid amino acid modifications, a variety of amino acid sequences of several humanized antibody heavy and light chain variable regions are designed. Several amino acid sequences of humanized antibody heavy chain variable region VH and VL are shown in Table 1: Table 1: amino acid sequences of humanized antibody heavy chain variable region VH and light chain variable region VL No. Amino Acid Sequence SEQ ID NO: E001V QVQLVQSGPELKKPGASVKISCKASGYTFTHYGM HWVKQTPGRGLKWVGWINTYTGEPTYDADFQGR FTFSLETSVSTAFLQINSLKDEDLATYFCARYDFDG FDYWGQGTTLTVSS E002V QVQLVQSGPELKKPGASVKISCKASGYTFTHYGM HWVKQTPGRGLKWVGWINTYTGEPTYDADFQGR FTFSLDTSVSTAFLQINSLKDEDLAVYFCARYDFD GFDYWGQGTTLTVSS E003V QVQLVQSGPELKKPGESVKISCKASGYTFTHYGM HWVKQTPGRGLKWVGWINTYTGEPTYDADFQGR FTFSLETSTSTAYLQINSLKDEDLATYFCARYDFDG FDYWGQGTTLTVSS E004V QVQLVQSGPELKKPGATVKVSCKASGYTFTHYGM HWVKQTPGRSLKWVGWINTYTGEPTYDADFQGR FTFSLETSVSTAFLQINTLKDEDLATYFCARYDFDG FDYWGQGTTLTVSS E005V QVQLVQSGPELKKPGASVKVSCKASGYTFTHYGM HWVRQTPGRGLKWVGWINTYTGEPTYDADFQGR FTFSLETSVSTAYLQINSLKDEDLATYFCARYDFDG FDYWGQGTTLTVSS F001V ENVLTQSPPILSASPGERVTMTCRASSSITFNYLH WYQQKSGDSPKVWIYSTSNLVSGVPSRFSGSGS GTSYSLTISSLEAEDAATYYCQQYSDYPYTFGGGT KLEIK F002V ENVLTQSPPILSASPGEEVTMTCRASSSITFNYLH WYQQKSGDSPKVWIYSTSNLVSGVPARFSGSGS GTSYSLTISSLEAEDFATYYCQQYSDYPYTFGGGT KLEIK F003V ENVLTQSPPIMSASPGERVTMTCRASSSITFNYLH WYQQKSGDSPKVLIYSTSNLVSGVPSRFSGSGSG TSYSLTISSVEAEDAATYYCQQYSDYPYTFGGGTK LEIK F004V ENVLTQSPPILSASPGERVTLTCRASSSITFNYLHW YQQKSGDSPKVWIYSTSNLVSGVPARFSGSGSGT SYSLTISSLEAEDAATYYCQQYSDYPYTFGGGTKL Example 5: Construction of pHu_anti-H1L1-of TNF! humanized antibody expression vector 1) Construction of gene for humanized antibody light chain First, the gene fragment for humanized antibody light chain variable region (F001VL) is prepared by synthetic method. And the preparation procedures include obtaining the nucleotides sequences by reverse translation from the amino acid sequence of the light chain variable region according to their genetic codons; adding a Kozak sequence and light chain leader sequence at 5' terminal; preparing the gene fragment for light chain variable region by synthetic method. The gene fragment was cloned into appropriate vectors to obtain pHu-VL1 plasmid. Subsequently, 5’ fragment is obtained by using a pHu-VL plasmid as template, 5' primer FVHX (SEQ ID NO: 17) and 3' primer VKCKO (SEQ ID NO: 21), which comprises the gene for humanized antibody light chain variable region (V ) and 7 amino acid at 5'-teminal of human # light chain constant region (C ). Meanwhile, A gene comprising human # light chain constant region (C ) encoding sequence is obtained from RNA prepared from human leucocyte by using 5' primer HuCKF (SEQ ID NO: 22) and 3' primer HUCKB (SEQ ID NO: 23) through reverse transcription and PCR. Finally, the fragment of humanized antibody light chain variable region and human C gene are linked by PCR using 5' primer (FVHX, SEQ ID NO: 17) and 3' primer (HUCKB, SEQ ID NO: 23) to obtain a gene fragment of length about 700 bp comprising light chain encoding sequence. The gene fragment is treated with endonuclease Hind III and Bam H1, and then inserted into vectors such as PUC19 (ref: Yanisch-Perron, C., Vieira, J. and Messing, J. (1985) Gene, 33, 103-119.). 2) Construction of gene for humanized antibody heavy chain First, the gene fragment for humanized antibody heavy chain variable region (E001VH) is prepared by synthetic method. And the preparation procedures include obtaining the nucleotides sequences by reverse translation from the amino acid sequence of the heavy chain variable region according to their genetic codons; adding a Kozak sequence and heavy chain leader sequence at 5' terminal; preparing the gene fragment for heavy chain variable region by synthetic method. The gene fragment was cloned into appropriate vectors to obtain pHu-VH1 plasmid. Subsequently, 5’ fragment is obtained by using a plasmid (pHu-V ) comprising humanized antibody heavy chain variable region V gene fragment as template, 5' primer FVHX (SEQ ID NO: 17) and 3' primer RVCG (SEQ ID NO: 18), which comprises the gene for humanized antibody heavy chain variable region (V ) and 7 amino acid at 5'-teminal of human IgG heavy chain constant region (C ). Meanwhile, A gene comprising human IgG heavy chain %1 1 constant region (C ) encoding sequence is obtained from RNA prepared from human leucocyte by using 5' primer HuCGF (SEQ ID NO: 19) and 3' primer HUCGE (SEQ ID NO: 20) through reverse transcription and PCR. Finally, the fragment of humanized antibody heavy chain variable region and human C gene are linked by PCR using 5' primer (FVHX, SEQ ID NO: 17) and 3' primer (HUCGE, SEQ ID NO: 20) to obtain a gene fragment of length about 1400 bp comprising heavy chain encoding sequence.
The gene fragment is treated with endonuclease Hind III and EcoR1, and then inserted into vectors such as PUC19 (ref: Yanisch-Perron, C., Vieira, J. and Messing, J. (1985) Gene, 33, 103-119) to express humanized antibody heavy chain protein. The sequence of the gene fragment has been verified to be correct by DNA sequencing. 3) Humanized antibody single-chain expression vector The cDNAs encoding the heavy chain or light chain which are obtained by aforesaid methods are inserted into pcDNA3 (Purchased from Invitrogen USA, Carlsbad, CA, U.S.A.) vector to construct pHu_anti-H1L1-TNF! humanized expression vector. The expression vector plasmid comprises cytomegalovirus early gene promoter-enhancer required for high level expression in mammal cells. Meanwhile, the vector plasmid also comprises optional maker gene, so as to have amicillin resistance in bacteria, have G418 resistance in mammal cells. Furthermore, the vector plasmid comprises DHFR gene. In suitable host cells, chimeric antibody gene and DHFR gene can be co-amplified by Methotrexate (MTX, Sigma) (see, for example, Axel, R., et al.
U.S Patent No. 5,179,017; Kaufman,R. and Sharp,P., J.Mol. Biol. 159:601-621,1982).
Example 6: The expression of humanized antibodies The constructed recombinant expression plasmid was transfected into mammalian host cells to express anti-hTNF! humanized antibody. In order to stabilize high level of expression, the preferred host cells are dihydrofolate reductase (DHFR) deficient Chinese hamster ovary (CHO) cells (see, for example, Chasind, L., et al, U.S.
Patent 4,818,679). The preferred method of transfection is electroporation, and other methods also can be used including calcium phosphate coprecipitation, lipid transfection and protoplast fusion. In electroporation, Gene Pulser (Bio-Rad Laboratories) set at 250V electric field and 960 µFd capacitor is used, 2(10 cells suspended in 0.8 mL of PBS is added into a cuvette, which also contains 10µg expression plasmid DNA linearized by using PvuI (TakaRa). After transfection for 2 days, 0.2 mg/mL of G418 and 200 nM methotrexate (methotrexate or MTX) are added.
In order to achieve a high level of expression, the transfected humanized antibody gene is co-amplified by using DHFR gene inhibited by MTX drugs. The subcloning transfectants are diluted, and the secretion rates of various cell lines are determined to screen out cell lines with high-level expression of humanized antibody.
Example 7: Study on the antibodies neutralizing TNF! killing effect on L929 cell L929 cells were trypsinized, centrifuged, resuspended in 1640 medium supplemented with 10% FCS, counted, and added to column 1 to 11 of 96-well plated at a certain concentration. And then, an appropriate concentration of TNF! was added to column 1 to 10 in 96-well plates. Respectively, 2-fold gradient dilutions of adalimumab (purchased from Abbott Laboratories), amino acid unmodified chimeric antibody AT(CE)-1, AT132, AT135, AT143, AT151, AT164 prepared in accordance with Examples 5 and 6 were added to row A, B, C, D, E, F, G and H, in a serial of concentrations from low to high (column 1 to 9). And column 10 is TNF! control, column 11 is cell control, and column 12 is medium control. After addition, the plate was placed in a 37 °C carbon dioxide incubator to cultivate. After incubation was completed, color reagent was added, and continued to incubate. And then absorbance was detected by microplate reader. The results were shown in Table 2.
Table 2: the antibodies neutralize TNF! killing effect on L929 cell Antibody Type Heavy Chain Light Chain EC (ng/mL) AT132 E001V F001V 20.4 AT135 E002V F001V 50 AT143 E003V F003V 39.8 AT151 E001V F002V 42.1 AT164 E002V F003V 45.5 AT(CE)-1 -- -- 15.2 Humira -- -- 19.1 The results in Table 2 show that humanized antibodies obtained by mutation in FR region still have good activities in neutralizing TNF!. The EC50 of AT132, 20.4 ng/mL, is similar to that of Humira.
Example 8: Study on AT132 neutralizing TNF! killing effect on human U937 cells (human lymphoma) U937 cells in good conditions were counted and adjusted to cell concentration of 3.75 ( 10 /well with 1640 medium with 10% FCS. And then were added to a 96-well plate, 75 µl/well. The cell culture medium containing 120 ng/mL of TNF! was used to respectively gradient dilute AT132 standard sample and test samples. The concentration in first well was 600 ng/mL, and dilution gradient was 1.5 fold. After dilutions, the samples were added to 96-well plate with 25 µl/well. The plate was incubated for 40 hours in a 37 °C carbon dioxide incubator. After completion of the incubation, each well was added 10 µl of CCK8 color reagent, and incubated for 3h, detected with microplate reader at 490nm/630nm dual-wavelength. A four-parameter curve fitting was performed to obtain ED50 of the standard sample and test samples, and calculate the specific activity (formula: 100% (ED50 of standard sample / ED50 of test samples).
Figure 1 shows the curve of antibody neutralizing TNF! killing effects of U937 cells The analysis results of figure 1 shows that, at extremely low concentrations of AT132, TNF! kills cells. With the concentration of AT132 increases, TNF! killing effects are gradually antagonized. When the concentration of AT132 reaches about 80 ng/mL, TNF! killing effects are completely antagonized. Clearly, it is dose-dependent according to the results of several experiments. The average median effective concentration of AT132 to neutralize 30 ng/mL of TNF! is 24.1 ng/mL.
Example 9: Determination of AT132 affinity AT132 affinity was determined by Biacore X100, and analyzed with Biacore X100 kinetics/affinity analysis software. Using indirect capture method, goat anti-human IgG Fc polyclonal antibody was coupled to the surface of CM5 chip as a capture molecule by using Amine Coupling Kit. By calculation, AT132 and Humira as control were respectively diluted to a a certain concentration to be lately used as ligand, and TNF! as the analyte. Analytes were diluted to 5 concentrations, and each concentration as a cycle. First, using HBS-EP buffer to run for three cycles, designing an analyte concentration of 0 concentration to run for two cycles, and finally, designing a repeat analyte concentration to run for a cycle. The whole process ran 11 cycles, and each cycle can draw a curve. The dynamics/affinity data of the measured antibody and humira were analyzed by Biacore X100 kinetics/affinity analysis software.
Results: AT132 dissociation constant (Kd) of 1.19 ( 10 M, that is, the -1 -10 affinity constant (Ka) of 8.4 ( 10 M ; Humira dissociation constant of 1.08 ( 10 M, 9 -1 that is, the affinity constant 9.3 ( 10 M .
Example 10: AT132 binding activity to mouse TNF! and monkey TNF! On a plate, respectively coat recombinant human TNF! 5 ng/well, mouse TNF! 25 ng/well, and monkey TNF! 5 ng/well at room temperature, and blocked for 1h, washed. And then 1.8-fold gradient dilutions of AT132 was added with an initial concentration of 250 ng/mL, and incubated at 25 °C for 2h, washed, HRP-labeled anti- human Fc antibody added, and incubated at room temperature for 1h, washed, and the substrate solution added as 100 µl/well, placed in dark at 37 °C for 30 min. In accordance with the order of the added color reagents, 0.2 M H SO , 50 µl/well was added to terminate the reaction. Within about 5 minutes after termination, OD values were test at 450 nm/630 nm on a microplate reader, and median effective concentrations were obtained by four-parameter fitting. Compare the binding activities of AT132 to different sources of TNF!.
Mouse TNF! binding activity results show that AT132 does not bind mouse TNF!. Calculation based on coating concentration and AT132 sample concentration suggests that AT132 binding activity to mouse TNF! is at least 1,000 times lower than that to human TNF!.
Monkey TNF! binding activity results show AT132 binding activity to monkey TNF! was about 50% of that to human TNF!.
Example 11: Preparation of injections The preparation of AT132, AT135 injection preparation is as follows: 1) Preparation of 20-L buffer (equivalent to 20.180 kg solution with a density: 1.009 g / mL) Weighing out the ingredients in the following weight: 240.0 g mannitol, 26.1 g monohydrate citric acid, 6.1 g sodium citrate, 30.6 g dihydrated disodium hydrogen phosphate, 17.2 g dihydrate phosphate monobasic sodium, 123.3 g sodium chloride, 20.0 g sorbitan polyoxyethylene(20) ether oleate and 19715.7 to 19716.1 g water.
Mixing 40.0 g sodium hydroxide and 1000.8 g water for injection to prepare sodium hydroxide solution.
Then, dissolving following pre-weighted ingredients (as described above) in about 90% water for injection to prepare buffer: mannitol monohydrate citric acid, sodium citrate dihydrate disodium hydrogen phosphate, ether oleate, sodium dihydrogen phosphate, sodium chloride and sorbitan polyethylene(20) ether oleate.
After adding all of the above buffer ingredients, adjusting pH of the solution with 1 M sodium hydroxide prepared by the above method. After adding sodium hydroxide, adding the final weight of water. And then, The buffer is filtered into a sterile container through a filter membrane (hydrophilic poly(vinylidene fluoride), 0.22 µm pore size). The filter media used in the filtration is ammonia for disinfection. 2) Preparation of 40 L formulation (equivalent to 40.88 kg) Filtered buffer solution was added to antibody concentrate (the active ingredient of the pharmaceutical formulation), which has been thawed and merged according to the following methods. Before the preparation of pharmaceutical formulation, the antibody (concentrate) was placed in a water bath to thaw. 34.207 g antibody concentrate was used, which was equivalent to 2.0 kg of protein, protein concentrate with a concentration of 60 mg protein/mL of. The density of the concentrate was 1.0262 g/mL. Any protein concentrate within 25.655-37.316 can be used, which is equivalent to a concentration of 50-80 mg/mL protein in the protein concentrate. Under stirring, the buffer was added until it reaches the final weight of the total solution.
Then, the formulation which comprising all its ingredients was filtered in accordance with the above method except that the formulation was filter by two layers of sterile 0.22 µm membrane filter. After disinfection, the formulation was packed for use in a vial or pre-filled syringe.
Example 12: AT132 acute toxicity test Test samples: AT132 lyophilized powder, 20 mg/bottle; adjuvant control: AT132 buffer (containing histidine, trehalose); solvent: sterilized water for injection.
Test animal grouping and dose: amount of 60 Kunming (KM) mice, 4-6 weeks old, 18- 22g weight, half male and half female, the SPF level. The animals were randomly divided into three groups, each group of 20, half male and half female. Single dosed and observed for 14 days. Table 3 shows the dose and route of administration: Table 3: dose and route of administration Dose in weight Dose in volume No. Group (mg/kg) (mL/kg) 1 Adjuvant control — — 2 Test sample, subcutaneous 500 25 injections Test sample, intravenous 3 500 25 injections Outcome measures: body weight, food intake, mental state, behavior, stool. After the end of the trial observation period, the animals were put euthanasia and pathology gross anatomical observed. And abnormal tissues or organs were histologically examined.
Results: During the trial, no animal died or was dying; all animals were in goold mental state, normal behavior, diet, water drinking, and showed no abnormal performance. The weights of each treatment group animals showed no regular change associated with the administration of dose. Pathology gross anatomy observation showed no abnormal changes related to the administration of dose.
Conclusion: under the present experimental condition, when AT132 powder was administrated to mice by intravenous and subcutaneous injection with a single injection of 500 mg/kg dose, no significant toxicity was observed, and the maximum tolerated dose (MTD) was greater than 500 mg/kg.
Example 13: AT132 protective effect on D-galactosamine sensitized mouse from rhTNF!-induced death An amount of 51 C57BL/6 mic, weight 20.0 ± 2.0 g, divided into 6 groups (Table 4). For group 2 to 5, each mouse was intraperitoneally injected 0.25 mL of AT132 solution, wherein, based on the amount of AT132, each mouse in group 2 was given a dose of 5.2 µg/mouse; in group 3, 26 µg/mouse; in group 4, 52 µg/mouse; in group 5, 26 µg/mouse. For group 1, each mouse was intraperitoneally injected 0.25 mL pH 5.63 citrate buffer. For group 6, each mouse was intraperitoneally injected 0.25 mL of human IgG1 (HuIgG1, negative control), 26 µg/mouse based on the amount of HuIgG1. 30 minutes later, every mouse in each group (except group 5) was intraperitoneally injected 0.25 mL mixture solution of rhTNF! (Primegene, batch number 1030109021) and D-galactosamine. For group 5, each mouse was intraperitoneally injected 0.25 mL of buffer. Observed the number of died mice in 48 hours, and calculated survival rates as shown in Table 4.
Table 4: The survival rates of mice in each group NO. Group n Survival/Total Survival Rate (%) 1 Buffer 9 0/9 0 2 5.2 µg AT132 9 3/9 33.33 3 26 µg AT132 9 9/9 100 4 52 µg AT132 10 10/10 100 26 µg AT132 7 7/7 100 (No rhTNF) 6 26µg HuIgG 7 0/7 0 Results: mice survival rates of buffer and HuIgG1 group were 0. But AT132 protected rhTNF! and D-amino-galactose sensitized mice in dose dependent manner. Therefore, AT132 showed protective effects on D-galactosamine sensitized mice from rhTNF!-induced death.
Example 14: Study on the effect of AT132 on type II collagen- induced arthritis An amount of 40 Wistar female rats were randomly divided into 4 groups, and each group is 10 rats. The groups are blank control group, inflammatory control group (model group), AT132 1 mg/kg group, and AT132 5 mg/kg group.
Except for the blank control group, the rats in the other groups were respectively intradermally injected on the back with collagen II immune. And the proinflammatory control group was injected with 0.1 mol/L acetic acid and complete or incomplete Freund's adjuvant emulsion (Sigma, lot number 129K8701) .
AT132 1 mg/kg group and AT132 5 mg/kg group began to be intraperitoneal injected with AT132 at the day of immunization. One week after the first immunization, the mice were immunized again in the same way to strengthen the immune, and were administrated for a total of 28 days. At different times before and after administration, rat joint swelling values were measured by YLS-7B toe volume measuring instrument. The joint was taken for pathological examination after the experiments. The swelling rate and inhibition rate were calculated, and the differences between groups were compared by t test. Calculated as follows: E = swelling values at different times after inflammation, E = swelling values before inflammation The experimental data were represented in mean values and standard deviation (s), and t test was used for statistical analysis. The results are shown in Figure 2. AT132 1 mg/kg group showed significant inhibitory effects, with a good effect starting from day 19, and the best effect is 63.10%. Since then, the inhibitory effect gradually decreased. The result is still good at day 28 with an inhibition rate of 55.71%.
AT132 5 mg/kg group showed significant inhibitory effects with significant treating effect starting from day 19. The rat paw joint swelling gradually subsided, and activities became normal, and the effect lasted until day 28 days. The highest inhibition rate of 47.27% appeared at day 21, and the other were about 40%.
Example 15: Pharmacodynamic Study of AT132 on Tg197 mouse arthritis model The experiment employed Tg197 transgenic mice (purchased from Cyagen Biosciences), and the mice were divided into 6 groups, each group of 10 mice, half male and half female. Specifically as follows: Group 1: AT132 1 mg/kg; Group 2: solvent group (a buffer containing citric acid and sodium chloride); Group 3: AT132 30 mg/kg,; Group 4: AT132 10 mg/kg); Group 5: Humira 10 mg/kg; Group 6: AT132 3 mg/kg. Another 4 Tg197 mice were selected as blank control group. 3 weeks old Tg197 mice were intraperitoneally injected AT132, twice a week, until 10 weeks old. AT132 was diluted to the desired concentration prior to administration, and each group was given a dose of 10 µl/g weight. Observe the degree of mice arthritis, and evaluate pathological scores of mice bare joint, and calculate the inhibition rate based on arthritis scores and pathological scores. 1) Study of AT132 on Tg197 mouse arthritis degree Mice joint morphological changes were evaluated weekly to assess the degree of arthritis, and the specific arthritis scoring criteria as follows: 0.0 = no arthritis (appearance normal, the mice are able to support the body weight, overall flexibility/evade capability normal, maximum grip strength); 0.5 = onset of arthritis (joints and paws slight swelling, appearance normal, the mice are able to support the body weight, overall flexibility/evade capability normal, maximum grip strength); 1.0 = mild arthritis (joints swelling and deformation, paws swelling, appearance normal, the mice are able to support the body weight, overall flexibility/evade capability normal, maximum grip strength); 1.5 = mild to moderate arthritis (joints and paws swelling, deformation, and the last fingers inward deformation, barely able to support the weight, overall flexibility reduced, grip strength decreased); 2.0 = moderate arthritis (severe joints, paws and fingers swelling, feet joints deformation, cannot support the upper body weight and fall, disappearance of the overall flexibility, grip strength disappeared, crawling/eating affected); 2.5 = moderate to severe arthritis (severe joints, paws and fingers swelling, feet joints deformation, cannot support the upper body weight and fall, disappearance of the overall flexibility, grip strength disappeared, crawling/eating affected, finger paws deformation, mouse activity impaired); 3.0 = severe arthritis (joint stiffness, bending detected and activity severely impared, the mice are dying).
Tg197 mice arthritis scores are shown below in Figure 3A. 2) Tg197 mice ankle joint histopathology study To monitor the disease status, four littermates of Tg197 mice of the trial (Numbered as Con1 to Con4) were sacrificed when 3 weeks old, as joint samples at the beginning of treatment. Experimental mice were sacrificed when 10 weeks old, and sample slices were taken from ankle joints. After hematoxylin/eosin staining, microscopic histopathological scores of arthritis were evaluated in a blinded manner, and the score of 0-4 as follows: 0 = no detectable pathology 1 = synovial proliferation, polymorphonuclear leukocytes infiltration 2 = pannus and fibrous tissue formation and subchondral bone erosion at focus site 3 = cartilage destruction and bone erosion 4 = expanded cartilage destruction and bone erosion Tg197 mice histopathological analysis were shown in Figure 3B and 3C 3) Calculation of inhibition rates according to the arthritis and pathological scores.
Inhibition rates were calculated according to the arthritis scores as follows: En = arthritis score of each group E0 = arthritis score of solvent group Inhibition rates were calculated according to the pathological score as follows: E = pathological score of each group E = pathological scores of solvent group The results are shown in Table 5 below.
Table 5: mice arthritis score inhibition rate and pathological score inhibition rate No. Group n arthritis score pathological inhibition rate (%) score inhibition rate Group 1 AT132 1 mg/kg 10 15 3 Group 2 Solvent 10 0 0 Group 3 AT132 30 mg/kg 10 77 84 Group 4 AT132 10 mg/kg 10 79 86 Group 5 Humira 10 mg/kg 10 65 76 Group 6 AT132 3 mg/kg 10 46 41 Results: Administrated with different AT132 doses (group 1, group 3, group 4, group 6), it shows significant inhibition effects on rheumatoid arthritis, which is typically dose dependent. Especially from group 1 (1 mg/kg) to group 6 (3 mg/kg), the inhibitions can be significantly distinguished therebetween. From group 6 (3 mg/kg) to group 4 (10 mg/kg), the inhibitions also can be significantly distinguished therebetween.
Only between group 4 (10 mg/kg) group and the highest dose group 3 (30 mg / kg), the inhibitions have no difference. It is important that at the same dose level of 10 mg/kg, AT132 treatment group has no significant difference from Humira treatment group 5, and have significantly improvement effect on arthritis histopathology when compared to solvent group 2 Example 16: AT132 monoclonal antibody tissue cross-reaction 1) AT132 monoclonal antibody human tissue cross-reaction Nasal polyps paraffin slices and four normal human tissue (donor A, B, C, D, provided by National Institutes for Food and Drug Control, and National Center for Safety Evaluation of Drugs) paraffin slices were divided into three groups, namely the experimental group (AT132 biotin marker), positive control group (biotin-labeled Humira), the negative control (buffer PBS). Observe the staining of tissue slicers after tissue cross-reaction.
The experimental results show that the negative control group of human nasal polyps and normal human tissues were not stained. Biotin-Humira positive control group, the human nasal polyps macrophages, the normal human lymph nodes macrophages and the lung alveolar macrophages were weakly to moderately stained, while the other tissues were not stained. Biotin-AT132 experimental group, the human nasal polyps macrophages showed weak to moderate staining. Normal human tissue cross-reactions were similar to Humira. 2) AT132 monoclonal antibody cynomolgus monkey tissue cross- reaction Nasal polyps paraffin slices and three sets of cynomolgus monkey tissues (donor A, B, C, provided by National Institutes for Food and Drug Control, and National Center for Safety Evaluation of Drugs) paraffin slices were divided into three groups, namely the experimental group (AT132 biotin marker), positive control group (biotin-labeled Humira), the negative control (buffer PBS). Observe the staining of tissue slicers after tissue cross-reaction.
The experimental results show that the negative control group of human nasal polyps and normal cynomolgus monkey tissues were not stained. Biotin-Humira positive control group, the human nasal polyps macrophages were weakly to moderately stained, while the normal cynomolgus monkey tissues were not stained.
Biotin-AT132 experimental group, the human nasal polyps macrophages showed weak to moderate staining. Normal cynomolgus monkey tissue cross-reactions were similar to Humira.

Claims (16)

WHAT IS CLAIMED IS:
1. A humanized anti-TNF monoclonal antibody comprising a heavy chain and a light chain, the heavy chain comprising complementary determining regions (CDRs) CDR-H1, CDR-H2 and CDR-H3, and heavy chain framework regions FR-H1, FR-H2, and FR-H3, the light chain comprising CDRs CDR-L1, CDR-L2, CDR-L3 and light framework regions FR-L1, FR-L2 and FR-L3, wherein CDR-H1 of the heavy chain variable region comprises a sequence of SEQ ID NO: 3; CDR-H2 comprises a sequence of SEQ ID NO: 4; CDR-H3 comprises a sequence of SEQ ID NO: 5; complementary determining region CDR-L1 comprises a sequence of SEQ ID NO: 6; CDR-L2 comprises a sequence of SEQ ID NO: 7; and CDR-L3 comprises a sequence of SEQ ID NO: 8; and wherein one or more of the heavy chain framework regions and light chain framework regions comprises at least one substitution selected from, within the heavy chain framework region FR-H1, I is optionally substituted by V at position 20 (SEQ ID NO: 11); within FR-H2, K is optionally substituted by R at position 3 (SEQ ID NO: 12); within FR-H3, T is optionally substituted by V at position 3, E is optionally substituted by D at position 7, F is optionally substituted by Y at position 14, and T is optionally substituted by V at position 27 (SEQ ID NO: 13); within framework region FR- L1, M is optionally substituted by I at position 21 (SEQ ID NO: 14); within FR-L2, W is optionally substituted by L at position 13 (SEQ ID NO: 15); and within FR-L3, A is optionally substituted by F at position 27 (SEQ ID NO: 16).
2. The humanized anti-TNF monoclonal antibody comprising a heavy chain and a light chain according to claim 1, wherein the heavy chain comprises a sequence of SEQ ID NO: 1, and the light chain comprises a sequence of SEQ ID NO: 2.
3. A nucleic acid sequence which encodes the humanized anti-TNF monoclonal antibody according to claim 1 or 2.
4. A vector comprising the nucleic acid sequence according to claim 3.
5. The vector according to claim 4, wherein the vector further comprises a promoter which is operational linked to the nucleic acid sequence to facilitate its expression.
6. A host cell comprising the vector according to claim 5 which is not present within a human.
7. A diagnostic agent or medicament comprising the humanized anti-TNF monoclonal antibody according to claim 1 or 2.
8. A method for preparing a medicament in diagnostic analysis of hTNFα, comprising including in the medicament an effective amount of the humanized anti-TNF monoclonal antibody according to claim 1 or 2.
9. Use of an effective amount of humanized anti-TNF monoclonal antibody according to claim 1 or 2 in the manufacture of a medicament for treating a hTNF related disease, wherein the hTNFα related disease is selected from pyaemia, autoimmune diseases, malignant tumor, lung function disorder, transplant rejection, bacterial meningitis, cerebral malaria, AIDS and AIDS related complex (ARC), and secondary cytomegalovirus infection after transplantation.
10. A pharmaceutical composition comprising the humanized anti-TNF monoclonal antibody according to claim 1 or 2 and at least one pharmaceutically accepted excipient.
11. The humanized anti-TNF monoclonal antibody according to claim 1 or 2, wherein the at least one substitution is within FR-H1, wherein I is substituted by V at position 20 (SEQ ID NO: 11).
12. The humanized anti-TNF monoclonal antibody according to any one of claim 1, 2 and 11, wherein the at least one substitution is within FR-H2, wherein K is substituted by R at position 3 (SEQ ID NO: 12).
13. The humanized anti-TNF monoclonal antibody according to any one of claim 1, 2, and11-12, wherein the at least one substitution is within FR-H3, wherein T is substituted by V at position 3, E is substituted by D at position 7, F is substituted by Y at position 14 and T is substituted by V at position 27 (SEQ ID NO: 13).
14. The humanized anti-TNF monoclonal antibody according to any one of claim 1, 2 and 11-13, wherein the at least one substitution is within FR-L1, wherein M is substituted by I at position 21 (SEQ ID NO: 14).
15. The humanized anti-TNF monoclonal antibody according to any one of claims 1, 2 and 11-14, wherein the at least one substitution is within FR-L2, wherein W is substituted by L at position 13 (SEQ ID NO: 15).
16. The humanized anti-TNF monoclonal antibody according to any one of claims 1, 2 and 11-15, wherein the at least one substitution is within FR-L3, wherein A is substituted by F at position 27 (SEQ ID NO: 16). 1 / 3
NZ615477A 2011-02-28 2012-02-13 Tumor necrosis factor-? humanized antibody NZ615477B2 (en)

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
CN201110048505 2011-02-28
CN201110048505.1 2011-02-28
CN201210026698.5A CN102675460B (en) 2011-02-28 2012-02-07 The humanized antibody of Tumor necrosis factorα
CN201210026698.5 2012-02-07
PCT/CN2012/071079 WO2012116595A1 (en) 2011-02-28 2012-02-13 Tumor necrosis factor-α humanized antibody

Publications (2)

Publication Number Publication Date
NZ615477A NZ615477A (en) 2015-11-27
NZ615477B2 true NZ615477B2 (en) 2016-03-01

Family

ID=

Similar Documents

Publication Publication Date Title
EP3689909A1 (en) Tigit antibody, antigen-binding fragment thereof, and medical use thereof
JP2023036605A (en) Anti-dll4/vegf dual variable domain immunoglobulin and uses thereof
JP5981347B2 (en) PCSK9 antagonist
RU2314316C2 (en) Antibodies raised to human mcp-1
US8236312B2 (en) VEGF-specific human antibody
AU2012222738B2 (en) Tumor necrosis factor-a humanized antibody
CA2977621C (en) Antibody binding to tfpi and composition comprising the same
US20220185875A1 (en) Bispecific antibody specifically bound to vegf and ang2
US20230159643A1 (en) Agent, uses and methods for treatment
EP3808774A1 (en) Human il-4r binding antibody, antigen binding fragment thereof, and medical use thereof
EP3683234A1 (en) Il-6r antibody and antigen binding fragment thereof and medical use
US9587015B2 (en) Anti-human CTGF antibody
WO2022268178A1 (en) Human epidermal growth factor receptor binding molecule and use thereof
US20240209114A1 (en) Anti-masp2 antibody, antigen-binding fragment thereof and medical use thereof
US8658171B2 (en) Humanized anti-TNFα antibodies
NZ615477B2 (en) Tumor necrosis factor-? humanized antibody
JP7202011B2 (en) Anti-RAMP2 antibody
TW202417509A (en) Slit2 related compositions and methods
EA041171B1 (en) ANTIBODIES TO ONCOSTATIN M RECEPTOR AND THEIR USE