WO1989009622A1 - Il-2 receptor-specific chimeric antibodies - Google Patents

Il-2 receptor-specific chimeric antibodies Download PDF

Info

Publication number
WO1989009622A1
WO1989009622A1 PCT/US1989/001578 US8901578W WO8909622A1 WO 1989009622 A1 WO1989009622 A1 WO 1989009622A1 US 8901578 W US8901578 W US 8901578W WO 8909622 A1 WO8909622 A1 WO 8909622A1
Authority
WO
WIPO (PCT)
Prior art keywords
human
sequence
segment
antibody
mouse
Prior art date
Application number
PCT/US1989/001578
Other languages
French (fr)
Inventor
Cary L. Queen
Original Assignee
Protein Design Labs, 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
Application filed by Protein Design Labs, Inc. filed Critical Protein Design Labs, Inc.
Priority to KR1019890702368A priority Critical patent/KR900700134A/en
Publication of WO1989009622A1 publication Critical patent/WO1989009622A1/en
Priority to FI895955A priority patent/FI895955A0/en
Priority to NO89895022A priority patent/NO895022L/en
Priority to DK631689A priority patent/DK631689A/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2866Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against receptors for cytokines, lymphokines, interferons
    • 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/46Hybrid immunoglobulins
    • C07K16/461Igs containing Ig-regions, -domains or -residues form different species
    • C07K16/462Igs containing a variable region (Fv) from one specie and a constant region (Fc) from another
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • 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
    • C07K2319/00Fusion polypeptide
    • C07K2319/01Fusion polypeptide containing a localisation/targetting motif
    • C07K2319/02Fusion polypeptide containing a localisation/targetting motif containing a signal sequence
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/33Fusion polypeptide fusions for targeting to specific cell types, e.g. tissue specific targeting, targeting of a bacterial subspecies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/55Fusion polypeptide containing a fusion with a toxin, e.g. diphteria toxin

Definitions

  • the present invention relates generally to the combination of recombinant DNA and monoclonal antibody technologies for developing novel therapeutic and other agents and, more particularly, to the production of chimeric antibodies specific for the human interleukin-2 receptor and the use of such chimeric antibodies in treating T-cell mediated human disorders.
  • the immune response is mediated by two types of cells that interact specifically with foreign material, i.e. , antigens.
  • B-cells are responsible for the production of antibodies.
  • T-cells include a wide variety of cellular subsets controlling the in vivo function of both B-cells and a wide variety of other hematopoietic cells, including T- cells.
  • IL-2 interleukin-2
  • T-cell growth factor T-cell growth factor 2
  • IL-2 interacts with a specific high-affinity membrane receptor (Greene, . , et al., Progress in Hematology XIV, E. Brown, ed. , Grune and Statton, New York (1986), at pgs. 283 ff) .
  • the human IL-2 receptor is a complex glycoprotein, with one chain 55kD in size (Leonard, . et al., J. Biol. Che . 260;1872 (1985)).
  • a gene encoding this protein has been isolated, and predicts a 272 a ino acid peptide, including a 21 amino acid signal peptide, (see, Leonard, . et al., Nature 311: 626 (1984)).
  • IL-2 receptors can be detected on T-cells, but also on cells of the onocyte-macrophage family, Kupffer cells of the liver, Langerhans 1 cells of the skin and, of course, activated T-cells.
  • resting T-cells, B-cells or circulating machrophages typically do not display the IL-2 receptor (Herrmann, et al., J. Exp. Med. 162:1111 (1985)).
  • the anti-Tac monoclonal antibody has also been used to define lymphocyte functions that require IL-2 interaction, and has been shown to inhibit various T-cell functions, including the generation of cytotoxic and suppressor T lymphocytes in cell culture. Also, based on studies with anti-Tac, a variety of disorders are now associated with improper IL-2 receptor expression by T-cells, in particular adult T-cell leukemia.
  • the IL-2 receptor has been shown to be an ideal target for novel therapeutic approaches to T-cell mediated diseases.
  • the anti-Tac monoclonal antibody can be used either alone or as an immunoconjugate (e.g. with Ricin A, isotopes and the like) to effectively remove cells bearing the IL-2 receptor.
  • These agents can, for example, theoretically eliminate IL-2 receptor-expressing leukemic cells, certain B-cells, or activated T-cells involved in a disease state, yet allow the retention of mature normal T- cells and their precursors to ensure the capability of mounting a normal T-cell immune response as needed.
  • other T-cell specific agents can destroy essentially all peripheral T-cells, which limits therapeutic efficacy.
  • the use of monoclonal antibodies specific for the IL-2 receptor can be expected to have therapeutic utility in autoimmune diseases, organ transplantation and any unwanted response by activated T-cells. Indeed, clinical trials have been initiated (see, generally, Waldman, T., Science 232:727- 732 (1986) , which is incorporated herein by reference) .
  • the use of the anti-Tac monoclonal antibody has certain drawbacks, particularly in repeated therapeutic regimens. As a mouse monoclonal, it does not fix human complement well, whereas a human equivalent may be more efficient. More importantly, however, anti-Tac monoclonal antibody contains substantial urine amino acid sequences that will be antigenic when injected into a human patient.
  • the present invention provides novel compositions useful in the treatment of T-cell mediated human disorders, the compositions containing a chimeric antibody specifically capable of binding to human IL-2 receptors, such as at the epitope bound by the anti-Tac monoclonal antibody.
  • the IL-2 chimeric antibody can have two pairs of light chain/heavy chain complexes, wherein at least one pair has chains comprising mouse variable regions joined with human constant region segments, with or without naturally-associated J and D segments.
  • the chimeric antibodies, or binding fragments thereof, of the present invention may be produced by a variety of recombinant DNA techniques, with ultimate expression in transfected cells, preferably immortalized eukaryotic cells, such as myeloma or hybridoma cells.
  • Polynucleotides comprising a first sequence coding for a human immunoglobulin constant region and a second sequence coding for the desired mouse immunoglobulin variable or hypervariable region can be produced synthetically or by combining appropriate cDNA and genomic DNA segments.
  • the compounds When the chimeric antibodies are complexed with a cytotoxic agent, such as a radionuclide, a ribosomal inhibiting protein or a cytotoxic agent active at cell surfaces, the compounds will be particularly useful in treating T-cell mediated disorders.
  • a cytotoxic agent such as a radionuclide, a ribosomal inhibiting protein or a cytotoxic agent active at cell surfaces
  • these compounds can be provided in a pharmaceutically accepted dosage form, which will vary depending on the mode of administration.
  • Figure 1 depicts the DNA coding sequence and putative amino acid sequence of the V and J regions of the anti-Tac light chain.
  • Figure 2 depicts the DNA coding sequence and putative amino acid sequence of the V and J regions of the anti-Tac heavy chain.
  • Figures 3-10 are schematic diagrams of the plasmids utilized to demonstrate the present invention.
  • Figure 11 represents an overview of a preferred strategy for preparation of V and J regions for insertion into plasmids.
  • DNA sequences encoding mouse variable/human constant region chimeric antibodies capable of binding epitopes on human IL-2 receptors are provided.
  • large quantities of chimeric antibodies can be produced.
  • the chimeric antibodies will have substantially the same binding profile or characteristics as (e.g. , be cross-reactive or capable of blocking) the binding of the anti-Tac monoclonal antibody, such as antibodies produced by the myeloma cell line deposited with the A.T.C.C. and designated accession number CRL 9688.
  • These chimeric antibodies find use, for example, in the treatment of T-cell mediated disorders in human patients.
  • the basic immunoglobin structural unit is known to comprise a tetramer.
  • Each tetramer is composed of two identical pairs of polypeptide chains, each pair having one "light" (about 25kD) and one "heavy" chain (about 50-70kD) .
  • the NH -terminus of each chain begins a variable region of about 100 to 110 or more amino acids primarily responsible for antigen recognition.
  • the COOH terminus of each chain defines a constant region primarily responsible for effector function.
  • Light chains are classified as either kappa or lambda.
  • Heavy chains are classified (and subclassified) as gamma, mu, alpha, delta, or epsilon, and define the immunoglobulin's isotype as IgG, IgM, IgA, IgD and IgE, respectively.
  • the variable and constant regions are joined by a "J" region of about 12 or more amino acids, with the heavy chain also including a "D” region of about 12 more amino acids.
  • Chimeric antibodies are antibodies whose light and heavy chain genes have been constructed, typically by genetic engineering, from immunoglobulin gene segments belonging to different species.
  • the variable (V) segments of the genes for a mouse monoclonal antibody may be joined to human constant (C) segments, such as 7 , and 7 .
  • a preferred therapeutic chimeric antibody is thus a hybrid protein consisting of the V or antigen-binding domain from a mouse antibody and the C or effector domain from a human antibody, although other mammalian species may be used.
  • Human chimeric antibodies have at least three potential advantages over mouse antibodies for use in human therapy: 1) Because the effector portion is human, it may interact better with the other parts of the human immune system (e.g. , destroy the target cells more efficiently by complement-dependent cytotoxicity (CDC) or antibody-dependent cellular cytotoxicity (ADCC) ) .
  • CDC complement-dependent cytotoxicity
  • ADCC antibody-dependent cellular cytotoxicity
  • the human immune system should not recognize the C region of the chimeric antibody as foreign, and therefore the antibody response against an injected chimeric antibody should be less than against a totally foreign mouse antibody.
  • Injected mouse antibodies have been reported to have a half-life in the human circulation much shorter than the half-life of normal antibodies (Shaw, D. et al., J. Immunol. 138:4534-4538 (1987)) . It is possible that injected chimeric antibodies will have a half-life more like that of human antibodies, allowing smaller and less frequent doses to be given.
  • the present invention is directed to recombinant DNA segments encoding the heavy and/or light chain variable or hypervariable regions from the anti-Tac monoclonal antibody.
  • the DNA segments encoding these regions will typically be joined to DNA segments encoding appropriate constant regions, such as human gamma heavy chain regions or human kappa light chain regions.
  • appropriate constant regions such as human gamma heavy chain regions or human kappa light chain regions.
  • the preferred variable region DNA sequences which on expression code for the polypeptide chains comprising the anti-Tac light and heavy chain variable regions (with naturally-associated J regions) , are shown in Figures 1 and 2, respectively. Due to codon degeneracy and non-critical amino-acid substitutions, other DNA sequences can be readily substituted for those sequences, as detailed below.
  • the DNA segments will typically further include an expression control DNA sequence operably linked to the chimeric antibody coding sequences, including naturally- associated or heterologous promoter regions.
  • the expression control sequences will be eukaryotic promoter systems in vectors capable of transforming or transfecting eukaryotic host cells. Once the vector has been incorporated into the appropriate host, the host is maintained under conditions suitable for high level expression of the nucleotide sequences, and, as desired, the collection and purification of the light chains, heavy chains, light/heavy chain dimers or intact chimeric antibodies may follow.
  • variable and constant regions are subject to substantial natural modification, yet are “substantially identical” and still capable of retaining their respective activities.
  • Human constant region DNA sequences can be isolated in accordance with well known procedures from a variety of human cells, but preferably immortalized B-cells. Suitable source cells for the DNA sequences and host cells for expression and secretion can be obtained from a number of sources, such as the American Type Culture Collection ("Catalogue of Cell Lines and Hybridomas," Fifth edition (1985) Rockville, Maryland, U.S.A., which is incorporated herein by reference).
  • substantially identical modified heavy and light chains can be readily designed and manufactured utilizing various recombinant DNA techniques well known to those skilled in the art.
  • the chains can vary from the naturally-occurring sequence at the primary structure level by several amino acid substitutions, terminal and intermediate additions and deletions, and the like.
  • polypeptide fragments comprising only a portion (usually at least about 60-80%, typically 90-95%) of the primary structure may be produced, which fragments possess one or more immunoglobulin activities (e.g.. complement fixation activity) , while exhibiting lower immunogenicity.
  • the immunoglobulin-related genes contain separate functional regions, each having one or more distinct biological activities. These may be fused to functional regions from other genes (e.g. r enzymes, see, commonly assigned U.S.S.N. 132,387, filed Dec. 15, 1987, which is incorporated herein by reference) to produce fusion proteins (e.g.. immunotoxins) having novel properties.
  • r enzymes see, commonly assigned U.S.S.N. 132,387, filed Dec. 15, 1987, which is incorporated herein by reference
  • modifications of the genes may be readily accomplished by a variety of well-known techniques, such as site-directed mutagenesis (see. Gillman and Smith, Gene 8.:81-97 (1979) ; Roberts, S. et al. Nature 328:731-734 (1987); and U.S. Patent no.
  • Preferred DNA segments encoding variable regions of the present invention will typically be substantially homologous to the sequences of Figures 1 and 2 (i.e., capable of hybridizing to the sequences under stringent conditions of low salt and high temperature) , most preferrably at least about 90-95% homologous or more.
  • nucleic acid sequences of the present invention capable of ultimately expressing the desired chimeric antibodies can be formed from a variety of different polynucleotides (genomic or cDNA, RNA, etc.) and components (e.g. , V, J, D, and C regions) , as well as by a variety of different techniques. Joining appropriate genomic sequences is presently the most common method of production, but cDNA sequences may also be utilized (see, European Patent Application Nos. 85102655.8, 85305604.2, 84302368.0 and 85115311.4, as well as PCT Application Nos. GB85/00392 and US86/02269, all of which are incorporated herein by reference) .
  • the DNA sequences of the present invention will be expressed in hosts after the sequences have been operably linked to (i.e. , positioned to ensure the functioning of) an expression control sequence.
  • These expression vectors are typically replicable in the host organisms either as episomes or as an integral part of the host chromosomal DNA.
  • expression vectors will contain selection markers, e.g. , tetracycline or neomycin, to permit detection of those cells transformed with the desired DNA sequences (see, e.g. , U.S. Patent 4,704,362, which is incorporated herein by reference) .
  • E. coli is one prokaryotic host useful particularly for cloning the DNA sequences of the present invention.
  • microbial hosts suitable for use include bacilli, such as Bacillus subtilus, and other enterobacteriaceae, such as Salmonella, Serratia, and various Pseudomonas species.
  • prokaryotic hosts one can also make expression vectors, which will typically contain expression control sequences compatible with the host cell (e.g. , an origin of replication) .
  • expression control sequences compatible with the host cell (e.g. , an origin of replication) .
  • any number of a variety of well- known promoters will be present, such as the lactose promoter system, a tryptophan (trp) promoter system, a beta-lactamase promoter system, or a promoter system from phage lambda.
  • the promoters will typically control expression, optionally with an operator sequence, and have ribosome binding site sequences and the like, for initiating and completing transcription and translation.
  • Saccharomvces is a preferred host, with suitable vectors having expression control sequences, such as promoters, including 3-phosphoglycerate kinase or other glycolytic enzymes, and an origin of replication, termination sequences and the like as desired.
  • mammalian tissue cell culture may also be used to produce the polypeptides of the present invention (see, Winnacker, "From Genes to Clones,” VCH Publishers, N.Y., N.Y. (1987), which is incorporated herein by reference) .
  • Eukaryotic cells are actually preferred, because a number of suitable host cell lines capable of secreting intact immunoglobulins have been developed in the art, and include the CHO cell lines, various COS cell lines, HeLa cells, myeloma cell lines, etc, but preferably transformed B-cells or hybrido as.
  • Expression vectors for these cells can include expression control sequences, such as an origin of replication, a promoter, an enhancer (Queen, C.
  • Preferred expression control sequences are promoters derived from immunoglobulin genes, SV40, Adenovirus, Bovine Papilloma Virus, and the like.
  • the vectors containing the DNA segments of interest can be transferred into the host cell by well-known methods, which vary depending on the type of cellular host. For example, calcium chloride transfection is commonly utilized for prokaryotic cells, whereas calcium phosphate treatment may be used for other cellular hosts. (See generally, Maniatis et al., Molecular Cloning: A Laboratory Manual, Cold Spring Harbor Press, (1982), which is incorporated herein by reference.)
  • the whole chimeric antibodies, their dimers, or individual light and heavy chains of the present invention can be purified according to standard procedures of the art, including ammonium sulfate precipitation, fraction column chro atography, gel electrophoresis and the like. (See, generally. Scopes, R. , Protein Purification, Springer-Verlag, N.Y. (1982).) Once purified, partially or to homogeneity as desired, the polypeptides may then be used therapeutically or in developing and performing assay procedures, immunofluorescent stainings, and the like. (See, generally, Immunological Methods, Vols. I and II, Eds. Lefkovits and Pernis, Academic Press, New York, N.Y. (1979 and 1981).)
  • the chimeric antibodies of the present invention will typically find use individually in treating a T-cell mediated disease state. Generally, where the cell linked to a disease has been identified as IL-2 receptor bearing, then the chimeric antibodies are suitable (see, U.S.S.N. 7- 085,707, entitled “Treating Human Malignancies and Disorders,” which is incorporated herein by reference) .
  • Other diseases include Type I diabetes, multiple sclerosis, rheumatoid arthritis, Lupus erythematosus, and Myasthenia Gravis.
  • the antibodies of the present invention may also be used in combination with other antibodies, particularly human chimeric antibodies or human monoclonal antibodies reactive with other markers on cells responsible for the disease.
  • suitable T-cell markers can include those grouped into the so-called "Clusters of Differentia ion," as named by the First International Leukocyte Differentiation Workshop, Leukocyte Typing, Eds. Bernard, et al., Springer-Verlag, N.Y. (1984) , which is incorporated herein by reference.
  • the chimeric antibodies can also be used as separately administered compositions given in conjunction with chemotherapeutic or immunosuppressive agents.
  • the agents will include a cephalosporin or a purine analog (e.g. , methotrexate, 6-mercaptopurine, or the like) , but numerous additional agents (e.g.. cyclophospha ide, sulfa drugs, etc.) well-known to those skilled in the art may also be utilized.
  • a preferred pharmaceutical composition of the present invention comprises the use of the subject chimeric antibodies in immunotoxins.
  • I munotoxins are characterized by two components and are particularly useful for killing selected cells in vitro or in vivo.
  • One component is a cytotoxic agent which is usually fatal to a cell when attached or absorbed.
  • the second component known as the "delivery vehicle,” provides a means for delivering the toxic agent to a particular cell type, such as cells comprising a carcinoma.
  • the two components are commonly chemically bonded together by any of a variety of well-known chemical procedures.
  • the cytotoxic agent is a protein and the second component is an intact immunoglobulin, such as a chimeric antibody
  • the linkage may be by way of heterobifunctional cross-linkers, e.g..
  • Cytotoxic agents can include radionuclides, such as Iodine-131, Yttrium-90, Rhenium-188, and Bismuth-212; a number of chemotherapeutic drugs, such as vindesine, methotrexate, adriamycin, and cisplatinum; and cytotoxic proteins such as ribosomal inhibiting proteins, pokeweed antiviral protein, abrin and ricin (or their A- chains, diphtheria toxin A-chains, Pseudomonas exotoxin A, etc.) or an agent active at the cell surface, such as the phospholipase enzymes (e.g. , phospholipase C) .
  • radionuclides such as Iodine-131, Yttrium-90, Rhenium-188, and Bismuth-212
  • chemotherapeutic drugs such as vindesine, methotrexate, adriamycin, and cisp
  • the delivery component of the immunotoxin will include the chimeric antibodies of the present invention. Intact chimeric immunoglobulins or their binding fragments, such as Fab, F(ab 2 ) , etc., are preferably used. Typically, the chimeric antibodies in the immunotoxins will be of the human IgM or IgG isotype, but other mammalian constant regions may be utilized as desired.
  • compositions for parenteral administration are particularly - useful for parenteral administration, i.e. , subcutaneously, intramuscularly or intravenously.
  • the compositions for parenteral administration will commonly comprise a solution of the antibody or a cocktail thereof dissolved in an accept- able carrier, preferably an aqueous carrier.
  • aqueous carriers can be used, e.g. , water, buffered water, 0.4% saline, 0.3% glycine and the like. These solutions are sterile and generally free of particulate matter.
  • These compositions may be sterilized by conventional, well known sterilization techniques.
  • compositions may contain pharmaceutically acceptable auxiliary substances as required to approximate physiological conditions such as pH adjusting and buffering agents, toxicity adjusting agents and the like, for example sodium acetate, sodium chloride, potassium chloride, calcium chloride, sodium lactate, etc.
  • concentration of antibody in these formulations can vary widely, i.e.. from less than about 0.5%, usually at or at least about 1% to as much as 15 or 20% by weight and will be selected primarily based on fluid volumes, viscosities, etc., in accordance with the particular mode of administration selected.
  • a typical pharmaceutical composition for intramuscular injection could be made up to contain 1 ml sterile buffered water, and 50 g of antibody.
  • a typical composition for intravenous infusion could be made up to contain 250 ml of sterile Ringer's solution, and 150 mg of antibody.
  • Actual methods for preparing parenterally administrable compositions will be known or apparent to those skilled in the art and are described in more detail in, for example, Remington's Pharmaceutical Science, 15th Ed. , Mack Publishing Company, Easton, Pennsylvania (1980) , which is incorporated herein by reference.
  • the antibodies of this -invention can be ly ⁇ philized for storage and reconstituted in a suitable carrier prior to use. This technique has been shown to be effective with conventional immune globulins and art-known lyophilization and reconstitution techniques can be employed. It will be appreciated by those skilled in the art that lyophilization and reconstitution can lead to varying degrees of antibody activity loss (e.g. , with conventional immune globulins, IgM antibodies tend to have greater activity loss than IgG antibodies) and that use levels may have to be adjusted to compensate.
  • compositions containing the present human chimeric antibodies or a cocktail thereof can be administered for the prophylactic and/or therapeutic treatments.
  • compositions are administered to a patient already, in an amount sufficient to cure or at least partially arrest the infection and its complications.
  • An amount adequate to accomplish this is defined as a "therapeutically effective dose.” Amounts effective for this use will depend upon the severity of the infection and the general state of the patient's own immune system, but generally range from about 1 to about 200 mg of antibody per dose, with dosages of from 5 to 25 mg per patient being more commonly used. It must be kept in mind that the materials of this invention may generally be employed in serious disease states, that is life-threatening or potentially life- threatening situations.
  • compositions containing the present antibodies or a cocktail thereof are administered to a patient not already in a disease state to enhance the patient's resistance.
  • Such an amount is defined to be a "prophylactically effective dose.”
  • the precise amounts again depend upon the patient's state of health and general level of immunity, but generally range from 0.1 to 25 mg per dose, especially 0.5 to 2.5 mg per patient.
  • Single or multiple administrations of the compositions can be carried out with dose levels and pattern being selected by the treating physician.
  • the pharmaceutical formulations should provide a quantity of the antibody(ies) of this invention sufficient to effectively treat the patient.
  • Chimeric antibodies of the present invention can further find a wide variety of utilities vitro.
  • the chimeric antibodies can be utilized for T-cell typing, for isolating specific IL-2 receptor bearing cells or fragments of the receptor, for vaccine preparation, or the like.
  • the chimeric antibodies may either be labeled or unlabeled.
  • Unlabeled antibodies can be used in combination with other labeled antibodies (second antibodies) that are reactive with the chimeric antibody, such as antibodies specific for human immunoglobulin constant regions.
  • second antibodies labeled antibodies
  • the chimeric antibodies can be directly labeled.
  • labels may be employed, such as radionuclides, fluorescers, enzymes, enzyme substrates, enzyme cofactors, enzyme inhibitors, ligands
  • Kits can also be supplied for use with the subject antibodies in the protection against or detection of a cellular activity or for the presence of a selected antigen.
  • the subject antibody composition of the present invention may be provided, usually in a lyophilized form in a container, either alone or in conjunction with additional antibodies specific for the desired cell type.
  • the antibodies which may be conjugated to a label or toxin, or unconjugated, are included in the kits with buffers, such as Tris, phosphate, carbonate, etc., stabilizers, biocides, inert proteins, e.g.
  • serum albumin or the like, and a set of instructions for use.
  • these materials will be present in less than about 5% wt. based on the amount of active antibody, and usually present in total amount of at least about 0.001% wt. based again on the antibody concentration.
  • a second antibody capable of binding to the chimeric antibody is employed in an assay, this will usually be present in a separate vial.
  • the second antibody is typically conjugated to a label and formulated in an analogous manner with the antibody formulations described above.
  • a splice and polyadenylation signal from the animal virus SV40 (850 bp) .
  • e Another part of SV40 containing its origin of replication (700 bp) ;
  • f Part of the plasmid pBR322, extending from the Sph I site to the Eco Rl site of the plasmid pMLl (Lusky, M and Botchan, M. Nature 293:79-81: 2300 bp) including the Amp gene and origin of replication.
  • pKcatH was cut with Bgl II, and the ends filled in with Klenow polymerase.
  • An Xba I linker having the sequence GCTCTAGAGC, was inserted at the filled-in Bgl II site. The resulting plasmid is called pKcatH - Xba.
  • pKcatH - Xba was cut partially with Xho II, run on an agar ⁇ se gel, and full-length linear plasmid (i.e. , plasmid cut exactly once with Xho II) was isolated.
  • the DNA was cut with Bam HI, run on a gel and the 5600 bp fragment isolated. Because of its size, this fragment had to extend from the Bam site to the Xho II site at the end of the CAT gene (Fig. 3). The fragment was ligated by itself.
  • the resulting plasmid is called pKcatH - Xba - BX. It is similar to pKcatH but has an Xba I site in place of the Bgl II site and is missing the Xho II - Bam HI SV40 fragment (Fig. 4) .
  • the plasmid pSV2neo (Southern, P.J. and Berg, P. J. Mol. App. Genet. 1:327-341 (1982)), was cut with Eco RI and Bam HI, and the large Eco RI - Bam HI fragment ligated to the same Eco RI - Bam HI fragment from pKcatH - Xba - BX used in (4) .
  • the resulting plasmid is called pSV2neo - EH - K , which is like pSV2gpt - EH - K, but has the Neo gene in place of the Gpt gene.
  • pSV2gpt - E réelle - c was cut with Xba I and Bam HI and the ends filled in with Klenow polymerase.
  • a fragment of the cloned human K constant segment gene (Hieter, P.A. et al., Cell 22:197-207 (1980)) was purified, extending from a Hind III site 336 bp before the coding region, to an Xba I site about 800 bp beyond the coding region, and the ends filled in.
  • the two fragments were ligated together, and a plasmid selected in which the Hind III site of the second*fragment was joined to the Xba I site and the Xba I site of the second fragment to the Bam HI site.
  • pV ⁇ l Fig. 6
  • An Xba I fragment containing the VJ region of the cloned anti-Tac light chain gene was prepared by in vitro mutagenesis (see, below) .
  • pV ⁇ l was cut with Xba I, treated with phosphatase, and ligated with the Xba I fragment.
  • a plasmid was selected in which the VJ region had the same orientation as the following C region, and called pLTAC2 (Fig. 7) .
  • pSV2neo - E réelle - K was cut with Xba I and Bam HI and the ends filled in with Klenow polymerase.
  • a 2800 bp fragment containing the human C7I gene was purified from the phage HG3A (Ellison, J.W. et al., Nucleic Acids Res. 10:4071-4079 (1982)), extending from a Hind III site 210 bp before the CHI exon to a Pvu II site about 1100 bp after the CH3 exon, and the ends filled in.
  • the two fragments were ligated together, and a plasmid selected in which the Hind III site of the second fragment was joined to the Xba I site and the Pvu II site of the second fragment to the Bam HI site. Because of the sequences of these sites, this recreated an Xba I site and a Bam HI site.
  • the new plasmid is called pV 7 lneo (Fig. 8) .
  • pSV2neo - E réelle - K was cut with Xba I and Bam HI and the ends filled in with Klenow polymerase.
  • a 3600 bp fragment containing the human C 7 3 gene (Takahashi, N. et al., Cell ⁇ 671-679 (1982)) was purified extending from a Hind III site 210 bp before the CHI exon to a Pvu II site about 1100 bp after the CH3 exon, and the ends filled in.
  • the two fragments were ligated together, and a plasmid selected in which the Hind III site of the second fragment was joined to the Xba I site and the Pvu II site of the second fragment to the Bam HI site.
  • the new plasmid is called pV 3neo and is identical to pV7lneo except it has the C 7 3 gene instead of C 7 I. 10.
  • An Xba I fragment containing the VJ region of the cloned anti-Tac heavy china gene was prepared by in vitro mutagenesis (see, below) .
  • pV7lneo was cut with Xba I, treated with phosphatase, and ligated with the Xba I fragment.
  • a plasmid was selected in which the VJ region had the same orientation as the following C region, and called pHTAC.
  • PV7I Three additional plasmids were prepared respectively from pV7lneo, pV 3neo and pHTAC, called respectively PV7I, PV73 and pGTACl.
  • the original plasmid was cut with Hind III and Bam HI, and the large Hind III - Bam HI fragment purified.
  • the plasmid pXBohph containing the Hyg gene (Blochlinger, K. and Diggelmann, H. Mol. Cell. Biol. 4:2929-2931 (1984)) was cut with Hind III and Bam HI and a 1600 bp fragment containing the Hyg gene purified.
  • the fragments from the original plasmids were each ligated to the pXbohph fragment.
  • PV 7 I is shown in Fig. 9 and pGTACl in Fig.
  • pHTAC was cut with Xba I and the small Xba I fragment, containing the heavy VJ region, purified.
  • PV 3 was cut with Xba I, treated with phosphatase, and ligated with the small Xba I fragment.
  • a plasmid was selected in which the VJ region had the same orientation as the following C region, and called pGTAC3. It is similar to pGTACl, but has the C 7 3 region instead of the C7I region.
  • mRNA was extracted from approximately 10-8 ascites cells of the anti-Tac hybrido a by the guanidium isothiocyanate method followed with poly A selection on Hybond- AP paper (Amersham) .
  • cDNA was prepared by the method of Gubler and Hoffman (Gubler, U. and Hoffman, B.J. Gene
  • plaques Approximately 10,000 plaques were screened with the oligonucleotide probes. About 100 plaques were positive for the 7 probe and 40 for the 7 probe.
  • VJ segments of the other pair of K clones were sequences completely and were identical (Fig. 1) .
  • This light chain uses the JK5 segment and contains all the amino acid residues conserved in mouse K light chains (Kabat, E. A. et al.. Sequences of Proteins of Immunological Interest, particularly pg. 45 et seq, all of which is incorporated herein by reference) .
  • Plasmid vectors were prepared for the construction and expression of chimeric light and heavy chain genes as described above.
  • the plasmid pV ⁇ l (Fig. 6) contains the human K genomic C segment, including 336 bp of the preceding intron and the poly A signal. It also contains a strong promoter sequence from the MOPC 41 K gene, and the heavy chain enhancer sequence. There is a unique Xba I site between the promoter and the C intron.
  • the plasmid also contains the gpt gene for selection. Two other very similar plasmids were prepared by using the human 71 and 7 3 C regions in place of the K C region.
  • the region inserted between the Xba I and Bam HI sites extended form about 210 bp 5* of the CHI exon to about 1100 bp 3 ' of the CH3 exon.
  • the gpt gene was replaced with the hyg gene to confer resistance to hygromycin.
  • the next 16 nucleotides were the same as the sequence that follows JK5 in mouse genomic DNA, and therefore included a splice donor signal.
  • the final nucleotides of the oligo consisted of an Xba I site followed by a short irrelevant sequence.
  • This oligonucleotide was hybridized to the Ml3 phage DNA containing the K CDNA and extended with Klenow polymerase (Fig. 3) .
  • the DNA was denatured and hybridized to a "reverse primer," which represents M13 DNA 5' to the cDNA insert.
  • the reverse primer was extended, and the DNA cut with Xba I.
  • the Xba I fragment consisting of the extended VJ segment of the cDNA was purified, and cloned in the correction orientation into the Xba I site of pV ⁇ l to obtain the plasmid pLTAC2 (Fig. 7) .
  • the final chimeric K gene has a VJ-C intron, of which the first 14 bp and last 209 bp are respectively the same as in mouse and human genomic DNA, as verified by direct sequencing of the construct.
  • the gene is transcribed from a K promoter, stimulated by the heavy chain enhancer. Based on results obtained with deleted introns, we expected that the intron would be correctly spliced from the transcribed RNA.
  • the VJ region from the anti-Tac 72a heavy chain cDNA, followed by a splice donor signal was inserted into the Xba I site of pV 7 lneo.
  • the resulting plasmid pHTAC contains a chimeric heavy chain gene, with a synthetic intron between the mouse VJ and human C 7 I segments.
  • RF DNA of the phage M13mpll was cut with Eco Rl and Xba I, the ends were filled-in, and the DNA ligated was ligated and transformed into JM101 cells. A plaque was picked and the DNA was sequenced to verify that the ends of the DNA had joined correctly, recreating the Eco Rl and Xba I sites with the intervening DNA segment deleted. This phage is designated M13mpllD. Eco Rl fragments containing the anti-Tac light and heavy chain cDNAs were separately cloned into the Eco Rl site of M13mpll, so that their 5' ends aubbed the Xba I site. The resulting phage are respectively denoted M13mpllL and M13mpllH.
  • the following 48-nucleotide long primer was synthesized and gel-purified: CCAGAATTCTAGAAAAGTGTACTTACGTTTCAGCTCCAGCTTGGTCCC. From the 3' end, the first 22 residues of the primer are the same as the last 22 bp of the JK5 segment (non-coding strand) . The next 16 nucleotides are the same as the sequence that follows JK5 in mouse genomic DNA and therefore includes a splice donor signal (abbreviated SD in Fig. 11) . The final nucleotides of the oligo consist of an Xba I site followed by a short irrelevant sequence.
  • Klenow polymerase The solution was incubated for 30 minutes at 37 X C. 50 ng of the "reverse primer" AACAGCTATGACCATG (New England Biolabs) , which can hybridize to the newly synthesized strand upstream of the Xba I site (Fig. 11) , was added. The solution was incubated at 95 ⁇ C for 3 min and put on ice. An additional 4 ul of 200 uM each dNTP and 5u Klenow polymerase was added, and the solution incubated for 30 minutes at 37 C. The solution was extracted with phenol- chloroform, precipitated with ethanol, resuspended, and digested with 20u Xba I.
  • AACAGCTATGACCATG New England Biolabs
  • the digested DNA was run on a 4% polyacrylamide gel and visualized with ethidiu bromide. In addition to high molecular weight DNA, an approximately 400 bp fragment was visible, corresponding to the VJ region of the light chain cDNA with a "tail" (Fig. 11) . The fragment was cloned directly into the Xba I site of pV ⁇ l in the correct orientation.
  • the first 21 residues of the primer are the same as the last 21 bp of the JH2 segment (non-coding strand) .
  • the next 19 nucleotides are the same as the sequence that follows JH2 in mouse genomic DNA and therefore includes a splice donor signal.
  • the final nucleotides of the oligo consist of an Xba I site followed by a short irrelevant sequence.
  • This primer was hybridized to M13mpllH DNA following the same protocol as above, in order to synthesize a fragment containing the VJ region of the heavy chain cDNA. The fragment was cloned directly into the Xba I site of pV 7 lneo in the correct orientation.
  • Chimeric Antibody Specificity CR2-2 and CEM cells are human T cell lines that are respectively positive and negative for surface expression of the IL2 receptor (IL2R) .
  • Whole CR2-2 cells were used to demonstrate binding specificity of the chimeric antibody in an ELISA assay, and CEM cells were used as a negative control.
  • IL2R IL2 receptor
  • Antibody to be tested was prepared in several ways.
  • Anti-Tac monoclonal antibody supplied by T. Waldmann, was purified from mouse ascites by passage through a DEAE-dextran column.
  • the cell line L40H4 created by transfecting SP2/0 cells with the chimeric light and gamma 1 heavy chain plasmids pLTAC2 and pGTACl (described above) , was injected into mice to form an ascites. From 3 mis of ascites fluid, 650 ⁇ g of chimeric gamma 1 antibody was purified by passage through a column of Baker Abx and a size exclusion column.
  • 2 x 10 6 cells were mixed with either purified antibody in 20 ⁇ l FACS buffer or 20 ⁇ l media supernatent from the transfected cell lines, and incubated on i ce f or 2 hr. The cells were washed 3 times with 1 ⁇ l of FACS buffer (being collected by brief centrifugation after each wash) .
  • the cells that had been incubated with anti-Tac antibody were mixed with 0.5 ⁇ l peroxidase-conjugated goat anti-mouse antibody (Fab) ' fragment (Tago Immunologicals, Burlingame, California) in 20 ⁇ l FACS buffer, and the cells that had been incubated with chimeric antibody were mixed with 0.5 ⁇ l peroxidase- conjugated goat anti-human gamma chain (Fab) ' fragment (Tago) in 20 ⁇ l FACS buffer.
  • the cells were incubated for 30 min on ice and then washed 3 times with FACS buffer. They were then mixed with 100 ⁇ l peroxidase development solution and incubated for 5 min at room temperature.
  • the cells were spun out and the supernatents were transferred to a 96-well plate and the OD's determined in an ELISA reader. The OD's at 414 nm for the cells treated with each antibody are given in Table l.
  • the anti-Tac antibody itself bound to the IL2R+ CR2-2 cells but not to the IL2R- CEM cells.
  • An equal amount of purified gamma 1 chimeric antibody gave an equivalent amount of binding to the CR2-2 cells as the anti- Tac, and also failed to bind to the CEM cells. All the supernatants from both the chimeric antibody producing cells bound to the CR2-2 cells. As additional negative controls, the 51.3 supernatant did not bind to the CEM cells, and supernatant from the parental SP2/0 cells did not bind the CR2-2 cells.
  • the plate was washed, incubated with peroxidase-conjugated goat anti-human gamma chain antibody, washed again, incubated with peroxidase developing solution and the OD's at 414 nm determined in an ELISA reader.
  • the media supernatants contained respectively about 8 and 7 ng per ⁇ l, that is the cells secreted 8 and 7 ⁇ g antibody per 10 6 cells per 24 hr.
  • the mixed lymphocyte reaction is a model for transplant rejection and was used to analyze the effectiveness of the chimeric anti-Tac antibodies.
  • Table 3 The experiments shown in Table 3 below were performed by standard methods (see, e.g. , Strong, D.M. et al., In Vitro Stimulation of Murine Spleen Cells Using a Microculture System and a .
  • the chimeric antibodies of the present invention offer numerous advantages of other human IL-2 receptor-specific antibodies.
  • the present human chimeric antibodies can be more economically produced and contain substantially less foreign amino acid sequences. This reduced likelihood of antigenicity after injection into a human patient represents a significant therapeutic improvement.

Abstract

Chimeric antibodies specifically reactive with human IL-2 receptors are prepared employing recombinant DNA technology for use in, e.g., treatment of T-cell mediated disorders.

Description

IL-2 RECEPTOR-SPECIFIC CHIMERIC ANTIBODIES
Field of the Invention
The present invention relates generally to the combination of recombinant DNA and monoclonal antibody technologies for developing novel therapeutic and other agents and, more particularly, to the production of chimeric antibodies specific for the human interleukin-2 receptor and the use of such chimeric antibodies in treating T-cell mediated human disorders.
Background of the Invention
In mammals, the immune response is mediated by two types of cells that interact specifically with foreign material, i.e. , antigens. One of these cell types, B-cells, are responsible for the production of antibodies. The second cell class, T-cells, include a wide variety of cellular subsets controlling the in vivo function of both B-cells and a wide variety of other hematopoietic cells, including T- cells.
One way in which T-cells exert this control is through the production of a lymphokine known as interleukin-2 (IL-2) , originally named T-cell growth factor. IL-2's prime function appears to be the stimulation and maintenance of T- cells. Indeed, some i munologists believe that IL-2 may be at the center of the entire immune response (see, Farrar, J. , et al., Immunol. Rev. 3:129-166 (1982), which is incorporated herein by reference) .
To exert its biological effects, IL-2 interacts with a specific high-affinity membrane receptor (Greene, . , et al., Progress in Hematology XIV, E. Brown, ed. , Grune and Statton, New York (1986), at pgs. 283 ff) . The human IL-2 receptor is a complex glycoprotein, with one chain 55kD in size (Leonard, . et al., J. Biol. Che . 260;1872 (1985)). A gene encoding this protein has been isolated, and predicts a 272 a ino acid peptide, including a 21 amino acid signal peptide, (see, Leonard, . et al., Nature 311: 626 (1984)). Much of the elucidation of the human IL-2 receptor's structure and function is due to the development of specifically reactive monoclonal antibodies. In particular, one monoclonal antibody, known as anti-Tac, (Uchiyama et al., . Immunol. 126:1393 (1981)) has shown that IL-2 receptors can be detected on T-cells, but also on cells of the onocyte-macrophage family, Kupffer cells of the liver, Langerhans1 cells of the skin and, of course, activated T-cells. Importantly, resting T-cells, B-cells or circulating machrophages typically do not display the IL-2 receptor (Herrmann, et al., J. Exp. Med. 162:1111 (1985)).
The anti-Tac monoclonal antibody has also been used to define lymphocyte functions that require IL-2 interaction, and has been shown to inhibit various T-cell functions, including the generation of cytotoxic and suppressor T lymphocytes in cell culture. Also, based on studies with anti-Tac, a variety of disorders are now associated with improper IL-2 receptor expression by T-cells, in particular adult T-cell leukemia.
More recently, the IL-2 receptor has been shown to be an ideal target for novel therapeutic approaches to T-cell mediated diseases. The anti-Tac monoclonal antibody can be used either alone or as an immunoconjugate (e.g. with Ricin A, isotopes and the like) to effectively remove cells bearing the IL-2 receptor. These agents can, for example, theoretically eliminate IL-2 receptor-expressing leukemic cells, certain B-cells, or activated T-cells involved in a disease state, yet allow the retention of mature normal T- cells and their precursors to ensure the capability of mounting a normal T-cell immune response as needed. In general, other T-cell specific agents can destroy essentially all peripheral T-cells, which limits therapeutic efficacy. Overall, the use of monoclonal antibodies specific for the IL-2 receptor can be expected to have therapeutic utility in autoimmune diseases, organ transplantation and any unwanted response by activated T-cells. Indeed, clinical trials have been initiated (see, generally, Waldman, T., Science 232:727- 732 (1986) , which is incorporated herein by reference) . Unfortunately, the use of the anti-Tac monoclonal antibody has certain drawbacks, particularly in repeated therapeutic regimens. As a mouse monoclonal, it does not fix human complement well, whereas a human equivalent may be more efficient. More importantly, however, anti-Tac monoclonal antibody contains substantial urine amino acid sequences that will be antigenic when injected into a human patient. Numerous studies have shown that the immune response elicited by a patient against the nonbinding portion of an injected mouse monoclonal antibody can be quite strong, essentially eliminating the antibody's therapeutic utility after an initial treatment. As increasing numbers of different mouse monoclonal antibodies can be expected to be developed to treat various diseases, after the first and second treatments with different mouse monoclonal antibodies, subsequent treatments can be dangerous in themselves.
Thus, there is a need for improved forms of the anti-Tac monoclonal antibody that are substantially less antigenic, yet easily and economically produced in a manner suitable for therapeutic formulation. The present invention fulfills these needs.
Summary of the Invention
The present invention provides novel compositions useful in the treatment of T-cell mediated human disorders, the compositions containing a chimeric antibody specifically capable of binding to human IL-2 receptors, such as at the epitope bound by the anti-Tac monoclonal antibody. The IL-2 chimeric antibody can have two pairs of light chain/heavy chain complexes, wherein at least one pair has chains comprising mouse variable regions joined with human constant region segments, with or without naturally-associated J and D segments.
The chimeric antibodies, or binding fragments thereof, of the present invention may be produced by a variety of recombinant DNA techniques, with ultimate expression in transfected cells, preferably immortalized eukaryotic cells, such as myeloma or hybridoma cells. Polynucleotides comprising a first sequence coding for a human immunoglobulin constant region and a second sequence coding for the desired mouse immunoglobulin variable or hypervariable region can be produced synthetically or by combining appropriate cDNA and genomic DNA segments.
When the chimeric antibodies are complexed with a cytotoxic agent, such as a radionuclide, a ribosomal inhibiting protein or a cytotoxic agent active at cell surfaces, the compounds will be particularly useful in treating T-cell mediated disorders. These compounds can be provided in a pharmaceutically accepted dosage form, which will vary depending on the mode of administration.
BRIEF DESCRIPTION OF THE FIGURES Figure 1 depicts the DNA coding sequence and putative amino acid sequence of the V and J regions of the anti-Tac light chain.
Figure 2 depicts the DNA coding sequence and putative amino acid sequence of the V and J regions of the anti-Tac heavy chain.
Figures 3-10 are schematic diagrams of the plasmids utilized to demonstrate the present invention.
Figure 11 represents an overview of a preferred strategy for preparation of V and J regions for insertion into plasmids.
DETAILED DESCRIPTION OF THE INVENTION In accordance with the present invention, DNA sequences encoding mouse variable/human constant region chimeric antibodies capable of binding epitopes on human IL-2 receptors are provided. When placed in expression vectors and suitable hosts, large quantities of chimeric antibodies can be produced. Preferably, the chimeric antibodies will have substantially the same binding profile or characteristics as (e.g. , be cross-reactive or capable of blocking) the binding of the anti-Tac monoclonal antibody, such as antibodies produced by the myeloma cell line deposited with the A.T.C.C. and designated accession number CRL 9688. These chimeric antibodies find use, for example, in the treatment of T-cell mediated disorders in human patients. The basic immunoglobin structural unit is known to comprise a tetramer. Each tetramer is composed of two identical pairs of polypeptide chains, each pair having one "light" (about 25kD) and one "heavy" chain (about 50-70kD) . The NH -terminus of each chain begins a variable region of about 100 to 110 or more amino acids primarily responsible for antigen recognition. The COOH terminus of each chain defines a constant region primarily responsible for effector function.
Light chains are classified as either kappa or lambda. Heavy chains are classified (and subclassified) as gamma, mu, alpha, delta, or epsilon, and define the immunoglobulin's isotype as IgG, IgM, IgA, IgD and IgE, respectively. Within light and heavy chains, the variable and constant regions are joined by a "J" region of about 12 or more amino acids, with the heavy chain also including a "D" region of about 12 more amino acids. (See, generally. Fundamental Immunology, Ed. Paul, W. Chapter 7, pgs. 131-166, Raven Press, N.Y. (1984), which is incorporated"herein by reference.)
Chimeric antibodies are antibodies whose light and heavy chain genes have been constructed, typically by genetic engineering, from immunoglobulin gene segments belonging to different species. For example, the variable (V) segments of the genes for a mouse monoclonal antibody may be joined to human constant (C) segments, such as 7 , and 7 . A preferred therapeutic chimeric antibody is thus a hybrid protein consisting of the V or antigen-binding domain from a mouse antibody and the C or effector domain from a human antibody, although other mammalian species may be used.
Human chimeric antibodies have at least three potential advantages over mouse antibodies for use in human therapy: 1) Because the effector portion is human, it may interact better with the other parts of the human immune system (e.g. , destroy the target cells more efficiently by complement-dependent cytotoxicity (CDC) or antibody-dependent cellular cytotoxicity (ADCC) ) .
2) The human immune system should not recognize the C region of the chimeric antibody as foreign, and therefore the antibody response against an injected chimeric antibody should be less than against a totally foreign mouse antibody.
3) Injected mouse antibodies have been reported to have a half-life in the human circulation much shorter than the half-life of normal antibodies (Shaw, D. et al., J. Immunol. 138:4534-4538 (1987)) . It is possible that injected chimeric antibodies will have a half-life more like that of human antibodies, allowing smaller and less frequent doses to be given.
In one aspect, the present invention is directed to recombinant DNA segments encoding the heavy and/or light chain variable or hypervariable regions from the anti-Tac monoclonal antibody. The DNA segments encoding these regions will typically be joined to DNA segments encoding appropriate constant regions, such as human gamma heavy chain regions or human kappa light chain regions. The preferred variable region DNA sequences, which on expression code for the polypeptide chains comprising the anti-Tac light and heavy chain variable regions (with naturally-associated J regions) , are shown in Figures 1 and 2, respectively. Due to codon degeneracy and non-critical amino-acid substitutions, other DNA sequences can be readily substituted for those sequences, as detailed below.
The DNA segments will typically further include an expression control DNA sequence operably linked to the chimeric antibody coding sequences, including naturally- associated or heterologous promoter regions. Preferably, the expression control sequences will be eukaryotic promoter systems in vectors capable of transforming or transfecting eukaryotic host cells. Once the vector has been incorporated into the appropriate host, the host is maintained under conditions suitable for high level expression of the nucleotide sequences, and, as desired, the collection and purification of the light chains, heavy chains, light/heavy chain dimers or intact chimeric antibodies may follow.
It is well known that native forms of "mature" immunoglobulins will vary somewhat in terms of length by deletions, substitutions, insertions or additions of one or more amino acids in the sequences. Thus, both the variable and constant regions are subject to substantial natural modification, yet are "substantially identical" and still capable of retaining their respective activities. Human constant region DNA sequences can be isolated in accordance with well known procedures from a variety of human cells, but preferably immortalized B-cells. Suitable source cells for the DNA sequences and host cells for expression and secretion can be obtained from a number of sources, such as the American Type Culture Collection ("Catalogue of Cell Lines and Hybridomas," Fifth edition (1985) Rockville, Maryland, U.S.A., which is incorporated herein by reference).
In addition to these naturally-occurring forms of immunoglobulin chains, "substantially identical" modified heavy and light chains can be readily designed and manufactured utilizing various recombinant DNA techniques well known to those skilled in the art. For example, the chains can vary from the naturally-occurring sequence at the primary structure level by several amino acid substitutions, terminal and intermediate additions and deletions, and the like. Alternatively, polypeptide fragments comprising only a portion (usually at least about 60-80%, typically 90-95%) of the primary structure may be produced, which fragments possess one or more immunoglobulin activities (e.g.. complement fixation activity) , while exhibiting lower immunogenicity. In particular, it is noted that like many genes, the immunoglobulin-related genes contain separate functional regions, each having one or more distinct biological activities. These may be fused to functional regions from other genes (e.g. r enzymes, see, commonly assigned U.S.S.N. 132,387, filed Dec. 15, 1987, which is incorporated herein by reference) to produce fusion proteins (e.g.. immunotoxins) having novel properties. In general, modifications of the genes may be readily accomplished by a variety of well-known techniques, such as site-directed mutagenesis (see. Gillman and Smith, Gene 8.:81-97 (1979) ; Roberts, S. et al. Nature 328:731-734 (1987); and U.S. Patent no. 4,703,008, all of which are incorporated herein by reference) . Preferred DNA segments encoding variable regions of the present invention will typically be substantially homologous to the sequences of Figures 1 and 2 (i.e., capable of hybridizing to the sequences under stringent conditions of low salt and high temperature) , most preferrably at least about 90-95% homologous or more.
The nucleic acid sequences of the present invention capable of ultimately expressing the desired chimeric antibodies can be formed from a variety of different polynucleotides (genomic or cDNA, RNA, etc.) and components (e.g. , V, J, D, and C regions) , as well as by a variety of different techniques. Joining appropriate genomic sequences is presently the most common method of production, but cDNA sequences may also be utilized (see, European Patent Application Nos. 85102655.8, 85305604.2, 84302368.0 and 85115311.4, as well as PCT Application Nos. GB85/00392 and US86/02269, all of which are incorporated herein by reference) .
As stated previously, the DNA sequences of the present invention (typcially at least about 30 contiguous nucleotides encoding 10 amino acids from the sequences in Figures l and 2) will be expressed in hosts after the sequences have been operably linked to (i.e. , positioned to ensure the functioning of) an expression control sequence. These expression vectors are typically replicable in the host organisms either as episomes or as an integral part of the host chromosomal DNA. Commonly, expression vectors will contain selection markers, e.g. , tetracycline or neomycin, to permit detection of those cells transformed with the desired DNA sequences (see, e.g. , U.S. Patent 4,704,362, which is incorporated herein by reference) .
E. coli is one prokaryotic host useful particularly for cloning the DNA sequences of the present invention.
Other microbial hosts suitable for use include bacilli, such as Bacillus subtilus, and other enterobacteriaceae, such as Salmonella, Serratia, and various Pseudomonas species. In these prokaryotic hosts, one can also make expression vectors, which will typically contain expression control sequences compatible with the host cell (e.g. , an origin of replication) . In addition, any number of a variety of well- known promoters will be present, such as the lactose promoter system, a tryptophan (trp) promoter system, a beta-lactamase promoter system, or a promoter system from phage lambda. The promoters will typically control expression, optionally with an operator sequence, and have ribosome binding site sequences and the like, for initiating and completing transcription and translation.
Other microbes, such as yeast may also be used for expression. Saccharomvces is a preferred host, with suitable vectors having expression control sequences, such as promoters, including 3-phosphoglycerate kinase or other glycolytic enzymes, and an origin of replication, termination sequences and the like as desired.
In addition to microorganisms, mammalian tissue cell culture may also be used to produce the polypeptides of the present invention (see, Winnacker, "From Genes to Clones," VCH Publishers, N.Y., N.Y. (1987), which is incorporated herein by reference) . Eukaryotic cells are actually preferred, because a number of suitable host cell lines capable of secreting intact immunoglobulins have been developed in the art, and include the CHO cell lines, various COS cell lines, HeLa cells, myeloma cell lines, etc, but preferably transformed B-cells or hybrido as. Expression vectors for these cells can include expression control sequences, such as an origin of replication, a promoter, an enhancer (Queen, C. et al., Immunol. Rev. 89:49-68 (1986), which is incorporated herein by reference) , and necessary processing information sites, such as ribosome binding sites, RNA splice sites, polyadenylation sites, and transcriptional terminator sequences. Preferred expression control sequences are promoters derived from immunoglobulin genes, SV40, Adenovirus, Bovine Papilloma Virus, and the like.
The vectors containing the DNA segments of interest (e.g.. the heavy and light chain encoding sequences and expression control sequences) can be transferred into the host cell by well-known methods, which vary depending on the type of cellular host. For example, calcium chloride transfection is commonly utilized for prokaryotic cells, whereas calcium phosphate treatment may be used for other cellular hosts. (See generally, Maniatis et al., Molecular Cloning: A Laboratory Manual, Cold Spring Harbor Press, (1982), which is incorporated herein by reference.)
Once expressed, the whole chimeric antibodies, their dimers, or individual light and heavy chains of the present invention can be purified according to standard procedures of the art, including ammonium sulfate precipitation, fraction column chro atography, gel electrophoresis and the like. (See, generally. Scopes, R. , Protein Purification, Springer-Verlag, N.Y. (1982).) Once purified, partially or to homogeneity as desired, the polypeptides may then be used therapeutically or in developing and performing assay procedures, immunofluorescent stainings, and the like. (See, generally, Immunological Methods, Vols. I and II, Eds. Lefkovits and Pernis, Academic Press, New York, N.Y. (1979 and 1981).)
The chimeric antibodies of the present invention will typically find use individually in treating a T-cell mediated disease state. Generally, where the cell linked to a disease has been identified as IL-2 receptor bearing, then the chimeric antibodies are suitable (see, U.S.S.N. 7- 085,707, entitled "Treating Human Malignancies and Disorders," which is incorporated herein by reference) .
For example, typical disease states suitable for treatment graft versus host disease and most patients undergoing an organ transplant, such as heart, lungs, kidneys, liver, etc. Other diseases include Type I diabetes, multiple sclerosis, rheumatoid arthritis, Lupus erythematosus, and Myasthenia Gravis. The antibodies of the present invention may also be used in combination with other antibodies, particularly human chimeric antibodies or human monoclonal antibodies reactive with other markers on cells responsible for the disease. For example, suitable T-cell markers can include those grouped into the so-called "Clusters of Differentia ion," as named by the First International Leukocyte Differentiation Workshop, Leukocyte Typing, Eds. Bernard, et al., Springer-Verlag, N.Y. (1984) , which is incorporated herein by reference.
The chimeric antibodies can also be used as separately administered compositions given in conjunction with chemotherapeutic or immunosuppressive agents. Typically, the agents will include a cephalosporin or a purine analog (e.g. , methotrexate, 6-mercaptopurine, or the like) , but numerous additional agents (e.g.. cyclophospha ide, sulfa drugs, etc.) well-known to those skilled in the art may also be utilized.
A preferred pharmaceutical composition of the present invention comprises the use of the subject chimeric antibodies in immunotoxins. I munotoxins are characterized by two components and are particularly useful for killing selected cells in vitro or in vivo. One component is a cytotoxic agent which is usually fatal to a cell when attached or absorbed. The second component, known as the "delivery vehicle," provides a means for delivering the toxic agent to a particular cell type, such as cells comprising a carcinoma. The two components are commonly chemically bonded together by any of a variety of well-known chemical procedures. For example, when the cytotoxic agent is a protein and the second component is an intact immunoglobulin, such as a chimeric antibody, the linkage may be by way of heterobifunctional cross-linkers, e.g.. SPDP, carbodiimide, glutaraldehyde, or the like. Production of various immunotoxins is well-known with the art, and can be found, for example in "Monoclonal Antibody-Toxin Conjugates: Aiming the Magic Bullet," Thorpe et al, Monoclonal Antibodies in Clinical Medicine, Academic Press, pp. 168-190 (1982) , which is incorporated herein by reference. A variety of cytotoxic agents are suitable for use in immunotoxins. Cytotoxic agents can include radionuclides, such as Iodine-131, Yttrium-90, Rhenium-188, and Bismuth-212; a number of chemotherapeutic drugs, such as vindesine, methotrexate, adriamycin, and cisplatinum; and cytotoxic proteins such as ribosomal inhibiting proteins, pokeweed antiviral protein, abrin and ricin (or their A- chains, diphtheria toxin A-chains, Pseudomonas exotoxin A, etc.) or an agent active at the cell surface, such as the phospholipase enzymes (e.g. , phospholipase C) . (See generally, "Chimeric Toxins," Olsnes and Phil, Pharmac. Ther.. 2J5:355-381 (1982), and "Monoclonal Antibodies for Cancer Detection and Therapy," eds. Baldwin and Byers, pp. 159-179, 224-266, Academic Press (1985), both of which are incorporated herein by reference.) The delivery component of the immunotoxin will include the chimeric antibodies of the present invention. Intact chimeric immunoglobulins or their binding fragments, such as Fab, F(ab2) , etc., are preferably used. Typically, the chimeric antibodies in the immunotoxins will be of the human IgM or IgG isotype, but other mammalian constant regions may be utilized as desired.
The chimeric antibodies and pharmaceutical compositions thereof of this invention are particularly - useful for parenteral administration, i.e. , subcutaneously, intramuscularly or intravenously. The compositions for parenteral administration will commonly comprise a solution of the antibody or a cocktail thereof dissolved in an accept- able carrier, preferably an aqueous carrier. A variety of aqueous carriers can be used, e.g. , water, buffered water, 0.4% saline, 0.3% glycine and the like. These solutions are sterile and generally free of particulate matter. These compositions may be sterilized by conventional, well known sterilization techniques. The compositions may contain pharmaceutically acceptable auxiliary substances as required to approximate physiological conditions such as pH adjusting and buffering agents, toxicity adjusting agents and the like, for example sodium acetate, sodium chloride, potassium chloride, calcium chloride, sodium lactate, etc. The concentration of antibody in these formulations can vary widely, i.e.. from less than about 0.5%, usually at or at least about 1% to as much as 15 or 20% by weight and will be selected primarily based on fluid volumes, viscosities, etc., in accordance with the particular mode of administration selected.
Thus, a typical pharmaceutical composition for intramuscular injection could be made up to contain 1 ml sterile buffered water, and 50 g of antibody. A typical composition for intravenous infusion could be made up to contain 250 ml of sterile Ringer's solution, and 150 mg of antibody. Actual methods for preparing parenterally administrable compositions will be known or apparent to those skilled in the art and are described in more detail in, for example, Remington's Pharmaceutical Science, 15th Ed. , Mack Publishing Company, Easton, Pennsylvania (1980) , which is incorporated herein by reference.
The antibodies of this -invention can be lyσphilized for storage and reconstituted in a suitable carrier prior to use. This technique has been shown to be effective with conventional immune globulins and art-known lyophilization and reconstitution techniques can be employed. It will be appreciated by those skilled in the art that lyophilization and reconstitution can lead to varying degrees of antibody activity loss (e.g. , with conventional immune globulins, IgM antibodies tend to have greater activity loss than IgG antibodies) and that use levels may have to be adjusted to compensate.
The compositions containing the present human chimeric antibodies or a cocktail thereof can be administered for the prophylactic and/or therapeutic treatments. In therapeutic application, compositions are administered to a patient already, in an amount sufficient to cure or at least partially arrest the infection and its complications. An amount adequate to accomplish this is defined as a "therapeutically effective dose." Amounts effective for this use will depend upon the severity of the infection and the general state of the patient's own immune system, but generally range from about 1 to about 200 mg of antibody per dose, with dosages of from 5 to 25 mg per patient being more commonly used. It must be kept in mind that the materials of this invention may generally be employed in serious disease states, that is life-threatening or potentially life- threatening situations. In such cases, in view of the minimization of extraneous substances and the lower probability of "foreign substance" rejections which are achieved by the present human chimeric antibodies of this in- vention, it is possible and may be felt desirable by the treating physician to administer substantial excesses of these antibodies.
In prophylactic applications, compositions containing the present antibodies or a cocktail thereof are administered to a patient not already in a disease state to enhance the patient's resistance. Such an amount is defined to be a "prophylactically effective dose." In this use, the precise amounts again depend upon the patient's state of health and general level of immunity, but generally range from 0.1 to 25 mg per dose, especially 0.5 to 2.5 mg per patient. Single or multiple administrations of the compositions can be carried out with dose levels and pattern being selected by the treating physician. In any event, the pharmaceutical formulations should provide a quantity of the antibody(ies) of this invention sufficient to effectively treat the patient.
Chimeric antibodies of the present invention can further find a wide variety of utilities vitro. By way of example, the chimeric antibodies can be utilized for T-cell typing, for isolating specific IL-2 receptor bearing cells or fragments of the receptor, for vaccine preparation, or the like.
For diagnostic purposes, the chimeric antibodies may either be labeled or unlabeled. Unlabeled antibodies can be used in combination with other labeled antibodies (second antibodies) that are reactive with the chimeric antibody, such as antibodies specific for human immunoglobulin constant regions. Alternatively, the chimeric antibodies can be directly labeled. A wide variety of labels may be employed, such as radionuclides, fluorescers, enzymes, enzyme substrates, enzyme cofactors, enzyme inhibitors, ligands
(particularly haptens) , etc. Numerous types of immunoassays are available and are well known to those skilled in the art. Kits can also be supplied for use with the subject antibodies in the protection against or detection of a cellular activity or for the presence of a selected antigen. Thus, the subject antibody composition of the present invention may be provided, usually in a lyophilized form in a container, either alone or in conjunction with additional antibodies specific for the desired cell type. The antibodies, which may be conjugated to a label or toxin, or unconjugated, are included in the kits with buffers, such as Tris, phosphate, carbonate, etc., stabilizers, biocides, inert proteins, e.g. , serum albumin, or the like, and a set of instructions for use. Generally, these materials will be present in less than about 5% wt. based on the amount of active antibody, and usually present in total amount of at least about 0.001% wt. based again on the antibody concentration. Frequently, it will be desirable to include an inert extender or excipient to dilute the active ingredients, where the excipient may be present in from about 1 to 99% wt. of the total composition. Where a second antibody capable of binding to the chimeric antibody is employed in an assay, this will usually be present in a separate vial. The second antibody is typically conjugated to a label and formulated in an analogous manner with the antibody formulations described above.
The following examples are offered by way of illustration, not by limitation.
EXPERIMENTAL
Construction of Plasmids
1. Construction began with the plasmid pKcatH described in the literature (Garcia, J.V. et al., Nature 332:383-385 (1986) ; Fig. 3) . This plasmid has the following parts; clockwise: a. A 700 bp fragment containing the mouse immunoglobulin heavy chain enhancer (E„ in the Fig. 3). b. An 1100 base pair (bp) fragment from the mouse immunoglobulin light chain kappa gene MOPC 41, containing its promoter. There is a Bgl II site 25 bp after the transcription startsite of this promoter. c. The bacterial CAT gene (800 bp) . d. A splice and polyadenylation signal from the animal virus SV40 (850 bp) . e. Another part of SV40 containing its origin of replication (700 bp) ; f. Part of the plasmid pBR322, extending from the Sph I site to the Eco Rl site of the plasmid pMLl (Lusky, M and Botchan, M. Nature 293:79-81: 2300 bp) including the Amp gene and origin of replication.
2. pKcatH was cut with Bgl II, and the ends filled in with Klenow polymerase. An Xba I linker, having the sequence GCTCTAGAGC, was inserted at the filled-in Bgl II site. The resulting plasmid is called pKcatH - Xba.
3. pKcatH - Xba was cut partially with Xho II, run on an agarσse gel, and full-length linear plasmid (i.e. , plasmid cut exactly once with Xho II) was isolated. The DNA was cut with Bam HI, run on a gel and the 5600 bp fragment isolated. Because of its size, this fragment had to extend from the Bam site to the Xho II site at the end of the CAT gene (Fig. 3). The fragment was ligated by itself. The resulting plasmid is called pKcatH - Xba - BX. It is similar to pKcatH but has an Xba I site in place of the Bgl II site and is missing the Xho II - Bam HI SV40 fragment (Fig. 4) .
4. pKcatH - Xba - BX was cut partially with Eco RI, run on an agarose gel, and full-length linear plasmid was isolated. This DNA was cut with Bam HI, and the 2600 bp fragment isolated. This fragment extended from the Eco Rl site before E„ to the Bam HI site. The plasmid pSV2gpt, described in the literature (Mulligan, R.C. and Berg, P., Proc. Nat. Acad. Sci. USA 78:2072-2976 (1980)), was cut with Eco Rl and Bam HI and the large Eco RI - Bam HI fragment ligated to the Eco RI - Bam HI fragment from pKcatH - Xba - BX. The resulting plasmid is called pSV2gpt - E„ - K , (Fig. 5) .
5. The plasmid pSV2neo (Southern, P.J. and Berg, P. J. Mol. App. Genet. 1:327-341 (1982)), was cut with Eco RI and Bam HI, and the large Eco RI - Bam HI fragment ligated to the same Eco RI - Bam HI fragment from pKcatH - Xba - BX used in (4) . The resulting plasmid is called pSV2neo - EH - K , which is like pSV2gpt - EH - K, but has the Neo gene in place of the Gpt gene.
6. pSV2gpt - E„ - c was cut with Xba I and Bam HI and the ends filled in with Klenow polymerase. A fragment of the cloned human K constant segment gene (Hieter, P.A. et al., Cell 22:197-207 (1980)) was purified, extending from a Hind III site 336 bp before the coding region, to an Xba I site about 800 bp beyond the coding region, and the ends filled in. The two fragments were ligated together, and a plasmid selected in which the Hind III site of the second*fragment was joined to the Xba I site and the Xba I site of the second fragment to the Bam HI site. Because of the sequences of these sites, this recreated an Xba I site and a Bam HI site. The new plasmid is called pVκl (Fig. 6) . 7. An Xba I fragment containing the VJ region of the cloned anti-Tac light chain gene was prepared by in vitro mutagenesis (see, below) . pVκl was cut with Xba I, treated with phosphatase, and ligated with the Xba I fragment. A plasmid was selected in which the VJ region had the same orientation as the following C region, and called pLTAC2 (Fig. 7) .
8. pSV2neo - E„ - K was cut with Xba I and Bam HI and the ends filled in with Klenow polymerase. A 2800 bp fragment containing the human C7I gene was purified from the phage HG3A (Ellison, J.W. et al., Nucleic Acids Res. 10:4071-4079 (1982)), extending from a Hind III site 210 bp before the CHI exon to a Pvu II site about 1100 bp after the CH3 exon, and the ends filled in. The two fragments were ligated together, and a plasmid selected in which the Hind III site of the second fragment was joined to the Xba I site and the Pvu II site of the second fragment to the Bam HI site. Because of the sequences of these sites, this recreated an Xba I site and a Bam HI site. The new plasmid is called pV7lneo (Fig. 8) .
9. pSV2neo - E„ - K was cut with Xba I and Bam HI and the ends filled in with Klenow polymerase. A 3600 bp fragment containing the human C73 gene (Takahashi, N. et al., Cell ϊ671-679 (1982)) was purified extending from a Hind III site 210 bp before the CHI exon to a Pvu II site about 1100 bp after the CH3 exon, and the ends filled in. The two fragments were ligated together, and a plasmid selected in which the Hind III site of the second fragment was joined to the Xba I site and the Pvu II site of the second fragment to the Bam HI site. Because of the sequences of these sites, this recreated an Xba I site and a Bam HI site. The new plasmid is called pV 3neo and is identical to pV7lneo except it has the C73 gene instead of C7I. 10. An Xba I fragment containing the VJ region of the cloned anti-Tac heavy china gene was prepared by in vitro mutagenesis (see, below) . pV7lneo was cut with Xba I, treated with phosphatase, and ligated with the Xba I fragment. A plasmid was selected in which the VJ region had the same orientation as the following C region, and called pHTAC.
11. Three additional plasmids were prepared respectively from pV7lneo, pV 3neo and pHTAC, called respectively PV7I, PV73 and pGTACl. In each case, the original plasmid was cut with Hind III and Bam HI, and the large Hind III - Bam HI fragment purified. The plasmid pXBohph containing the Hyg gene (Blochlinger, K. and Diggelmann, H. Mol. Cell. Biol. 4:2929-2931 (1984)) was cut with Hind III and Bam HI and a 1600 bp fragment containing the Hyg gene purified. The fragments from the original plasmids were each ligated to the pXbohph fragment. PV7I is shown in Fig. 9 and pGTACl in Fig.
10.
12. pHTAC was cut with Xba I and the small Xba I fragment, containing the heavy VJ region, purified. PV 3 was cut with Xba I, treated with phosphatase, and ligated with the small Xba I fragment. A plasmid was selected in which the VJ region had the same orientation as the following C region, and called pGTAC3. It is similar to pGTACl, but has the C73 region instead of the C7I region.
Cloning of Light and Heavy Chain cDNA. mRNA was extracted from approximately 10-8 ascites cells of the anti-Tac hybrido a by the guanidium isothiocyanate method followed with poly A selection on Hybond- AP paper (Amersham) . cDNA was prepared by the method of Gubler and Hoffman (Gubler, U. and Hoffman, B.J. Gene
25:263-269 (1983)), treated with Eco Rl methylase, ligated to Eco RI linkers, cloned into λgtlO arms (Promega Biotec) , packaged (Strategene packaging extract) and plated on C600Hfl cells. Oligonucleotides respectively 43 and 37 nucleotides long that hybridized to the 5' ends of the mouse C and C7I segments were synthesized (Applied Biosystems Model 38OB DNA synthesizer) . The oligonucleotides were 5' end-labelled and used to screen the plaques (Maniatis, et al. , Molecular Cloning A Laboratory Manual, Cold Spring Harbor Press, (1982) , which is incorporated herein by reference) .
Approximately 10,000 plaques were screened with the oligonucleotide probes. About 100 plaques were positive for the 7 probe and 40 for the 7 probe.
The cDNA inserts from 4 K positive and 4 72a positive phage were subcloned into the Eco Rl sites of pUC19 and M13mpl9. Partial sequencing by the dideoxy method (Sanger, F. et al., Proc. Nat. Acad. Sci. USA 74:5463-5467 (1977) ) showed that two of the K isolates had one sequence, and the other two had another sequence. In one pair, a tz V gene segment was joined to the JK2 segment out of its reading frame. In addition, the conserved Cys at position 23 was absent in this V segment, and the sequences of the two isolates differed slightly. Presumably, these clones were the result of an aberrant joining event in one K allele, which continued to undergo somatic mutation after the formation of the hybridoma.
The VJ segments of the other pair of K clones were sequences completely and were identical (Fig. 1) . This light chain uses the JK5 segment and contains all the amino acid residues conserved in mouse K light chains (Kabat, E. A. et al.. Sequences of Proteins of Immunological Interest, particularly pg. 45 et seq, all of which is incorporated herein by reference) .
Partial sequencing of the four 72a clones showed they were from the same gene. The VJ segments of two were sequenced completely and had no differences. This heavy chain (Fig. 2) uses the JH2 segment and is of subgroup II (Kabat, E.A. et al., supra, at pgs. 121-127). The sequence is somewhat unusual because it contains no detectable D segment and has an N region containing 10 Gs. As both alleles of the K light chain were accounted for and only one heavy chain sequence was detected, we tentatively assigned these sequences to the anti-Tac antibody genes.
Preparation of the Chimeric Genes
Plasmid vectors were prepared for the construction and expression of chimeric light and heavy chain genes as described above. The plasmid pVκl (Fig. 6) contains the human K genomic C segment, including 336 bp of the preceding intron and the poly A signal. It also contains a strong promoter sequence from the MOPC 41 K gene, and the heavy chain enhancer sequence. There is a unique Xba I site between the promoter and the C intron. The plasmid also contains the gpt gene for selection. Two other very similar plasmids were prepared by using the human 71 and 73 C regions in place of the K C region. In each case, the region inserted between the Xba I and Bam HI sites extended form about 210 bp 5* of the CHI exon to about 1100 bp 3 ' of the CH3 exon. In addition, the gpt gene was replaced with the hyg gene to confer resistance to hygromycin.
Our strategy was to insert the VJ region from the anti-Tac K cDNA, followed by a splice donor signal, at the Xba I site of pV/cl. Doing so would create a chimeric K gene, with a synthetic intron between the mouse VJ and human C segments. For this purpose, we devised a form of primer- directed mutagenesis (Fig. 11) . The K Eco Rl cDNA clone was moved into a variant of M13mpll, in which the Xba 1 and Eco Rl sites in the polylinker were abuting. An oligonucleotide was synthesized, of which the first 22 residues hybridized to the last 22 bp of the JK5 segment. The next 16 nucleotides were the same as the sequence that follows JK5 in mouse genomic DNA, and therefore included a splice donor signal. The final nucleotides of the oligo consisted of an Xba I site followed by a short irrelevant sequence.
This oligonucleotide was hybridized to the Ml3 phage DNA containing the K CDNA and extended with Klenow polymerase (Fig. 3) . The DNA was denatured and hybridized to a "reverse primer," which represents M13 DNA 5' to the cDNA insert. The reverse primer was extended, and the DNA cut with Xba I. The Xba I fragment consisting of the extended VJ segment of the cDNA was purified, and cloned in the correction orientation into the Xba I site of pVκl to obtain the plasmid pLTAC2 (Fig. 7) . Hence, the final chimeric K gene has a VJ-C intron, of which the first 14 bp and last 209 bp are respectively the same as in mouse and human genomic DNA, as verified by direct sequencing of the construct. The gene is transcribed from a K promoter, stimulated by the heavy chain enhancer. Based on results obtained with deleted introns, we expected that the intron would be correctly spliced from the transcribed RNA. In an analogous manner, the VJ region from the anti-Tac 72a heavy chain cDNA, followed by a splice donor signal, was inserted into the Xba I site of pV7lneo. The resulting plasmid pHTAC contains a chimeric heavy chain gene, with a synthetic intron between the mouse VJ and human C7I segments. The additional plasmids pGTACl and pGTAC3, containing the Hyg gene and chimeric heavy genes with the human C7I C73 constant regions respectively, were constructed as described above (Fig. 10) . Details of Insertion of VJ cDNA Regions Into Plasmids
RF DNA of the phage M13mpll was cut with Eco Rl and Xba I, the ends were filled-in, and the DNA ligated was ligated and transformed into JM101 cells. A plaque was picked and the DNA was sequenced to verify that the ends of the DNA had joined correctly, recreating the Eco Rl and Xba I sites with the intervening DNA segment deleted. This phage is designated M13mpllD. Eco Rl fragments containing the anti-Tac light and heavy chain cDNAs were separately cloned into the Eco Rl site of M13mpll, so that their 5' ends abuted the Xba I site. The resulting phage are respectively denoted M13mpllL and M13mpllH.
The following 48-nucleotide long primer was synthesized and gel-purified: CCAGAATTCTAGAAAAGTGTACTTACGTTTCAGCTCCAGCTTGGTCCC. From the 3' end, the first 22 residues of the primer are the same as the last 22 bp of the JK5 segment (non-coding strand) . The next 16 nucleotides are the same as the sequence that follows JK5 in mouse genomic DNA and therefore includes a splice donor signal (abbreviated SD in Fig. 11) . The final nucleotides of the oligo consist of an Xba I site followed by a short irrelevant sequence. Approximately 1 ug of single- stranded M13mpllL DNA was mixed with 50 ng of primer in 35 ul of 10 mM Tris, pH 7.4, 60 mM NaCl, 10 mM MgCl2 and incubated for 15 min at 50XC and then 15 min at 23λC. 4 ul of a solution of 200 uM each dNTP was added, together with 5u
Klenow polymerase. The solution was incubated for 30 minutes at 37XC. 50 ng of the "reverse primer" AACAGCTATGACCATG (New England Biolabs) , which can hybridize to the newly synthesized strand upstream of the Xba I site (Fig. 11) , was added. The solution was incubated at 95λC for 3 min and put on ice. An additional 4 ul of 200 uM each dNTP and 5u Klenow polymerase was added, and the solution incubated for 30 minutes at 37 C. The solution was extracted with phenol- chloroform, precipitated with ethanol, resuspended, and digested with 20u Xba I. The digested DNA was run on a 4% polyacrylamide gel and visualized with ethidiu bromide. In addition to high molecular weight DNA, an approximately 400 bp fragment was visible, corresponding to the VJ region of the light chain cDNA with a "tail" (Fig. 11) . The fragment was cloned directly into the Xba I site of pVκl in the correct orientation.
The following 50-nucleotide long primer was synthesized and gel-purified:
CCAGAATTCTAGAGGTTTTAAGGACTCACCTGAGGAGACTGTGAGAGTGG. From the 3 • end, the first 21 residues of the primer are the same as the last 21 bp of the JH2 segment (non-coding strand) . The next 19 nucleotides are the same as the sequence that follows JH2 in mouse genomic DNA and therefore includes a splice donor signal. The final nucleotides of the oligo consist of an Xba I site followed by a short irrelevant sequence. This primer was hybridized to M13mpllH DNA following the same protocol as above, in order to synthesize a fragment containing the VJ region of the heavy chain cDNA. The fragment was cloned directly into the Xba I site of pV7lneo in the correct orientation.
Chimeric Antibody Specificity CR2-2 and CEM cells are human T cell lines that are respectively positive and negative for surface expression of the IL2 receptor (IL2R) . Whole CR2-2 cells were used to demonstrate binding specificity of the chimeric antibody in an ELISA assay, and CEM cells were used as a negative control.
Antibody to be tested was prepared in several ways. Anti-Tac monoclonal antibody, supplied by T. Waldmann, was purified from mouse ascites by passage through a DEAE-dextran column. The cell line L40H4, created by transfecting SP2/0 cells with the chimeric light and gamma 1 heavy chain plasmids pLTAC2 and pGTACl (described above) , was injected into mice to form an ascites. From 3 mis of ascites fluid, 650 μg of chimeric gamma 1 antibody was purified by passage through a column of Baker Abx and a size exclusion column. Also, to detect antibody production in tissue culture, 106 cells each of L40H4 and the cell lines 51.3 and L40H2 (which had been created by transfecting SP2/0 with pLTAC2 and the gamma 3 heavy chain plasmid pGTAC3 described before) and the non-producing line Sp2/0 itself, were plated in 1 ml of DME medium each. The media supernatent, containing any secreted chimeric antibodies, was harvested after 24 hr. and used directly in the assay, Cultured cells of each type were washed in FACS buffer (0.1% BSA, .01% sodium azide in Dulbecco's phosphate buffered saline) . 2 x 106 cells were mixed with either purified antibody in 20 μl FACS buffer or 20 μl media supernatent from the transfected cell lines, and incubated on ice for 2 hr. The cells were washed 3 times with 1 μl of FACS buffer (being collected by brief centrifugation after each wash) . The cells that had been incubated with anti-Tac antibody (or no antibody as a control) were mixed with 0.5 μl peroxidase-conjugated goat anti-mouse antibody (Fab) ' fragment (Tago Immunologicals, Burlingame, California) in 20 μl FACS buffer, and the cells that had been incubated with chimeric antibody were mixed with 0.5 μl peroxidase- conjugated goat anti-human gamma chain (Fab) ' fragment (Tago) in 20 μl FACS buffer. The cells were incubated for 30 min on ice and then washed 3 times with FACS buffer. They were then mixed with 100 μl peroxidase development solution and incubated for 5 min at room temperature. The cells were spun out and the supernatents were transferred to a 96-well plate and the OD's determined in an ELISA reader. The OD's at 414 nm for the cells treated with each antibody are given in Table l.
As expected the anti-Tac antibody itself bound to the IL2R+ CR2-2 cells but not to the IL2R- CEM cells. An equal amount of purified gamma 1 chimeric antibody gave an equivalent amount of binding to the CR2-2 cells as the anti- Tac, and also failed to bind to the CEM cells. All the supernatants from both the chimeric antibody producing cells bound to the CR2-2 cells. As additional negative controls, the 51.3 supernatant did not bind to the CEM cells, and supernatant from the parental SP2/0 cells did not bind the CR2-2 cells.
In combination with the derivation of the light and heavy chain genes from the anti-Tac hybridoma, these results show that the chimeric antibodies retained specificity for the human IL2R.
TABLE 1
Whole-Cell ELISA Assay of Chimeric Antibodies
CR2-2 cells CEM cells (IL2R +) (IL2R -)
Mouse Gamma 2a none .037 anti-Tac (lOOng) .679 .024
Human Gamma 1 none .084 purified ascites (lOOng) .641 .044
L40H4 (20 μl sup.) .481
Human Gamma 3
51.3 (20 μl sup.) .381 .038
L40H2 (20 μl sup.) 932
To quantitate the rate of secretion of the chimeric antibodies, an ELISA assay was performed, using standard methods. The results are presented in Table 2. About 106 cells each of the chimeric gamma 1 secreting line L40H0 and the chimeric gamma 3 secreting line 51.3 were plated in 1 ml of DME medium with 5% fetal calf serum, and the supernatents collected after 24 hr. A 96-well plate was coated with goat anti-human antibodies (Tago Immunologicals) . Different wells were incubated with increasing known amounts of gamma 1 chimeric antibody (purified from ascites) and with 1 μl of the chimeric antibody supernatents. Briefly, the plate was washed, incubated with peroxidase-conjugated goat anti-human gamma chain antibody, washed again, incubated with peroxidase developing solution and the OD's at 414 nm determined in an ELISA reader. By comparison with the standard curve from the purified antibody, the media supernatants contained respectively about 8 and 7 ng per μl, that is the cells secreted 8 and 7 μg antibody per 106 cells per 24 hr.
TABLE 2
Rate of Chimeric Antibody Secretion
Figure imgf000033_0001
Purified gamma 1 chimeric
3 ng .426 4 ng .535
6 ng .734
8 ng 1.004
L40H4 (1 μl sup.) 1.014 51.3 (1 μl sup.) .845
The mixed lymphocyte reaction is a model for transplant rejection and was used to analyze the effectiveness of the chimeric anti-Tac antibodies. The experiments shown in Table 3 below were performed by standard methods (see, e.g. , Strong, D.M. et al., In Vitro Stimulation of Murine Spleen Cells Using a Microculture System and a .
Multiple Automated Sample Harvester, J. Immunol. Meth.. :279 (1973)). Briefly, human peripheral blood lymphocytes from two unrelated human donors were purified. The lymphocytes from the two donors were incubated in wells of a 96-well plate, either separately, mixed together, or mixed together with 1 μg/ml of the indicated antibody added each day. When mixed together, the T-cells recognize each other as foreign, as T cells recognize an organ transplant as foreign. They therefore proliferate, as measured by uptake of 3H-labeled thymidine after three days. The numbers in Table 3 are the average thymidine uptake in cpm from triplicate wells + the standard deviation. Addition of either the original anti-Tac antibody or either of the chimeric antibodies strongly inhibits proliferation of the cells (percent inhibition shown in parentheses) , by binding to the IL2R. This prevents IL2 from binding to the IL2R, which is required for T-cell proliferation. These experiments show that the chimeric antibody can be used to reduce the response of T-cells to foreign cells and may therefore have medical applications in preventing transplant rejection.
TABLE 3
Anti-IL2R Antibody Inhibition of Mixed Lymphocyte Reaction
Exp. 1 Exp. 2
Donor A 510 + 292. 151 ± 5
Donor B 901 ± 38 712 ± 29
A + B 11740 + 1593 19806 ± 2034 A + B + anti-Tac 4262 ± 346 (73%) 7311 ± 461 (66%)
A + B + Human Gamma 1 4126 ± 370 (74%) 6721 ± 981 (69%)
A + B + Human Gamma 3 - 6423 + 423 (71%)
From the foregoing, it will be appreciated that the chimeric antibodies of the present invention offer numerous advantages of other human IL-2 receptor-specific antibodies. In comparison to anti-Tac mouse monoclonal antibodies, the present human chimeric antibodies can be more economically produced and contain substantially less foreign amino acid sequences. This reduced likelihood of antigenicity after injection into a human patient represents a significant therapeutic improvement.
Although the present invention has been described in some detail by way of illustration and example for purposes of clarity and understanding, it will be apparent that certain changes and modifications may be practiced within the scope of the appended claims.

Claims

1. A method of treating T-cell mediated disorders in a human patient, said method comprising administering to said patient a therapeutically effective dose of a mouse variable/human constant region chimeric antibody composition specifically reactive with IL-2 receptors on human T-cells.
2. A method according to Claim 1, wherein the mouse variable region comprises the entire variable region from heavy and light chains of an anti-Tac monoclonal antibody as produced by the cell line designated A.T.C.C. Accession No. CRL 9688.
3. A method according to Claim 1, wherein at least one of the mouse variable regions is joined with a naturally-associated mouse J segment.
4. A method according to Claim 1, wherein the human light chain constant region comprises a K chain constant region.
5. A method according to Claim 1, wherein the human light chain constant region comprises a 7 or >γ chain constant region.
6. A method according to Claim 1, wherein the composition comprises the chimeric antibody complexed with a cytotoxic agent.
7. A method according to Claim 1, wherein the cytotoxic agent is a ribosomal inhibiting protein, a radionuclide or cytotoxic agent active at cell surfaces.
8. A chimeric antibody capable of binding to a human IL-2 receptor epitope, said antibody having two pairs of light chain/heavy chain complexes, wherein at least one pair has chains comprising mouse variable region and human constant region segments.
5
9. A chimeric antibody according to Claim 8, wherein the mouse variable region is adjacent to a naturally associated J segment.
0 10. A chimeric antibody according to Claim 8, which inhibits the binding of monoclonal antibodies secreted by the cell line designated A.T.C.C. Accession No. CRL 9688.
11. A chimeric antibody according to Claim 10, 5 wherein at least one variable region comprises the amino acid sequence of a variable region of the anti-Tac monoclonal antibody.
12. A chimeric antibody according to Claim 8, which 0 was produced in a myeloma or hybridoma cell.
13. A chimeric antibody according to Claim 8, which was expressed in a myeloma cell from a transfected DNA sequence comprising a mouse cDNA segment joined to a human genomic DNA segment.
14. A chimeric antibody according to Claim 8 "complexed with a cytotoxic agent or signal agent.
JU 15. A heavy immunoglobulin chain comprising a human heavy chain constant region and a variable chain region which is substantially identical to a monoclonal antibody heavy chain variable region secreted by the cell line designated A.T.C.C. Accession No. CRL 9688. 35
16. _heavy immunoglobulin chain according to Claim 15, wherein the variable and constant region are joined by a mouse J segment.
17. A light immunoglobulin chain comprising a human light chain constant region and a variable chain region which is substantially identical to a monoclonal antibody light chain variable region secreted by the cell line designated A.T.C.C. Accession No. CRL 9688.
18. A polynucleotide molecule comprising a first sequence coding for a human immunoglobulin constant region and a second sequence coding for a mouse immunoglobulin variable region, wherein said second sequence encodes substantially for one of the amino acid sequences of Figures 1 or 2.
19. A cell line transfected with a polynucleotide of Claim 18.
20. The cell line designated A.T.C.C. Accession No. CRL 9688.
21. A DNA segment encoding a portion of an immunoglobulin light chain, said segment comprising at least about 30 contiguous nucleotides from the sequence of Figure 1.
22. A DNA segment according to Claim 21, wherein the segment encodes a full length light chain variable region substantially homologous to the sequence in Figure 1.
23. A DNA segment encoding a portion of an immunoglobulin heavy chain, said segment comprising at least about 30 contiguous nucleotides from the sequence of Figure 2.
24. A DNA segment according to Claim 23, wherein the segment encodes a full length heavy chain variable region substantially homologous to the sequence in Figure 2.
25. An expression vector comprising a heterologous promoter operably linked to a DNA segment according to Claim 21 or 23.
26. An immortalized cell line transformed with an expression vector according to Claim 25.
27. A protein composition comprising at least about ten contiguous amino acids from the protein sequence of Figure 1 or Figure 2 fused to a heterologous polypeptide.
28. A protein composition according to Claim 27, wherein the heterologous polypeptide is an immunoglobulin constant region.
29. A protein composition according to Claim 27, wherein the protein is glycosylated.
30. A recombinantly produced antibody exhibiting substantially the same antigen binding profile as an immunoglobulin secreted by a cell line designated A.T.C.C. Accession No. CRL 9688.
PCT/US1989/001578 1988-04-15 1989-04-13 Il-2 receptor-specific chimeric antibodies WO1989009622A1 (en)

Priority Applications (4)

Application Number Priority Date Filing Date Title
KR1019890702368A KR900700134A (en) 1988-04-15 1989-04-13 IL-2 Receptor-Specific Chimeric Antibodies
FI895955A FI895955A0 (en) 1988-04-15 1989-12-13 IL-2 RECEPTOR SPECIFICATION CHIMERA ANTIKROPPAR.
NO89895022A NO895022L (en) 1988-04-15 1989-12-14 IL-2 RECEPTOR SPECIFIC CHIMER ANTIBODIES.
DK631689A DK631689A (en) 1988-04-15 1989-12-14 IL-2 RECEPTOR-SPECIFIC CHEMICAL ANTIBODIES

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US18186288A 1988-04-15 1988-04-15
US181,862 1988-04-15

Publications (1)

Publication Number Publication Date
WO1989009622A1 true WO1989009622A1 (en) 1989-10-19

Family

ID=22666118

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US1989/001578 WO1989009622A1 (en) 1988-04-15 1989-04-13 Il-2 receptor-specific chimeric antibodies

Country Status (6)

Country Link
EP (1) EP0362371A4 (en)
JP (1) JPH02503867A (en)
KR (1) KR900700134A (en)
AU (1) AU631545B2 (en)
FI (1) FI895955A0 (en)
WO (1) WO1989009622A1 (en)

Cited By (160)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0380068A1 (en) * 1989-01-24 1990-08-01 Miles Inc. An expression system for production of chimeric monoclonal antibodies
FR2649488A1 (en) * 1989-07-07 1991-01-11 Inst Nat Sante Rech Med FRAGMENTS OF MONOCLONAL ANTIBODIES SPECIFIC TO THE PRESENCE OF ACTIVE LEUKOCYTES - THEIR PROCESS FOR OBTAINING AND THEIR APPLICATION IN THE CASE OF A GRAFT REJECT
EP0421876A1 (en) * 1989-10-06 1991-04-10 PASTEUR MERIEUX SERUMS ET VACCINS, Société Anonyme : Use of active agents in the preparation of a drug for the treatment of lymphomes or related diseases
FR2652747A1 (en) * 1989-10-06 1991-04-12 Merieux Inst Application of active agents for the preparation of a medicament intended for the treatment of autoimmune diseases
EP0449769A1 (en) * 1990-03-16 1991-10-02 Sandoz Ltd. CD 25 binding molecules
EP0451216A1 (en) * 1988-12-28 1991-10-16 Protein Design Labs Inc CHIMERIC IMMUNOGLOBULINS SPECIFIC FOR p55 TAC PROTEIN OF THE IL-2 RECEPTOR.
EP0460674A2 (en) * 1990-06-08 1991-12-11 Roche Diagnostics GmbH Recombinant DNA and process for the production of chimaeric antibodies
WO1992004051A1 (en) * 1990-09-12 1992-03-19 Boehringer Mannheim Gmbh Monoclonal antibody active against the interleukin-2 receptor
GR900100009A (en) * 1990-01-09 1992-06-25 Protein Design Labs Particular human immunoglobulin receiver of il-2 new type
US5196320A (en) * 1989-09-20 1993-03-23 Abbott Biotech, Inc. Method of producing engineered binding proteins
US5621083A (en) * 1991-11-04 1997-04-15 Xoma Corporation Immunotoxins comprising ribosome-inactivating proteins
WO1997017374A1 (en) * 1995-11-08 1997-05-15 Medac Gesellschaft Für Klinische Spezialpräparate Gmbh Recombinant ligands for the human cell membrane antigen cd30
WO1997028828A1 (en) 1996-02-09 1997-08-14 Amgen Boulder Inc. Composition comprising interleukin-1 inhibitor and controlled release polymer
US5766886A (en) * 1991-12-13 1998-06-16 Xoma Corporation Modified antibody variable domains
US5837491A (en) * 1991-11-04 1998-11-17 Xoma Corporation Polynucleotides encoding gelonin sequences
US5869619A (en) * 1991-12-13 1999-02-09 Xoma Corporation Modified antibody variable domains
US6013256A (en) * 1996-09-24 2000-01-11 Protein Design Labs, Inc. Method of preventing acute rejection following solid organ transplantation
WO2000006604A2 (en) * 1998-07-27 2000-02-10 Novartis Ag Use of cd25 binding molecules in the treatment of rheumatoid arthritis or skin diseases
US6054297A (en) * 1991-06-14 2000-04-25 Genentech, Inc. Humanized antibodies and methods for making them
US6146850A (en) * 1991-11-04 2000-11-14 Xoma Corporation Proteins encoding gelonin sequences
WO2000069459A1 (en) 1999-05-14 2000-11-23 Imclone Systems Incorporated Treatment of refractory human tumors with epidermal growth factor receptor antagonists
WO2001002853A2 (en) 1999-07-05 2001-01-11 K.U. Leuven Research & Development DETECTION OF VON-WILLEBRANDFACTOR (vWF) ACTIVITY
US6180370B1 (en) 1988-12-28 2001-01-30 Protein Design Labs, Inc. Humanized immunoglobulins and methods of making the same
US6407213B1 (en) 1991-06-14 2002-06-18 Genentech, Inc. Method for making humanized antibodies
WO2004002421A2 (en) 2002-06-28 2004-01-08 The Government Of The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services Method for the treatment of multiple sclerosis
US6800738B1 (en) 1991-06-14 2004-10-05 Genentech, Inc. Method for making humanized antibodies
US6890532B2 (en) 2000-05-16 2005-05-10 Thomas Jefferson University Rabies virus-specific neutralizing human monoclonal antibodies and nucleic acids and related methods
WO2005042581A2 (en) 2003-11-01 2005-05-12 Biovation Ltd. Modified anti-cd52 antibody
WO2005116657A2 (en) 2004-05-24 2005-12-08 Universität Zu Köln Identification of ergothioneine transporter and therapeutic uses thereof
WO2006021893A2 (en) 2004-08-26 2006-03-02 The University Of Western Ontario Pharmaceutical compositions comprising inhibitors of iron transport, and method of identifying iron transport inhibitors, fn staphylococcus aureus
WO2006034507A2 (en) 2004-09-24 2006-03-30 Beth Israel - Deaconess Medical Center Methods of diagnosing and treating complications of pregnancy
US7071319B2 (en) 2000-05-16 2006-07-04 Thomas Jefferson University Recombinant antibodies, and compositions and methods for making and using the same
WO2006083355A2 (en) 2004-11-19 2006-08-10 Cornell Research Foundation, Inc. Use of vascular endothelial growth factor receptor 1+ cells in treating and monitoring cancer and in screening for chemotherapeutics
US7098006B1 (en) 1990-09-17 2006-08-29 Burroughs Wellcome Co. Chimeric antibody, pharmaceutical composition and process of its production
WO2006105448A2 (en) 2005-03-30 2006-10-05 Minerva Biotechnologies Corporation Proliferation of muc1 expressing cells
EP1724282A2 (en) 1997-05-21 2006-11-22 Biovation Limited Method for the production of non-immunogenic proteins
WO2007027751A2 (en) 2005-08-30 2007-03-08 University Of Miami Immunomodulating tumor necrosis factor receptor 25 (tnfr25) agonists, antagonists and immunotoxins
US7252823B2 (en) * 1991-03-18 2007-08-07 Centocor, Inc. Recombinant A2-specific TNFα-specific antibodies
WO2008054603A2 (en) 2006-10-02 2008-05-08 Amgen Inc. Il-17 receptor a antigen binding proteins
EP1923702A2 (en) 2004-06-04 2008-05-21 University of Geneva Novel means and methods for the treatment of hearing loss and phantom hearing
WO2008081008A1 (en) 2007-01-05 2008-07-10 University Of Zurich Method of providing disease-specific binding molecules and targets
WO2008110372A1 (en) 2007-03-13 2008-09-18 University Of Zurich Monoclonal human tumor-specific antibody
WO2008118324A2 (en) 2007-03-26 2008-10-02 Macrogenics, Inc. Composition and method of treating cancer with an anti-uroplakin ib antibody
US7442778B2 (en) 2004-09-24 2008-10-28 Amgen Inc. Modified Fc molecules
EP1998266A2 (en) 2001-02-19 2008-12-03 Merck Patent GmbH Method for identification of T-cell epitopes and use for preparing molecules with reduced immunogenicity
EP2002846A2 (en) 1996-12-06 2008-12-17 Amgen Inc. Combination therapy using an IL-1 inhibitor for treating IL-1 mediated diseases
WO2009014835A2 (en) 2007-06-21 2009-01-29 Angelica Therapeutics, Inc. Modified toxins
EP2070947A1 (en) 2003-12-05 2009-06-17 multimmune GmbH Therapeutic and diagnostic anti-Hsp70 antibodies
EP2100614A2 (en) 2005-06-17 2009-09-16 Imclone LLC PDGFR-alpha antagonists for treatment of metastatic bone cancer
WO2010027364A1 (en) 2008-09-07 2010-03-11 Glyconex Inc. Anti-extended type i glycosphingolipid antibody, derivatives thereof and use
EP2172220A1 (en) 2004-02-04 2010-04-07 Beth Israel Deaconess Medical Center Methods of diagnosing and treating pre-eclampsia or eclampsia
US7740841B1 (en) 2000-01-28 2010-06-22 Sunnybrook Health Science Center Therapeutic method for reducing angiogenesis
WO2010069603A1 (en) 2008-12-19 2010-06-24 Neurimmune Therapeutics Ag Human anti-alpha-synuclein autoantibodies
US7744885B2 (en) 1991-03-18 2010-06-29 Centocor, Inc. Methods of treating vascular inflammatory pathology using anti-TNF antibodies and fragments thereof
WO2011015379A1 (en) 2009-08-05 2011-02-10 Nexigen Gbmh Human hcv-interacting proteins and methods of use
EP2287339A1 (en) 2004-05-18 2011-02-23 Georg Dewald Methods and kits to detect hereditary angioedema type III
WO2011024114A1 (en) 2009-08-25 2011-03-03 Ecole Polytechnique Federale De Lausanne (Epfl) Targeting extracellular matrix molecules for the treatment of cancer
EP2305301A2 (en) 2002-07-19 2011-04-06 Beth Israel Deaconess Medical Center Methods of diagnosing and treating pre-eclampsia or eclampsia
EP2305285A1 (en) 2009-09-29 2011-04-06 Julius-Maximilians-Universität Würzburg Means and methods for treating ischemic conditions
WO2011039724A1 (en) 2009-10-02 2011-04-07 Sanofi-Aventis Antibodies that specifically bind to the epha2 receptor
WO2011046958A1 (en) 2009-10-12 2011-04-21 Amgen Inc. Use of il-17 receptor a antigen binding proteins
US7951370B2 (en) 2008-03-12 2011-05-31 Imclone Llc Anti-TYRP1 antibodies
EP2332990A1 (en) 2004-03-19 2011-06-15 Imclone LLC Human anti-epidermal growth factor receptor antibody
WO2011100403A1 (en) 2010-02-10 2011-08-18 Immunogen, Inc Cd20 antibodies and uses thereof
WO2011103389A1 (en) 2010-02-19 2011-08-25 Cornell University Method to treat autoimmune demyelinating diseases and other autoimmune or inflammatory diseases
US8008007B2 (en) 2005-04-14 2011-08-30 Rwth Aachen S-adenosyl-L-methionine analogs with extended activated groups for transfer by methyltransferases
EP2363410A1 (en) 2002-11-27 2011-09-07 Minerva Biotechnologies Corporation Isoforms of MUC1
EP2365001A2 (en) 2003-05-01 2011-09-14 Imclone LLC Fully human antibodies directed against the human insulin-like growth factor-1 receptor
WO2011123381A1 (en) 2010-04-01 2011-10-06 Imclone Llc Antibodies against csf-1r
EP2377555A2 (en) 2004-11-18 2011-10-19 Imclone LLC Antibodies against vascular endothelial growth factor receptor-1
US8063182B1 (en) 1989-09-12 2011-11-22 Hoffman-Laroche Inc. Human TNF receptor fusion protein
WO2011145085A2 (en) 2010-05-21 2011-11-24 Procognia (Israel) Ltd Novel antibodies and methods of use for the treatment and diagnosis of cancer
US8071099B2 (en) 2008-05-30 2011-12-06 ImClone, LLC Anti-FLT3 antibodies
DE202011103324U1 (en) 2011-07-12 2012-01-02 Nekonal S.A.R.L. Therapeutic anti-TIRC7 antibodies for use in immune and other diseases
WO2012025636A1 (en) 2010-08-27 2012-03-01 University Of Zurich Method for target and drug validation in inflammatory and/or cardiovascular diseases
WO2012028697A1 (en) 2010-09-01 2012-03-08 Eth Zürich, Institute Of Molecular Biology And Biophysics Affinity purification system based on donor strand complementation
EP2433966A1 (en) 2006-11-03 2012-03-28 U3 Pharma GmbH FGFR4 antibodies
US8153765B2 (en) 2006-10-19 2012-04-10 Sanof Aventis Anti-CD38 antibodies for the treatment of cancer
WO2012049570A1 (en) 2010-10-11 2012-04-19 Panima Pharmaceuticals Ag Human anti-tau antibodies
WO2012052230A1 (en) 2010-10-18 2012-04-26 Mediapharma S.R.L. Erbb3 binding antibody
WO2012080518A1 (en) 2010-12-17 2012-06-21 Neurimmune Holding Ag Human anti-sod1 antibodies
WO2012104824A1 (en) 2011-02-04 2012-08-09 Ecole polytechnique fédérale de Lausanne (EPFL) Therapeutic antibodies targeting app-c99
WO2012113775A1 (en) 2011-02-21 2012-08-30 University Of Zurich Ankyrin g and modulators thereof for the treatment of neurodegenerative disorders
WO2012125775A1 (en) 2011-03-16 2012-09-20 Sanofi Uses of a dual v region antibody-like protein
EP2527369A2 (en) 2007-09-13 2012-11-28 University Of Zurich Prorektorat Forschung Monoclonal amyloid beta (abeta)-specific antibody and uses thereof
EP2530088A1 (en) 2011-05-30 2012-12-05 Klinikum rechts der Isar der Technischen Universität München Means and methods for diagnosing and treating multiple sclerosis
WO2012177972A1 (en) 2011-06-23 2012-12-27 Biogen Idec International Neuroscience Gmbh Anti-alpha synuclein binding molecules
WO2013016220A1 (en) 2011-07-22 2013-01-31 Amgen Inc. Il-17 receptor a is required for il-17c biology
US8388965B2 (en) 2007-10-15 2013-03-05 Sanofi Antibodies that bind IL-4 and/or IL-13 and their uses
WO2013041962A1 (en) 2011-09-19 2013-03-28 Axon Neuroscience Se Protein-based therapy and diagnosis of tau-mediated pathology in alzheimer's disease
WO2013050540A1 (en) 2011-10-05 2013-04-11 University Of Bremen Wnt4 and med12 for use in the diagnosis and treatment of tumor diseases
US8460667B2 (en) 2006-07-18 2013-06-11 Sanofi EPHA2 receptor antagonist antibodies
EP2602621A1 (en) 2011-12-08 2013-06-12 Julius-Maximilians-Universität Würzburg LASP-1, a novel urinary marker for transitional cell carcinoma detection
WO2013093122A2 (en) 2011-12-23 2013-06-27 Phenoquest Ag Antibodies for the treatment and diagnosis of affective and anxiety disorders
WO2013098419A1 (en) 2011-12-28 2013-07-04 Immunoqure Ag Method of providing monoclonal auto-antibodies with desired specificity
WO2013149526A1 (en) 2012-04-01 2013-10-10 上海益杰生物技术有限公司 Multi-functional antibody polypeptide for cryptic epitope of epidermal growth factor receptor and t cell antigen
US8647622B2 (en) 2007-08-29 2014-02-11 Sanofi Humanized anti-CXCR5 antibodies, derivatives thereof and their use
EP2711016A1 (en) 2012-09-21 2014-03-26 Covagen AG Novel IL-17A binding molecules and medical uses thereof
DE102012020496A1 (en) 2012-10-18 2014-04-24 Charité - Universitätsmedizin Berlin Invitro diagnosis and/or early detection of neurofibromatosis type 1 and/or neurofibromatosis type 1-associated tumor from sample of a patient for treating the patient with the tumor, comprises detecting protein or its fragment in sample
WO2014102399A1 (en) 2012-12-31 2014-07-03 Neurimmune Holding Ag Recombinant human antibodies for therapy and prevention of polyomavirus-related diseases
WO2014180306A1 (en) 2013-05-08 2014-11-13 上海益杰生物技术有限公司 Nucleic acid of coded gpc3 chimeric antigen receptor protein and t lymphocyte expressing gpc3 chimeric antigen receptor protein
US8980864B2 (en) 2013-03-15 2015-03-17 Moderna Therapeutics, Inc. Compositions and methods of altering cholesterol levels
US8999380B2 (en) 2012-04-02 2015-04-07 Moderna Therapeutics, Inc. Modified polynucleotides for the production of biologics and proteins associated with human disease
WO2015106641A1 (en) 2014-01-15 2015-07-23 上海易乐生物技术有限公司 Anti-human probdnf monoclonal antibody, and uses thereof in pains
US9107886B2 (en) 2012-04-02 2015-08-18 Moderna Therapeutics, Inc. Modified polynucleotides encoding basic helix-loop-helix family member E41
US9114175B2 (en) 2005-08-12 2015-08-25 Amgen Inc. Modified Fc molecules
EP2918605A1 (en) 2007-11-12 2015-09-16 U3 Pharma GmbH Axl antibodies
US9150650B2 (en) 2007-06-13 2015-10-06 Pharmabcine Inc. Human monoclonal antibody neutralizing vascular endothelial growth factor receptor and use thereof
WO2015153144A1 (en) 2014-03-31 2015-10-08 Kirin-Amgen, Inc. Methods of treating nail and scalp psoriasis
US9163086B2 (en) 2009-08-18 2015-10-20 President And Fellows Of Harvard College Methods and compositions for the treatment of proliferative and pathogenic diseases
US9181319B2 (en) 2010-08-06 2015-11-10 Moderna Therapeutics, Inc. Engineered nucleic acids and methods of use thereof
US9186372B2 (en) 2011-12-16 2015-11-17 Moderna Therapeutics, Inc. Split dose administration
WO2015198146A2 (en) 2014-06-27 2015-12-30 Sanofi Anti-il4-il 13 bispecific antibodies
US9283287B2 (en) 2012-04-02 2016-03-15 Moderna Therapeutics, Inc. Modified polynucleotides for the production of nuclear proteins
WO2016050822A2 (en) 2014-09-30 2016-04-07 Neurimmune Holding Ag Human-derived anti-dipeptide repeats (dprs) antibody
US9334328B2 (en) 2010-10-01 2016-05-10 Moderna Therapeutics, Inc. Modified nucleosides, nucleotides, and nucleic acids, and uses thereof
WO2016081796A1 (en) 2014-11-21 2016-05-26 Yale University Compositions and methods for modulating salm5 and hvem
US9428535B2 (en) 2011-10-03 2016-08-30 Moderna Therapeutics, Inc. Modified nucleosides, nucleotides, and nucleic acids, and uses thereof
WO2016140910A2 (en) 2015-03-04 2016-09-09 University Of Rochester Compositions and methods of using anti-mullerian hormone for treatment of infertility
WO2016149621A1 (en) 2015-03-18 2016-09-22 The Johns Hopkins University Novel monoclonal antibody inhibitors targeting potassium channel kcnk9
WO2016146702A1 (en) 2015-03-16 2016-09-22 Helmholtz Zentrum München - Deutsches Forschungszentrum für Gesundheit und Umwelt (GmbH) Trispecific binding molecules for treating hbv infection and associated conditions
US9464124B2 (en) 2011-09-12 2016-10-11 Moderna Therapeutics, Inc. Engineered nucleic acids and methods of use thereof
US9533047B2 (en) 2011-03-31 2017-01-03 Modernatx, Inc. Delivery and formulation of engineered nucleic acids
US9572897B2 (en) 2012-04-02 2017-02-21 Modernatx, Inc. Modified polynucleotides for the production of cytoplasmic and cytoskeletal proteins
US9592289B2 (en) 2012-03-26 2017-03-14 Sanofi Stable IgG4 based binding agent formulations
US9597380B2 (en) 2012-11-26 2017-03-21 Modernatx, Inc. Terminally modified RNA
EP3202788A1 (en) 2016-02-05 2017-08-09 MediaPharma S.r.l. Endosialin-binding antibody
WO2017167967A1 (en) 2016-04-01 2017-10-05 Istituto Biochimico Italiano Giovanni Lorenzini S.P.A. Erbb2-targeting antibody
EP3269740A1 (en) 2016-07-13 2018-01-17 Mabimmune Diagnostics AG Novel anti-fibroblast activation protein (fap) binding agents and uses thereof
US9937205B2 (en) 2012-09-04 2018-04-10 The Trustees Of The University Of Pennsylvania Inhibition of diacylglycerol kinase to augment adoptive T cell transfer
WO2018083282A1 (en) 2016-11-07 2018-05-11 Biouniversa S.R.L. Anti-bag3 antibodies in combination with inhibitors of immune check-point for therapeutic use
WO2018129029A1 (en) 2017-01-04 2018-07-12 Immunogen, Inc. Met antibodies and immunoconjugates and uses thereof
US10059750B2 (en) 2013-03-15 2018-08-28 Angelica Therapeutics, Inc. Modified toxins
US10072085B2 (en) 2010-01-15 2018-09-11 Kirin-Amgen, Inc. Method of treating psoriasis using an IL-17 receptor antibody formulation
WO2018172465A1 (en) 2017-03-22 2018-09-27 Sanofi Treatment of lupus using humanized anti-cxcr5 antibodies
WO2018185050A1 (en) 2017-04-03 2018-10-11 Covagen Ag Fgfr3 binding molecules
US10183965B2 (en) 2014-08-29 2019-01-22 Katholieke Universiteit Leuven Cofactor analogues for methyltransferases
US10316037B1 (en) 2016-11-04 2019-06-11 Yale University Compounds and methods for treating cancer
US10323076B2 (en) 2013-10-03 2019-06-18 Modernatx, Inc. Polynucleotides encoding low density lipoprotein receptor
US10345298B2 (en) 2014-04-30 2019-07-09 Klinikum Rechts Der Isar Der Technischen Universität München Diagnosis of multiple sclerosis
US10377822B2 (en) 2014-07-17 2019-08-13 Carsgen Therapeutics Ltd. Immunologic effector cell of targeted CLD18A2, and preparation method and use thereof
WO2019207159A1 (en) 2018-04-27 2019-10-31 Fondazione Ebri Rita Levi-Montalcini Antibody directed against a tau-derived neurotoxic peptide and uses thereof
WO2020014306A1 (en) 2018-07-10 2020-01-16 Immunogen, Inc. Met antibodies and immunoconjugates and uses thereof
WO2020053808A1 (en) 2018-09-12 2020-03-19 Georg Dewald Method of diagnosing vasoregulatory disorders
WO2020099235A1 (en) 2018-11-12 2020-05-22 Mediapharma S.R.L. Bispecific antibodies directed against human 90k and either endosialin or her3
US10703821B2 (en) 2005-03-30 2020-07-07 Minerva Biotechnologies Corporation Method for stimulating or enhancing proliferation of cells by activating the mucin 1 (MUC1) receptor
WO2020157122A1 (en) 2019-01-29 2020-08-06 Helmholtz Zentrum München - Deutsches Forschungszentrum für Gesundheit und Umwelt (GmbH) Treating the causative agent in adhesiogenesis
EP3693387A1 (en) 2015-11-05 2020-08-12 Intrepida Bio, Inc. Humanized anti-bag3 antibodies
WO2020167912A1 (en) 2019-02-13 2020-08-20 The Brigham And Women's Hospital, Inc. Anti-peripheral lymph node addressin antibodies and uses thereof
EP3699284A1 (en) 2012-07-05 2020-08-26 The Trustees of the University of Pennsylvania U1 snrnp regulates gene expression and modulates oncogenicity
US10815291B2 (en) 2013-09-30 2020-10-27 Modernatx, Inc. Polynucleotides encoding immune modulating polypeptides
WO2020242989A1 (en) 2019-05-24 2020-12-03 Sanofi Methods for treating systemic sclerosis
EP3792278A2 (en) 2012-12-21 2021-03-17 Biogen MA Inc. Human anti-tau antibodies
WO2021140173A1 (en) 2020-01-10 2021-07-15 Biouniversa S.R.L. Methods and uses for treating fibrotic solid tumors with bags inhibitors
WO2021183207A1 (en) 2020-03-10 2021-09-16 Massachusetts Institute Of Technology COMPOSITIONS AND METHODS FOR IMMUNOTHERAPY OF NPM1c-POSITIVE CANCER
US11155638B2 (en) 2018-05-08 2021-10-26 Rhode Island Hospital Anti-CHI3L1 antibodies for the detection and/or treatment of nonalcoholic fattly liver disease/nonalcoholic steatonhepatitis and subsequent complications
EP4056591A1 (en) 2013-07-03 2022-09-14 ImmunoQure AG Human anti-ifn-alpha antibodies
WO2022189632A1 (en) 2021-03-12 2022-09-15 Fibrosys S.R.L. Monoclonal antibodies for the treatment of viral infections
US11773182B2 (en) 2017-01-05 2023-10-03 The Johns Hopkins University Development of new monoclonal antibodies recognizing human prostate-specific membrane antigen (PSMA)
US11898160B2 (en) 2008-10-09 2024-02-13 Minerva Biotechnologies Corporation Method for maintaining pluripotency in cells

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0173494A2 (en) * 1984-08-27 1986-03-05 The Board Of Trustees Of The Leland Stanford Junior University Chimeric receptors by DNA splicing and expression
EP0184187A2 (en) * 1984-12-04 1986-06-11 Teijin Limited Mouse-human chimaeric immunoglobulin heavy chain, and chimaeric DNA encoding it
US4664911A (en) * 1983-06-21 1987-05-12 Board Of Regents, University Of Texas System Immunotoxin conjugates employing toxin B chain moieties
EP0125023B1 (en) * 1983-04-08 1991-06-05 Genentech, Inc. Recombinant immunoglobulin preparations, methods for their preparation, dna sequences, expression vectors and recombinant host cells therefor

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US664911A (en) * 1900-09-17 1901-01-01 William H Voss Mechanical movement.
GB2188941B (en) * 1986-04-14 1990-06-06 Bayer Ag Monoclonal antibodies recognizing human interleukin-2-receptor

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0125023B1 (en) * 1983-04-08 1991-06-05 Genentech, Inc. Recombinant immunoglobulin preparations, methods for their preparation, dna sequences, expression vectors and recombinant host cells therefor
US4664911A (en) * 1983-06-21 1987-05-12 Board Of Regents, University Of Texas System Immunotoxin conjugates employing toxin B chain moieties
EP0173494A2 (en) * 1984-08-27 1986-03-05 The Board Of Trustees Of The Leland Stanford Junior University Chimeric receptors by DNA splicing and expression
EP0184187A2 (en) * 1984-12-04 1986-06-11 Teijin Limited Mouse-human chimaeric immunoglobulin heavy chain, and chimaeric DNA encoding it

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
See also references of EP0362371A4 *
The Journal of Immunology, Vol. 126, No. 4, issued April 1981 (USA), UCHIYAMA et al, "A Monoclonal Antibody (ANTI-Tac) Reactive with Activated and Functionally Mature Human T Cells," see pages 1393 to 1397. *

Cited By (308)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7022500B1 (en) 1988-12-28 2006-04-04 Protein Design Labs, Inc. Humanized immunoglobulins
EP1477497B1 (en) * 1988-12-28 2014-11-26 PDL BioPharma, Inc. Method for the production of humanized immunoglobulins
EP0451216A1 (en) * 1988-12-28 1991-10-16 Protein Design Labs Inc CHIMERIC IMMUNOGLOBULINS SPECIFIC FOR p55 TAC PROTEIN OF THE IL-2 RECEPTOR.
EP0451216A4 (en) * 1988-12-28 1992-01-02 Protein Design Labs, Inc. Chimeric immunoglobulins specific for p55 tac protein of the il-2 receptor
US6180370B1 (en) 1988-12-28 2001-01-30 Protein Design Labs, Inc. Humanized immunoglobulins and methods of making the same
EP0380068A1 (en) * 1989-01-24 1990-08-01 Miles Inc. An expression system for production of chimeric monoclonal antibodies
FR2649488A1 (en) * 1989-07-07 1991-01-11 Inst Nat Sante Rech Med FRAGMENTS OF MONOCLONAL ANTIBODIES SPECIFIC TO THE PRESENCE OF ACTIVE LEUKOCYTES - THEIR PROCESS FOR OBTAINING AND THEIR APPLICATION IN THE CASE OF A GRAFT REJECT
WO1991000921A1 (en) * 1989-07-07 1991-01-24 Institut National De La Sante Et De La Recherche Medicale Fragments of monoclonal antibodies specific to the presence of activated leucocytes, process for obtaining them and their application in the case of a transplant being rejected
US8163522B1 (en) 1989-09-12 2012-04-24 Hoffman-Laroche Inc. Human TNF receptor
US8063182B1 (en) 1989-09-12 2011-11-22 Hoffman-Laroche Inc. Human TNF receptor fusion protein
US5196320A (en) * 1989-09-20 1993-03-23 Abbott Biotech, Inc. Method of producing engineered binding proteins
US5338669A (en) * 1989-09-20 1994-08-16 Abbott Biotech, Incorporated Method of producing fusion proteins
FR2652746A1 (en) * 1989-10-06 1991-04-12 Merieux Inst APPLICATION OF ACTIVE AGENTS FOR THE PREPARATION OF A MEDICINAL PRODUCT FOR THE TREATMENT OF LYMPHOMA OR THE LIKE.
FR2652747A1 (en) * 1989-10-06 1991-04-12 Merieux Inst Application of active agents for the preparation of a medicament intended for the treatment of autoimmune diseases
EP0421876A1 (en) * 1989-10-06 1991-04-10 PASTEUR MERIEUX SERUMS ET VACCINS, Société Anonyme : Use of active agents in the preparation of a drug for the treatment of lymphomes or related diseases
GR900100009A (en) * 1990-01-09 1992-06-25 Protein Design Labs Particular human immunoglobulin receiver of il-2 new type
US6521230B1 (en) 1990-03-16 2003-02-18 Novartis Ag CD25 binding molecules
EP0449769A1 (en) * 1990-03-16 1991-10-02 Sandoz Ltd. CD 25 binding molecules
US6383487B1 (en) * 1990-03-16 2002-05-07 Novartis Ag Methods of treatment using CD25 binding molecules
EP0460674A3 (en) * 1990-06-08 1992-08-19 Boehringer Mannheim Gmbh Recombinant dna and process for the production of chimaeric antibodies
EP0460674A2 (en) * 1990-06-08 1991-12-11 Roche Diagnostics GmbH Recombinant DNA and process for the production of chimaeric antibodies
WO1992004051A1 (en) * 1990-09-12 1992-03-19 Boehringer Mannheim Gmbh Monoclonal antibody active against the interleukin-2 receptor
US6538110B1 (en) 1990-09-12 2003-03-25 Roche Diagnostics Monoclonal antibodies against the interleukin 2 receptor
US7098006B1 (en) 1990-09-17 2006-08-29 Burroughs Wellcome Co. Chimeric antibody, pharmaceutical composition and process of its production
USRE46877E1 (en) 1990-09-17 2018-05-29 Btg International Limited Altered antibodies and their preparation
USRE43898E1 (en) 1990-09-17 2013-01-01 Glaxo Wellcome Inc. Research Altered antibodies and their preparation
US7252823B2 (en) * 1991-03-18 2007-08-07 Centocor, Inc. Recombinant A2-specific TNFα-specific antibodies
US7276239B2 (en) 1991-03-18 2007-10-02 Centocor, Inc. Recombinant A2-specific TNFα-specific antibodies
US7744885B2 (en) 1991-03-18 2010-06-29 Centocor, Inc. Methods of treating vascular inflammatory pathology using anti-TNF antibodies and fragments thereof
US6800738B1 (en) 1991-06-14 2004-10-05 Genentech, Inc. Method for making humanized antibodies
US6054297A (en) * 1991-06-14 2000-04-25 Genentech, Inc. Humanized antibodies and methods for making them
US6719971B1 (en) 1991-06-14 2004-04-13 Genentech, Inc. Method for making humanized antibodies
US6639055B1 (en) 1991-06-14 2003-10-28 Genentech, Inc. Method for making humanized antibodies
US8075890B2 (en) 1991-06-14 2011-12-13 Genentech, Inc. Method for making humanized antibodies
US6407213B1 (en) 1991-06-14 2002-06-18 Genentech, Inc. Method for making humanized antibodies
US6146850A (en) * 1991-11-04 2000-11-14 Xoma Corporation Proteins encoding gelonin sequences
US5837491A (en) * 1991-11-04 1998-11-17 Xoma Corporation Polynucleotides encoding gelonin sequences
US5744580A (en) * 1991-11-04 1998-04-28 Xoma Corporation Immunotoxins comprising ribosome-inactivating proteins
US5621083A (en) * 1991-11-04 1997-04-15 Xoma Corporation Immunotoxins comprising ribosome-inactivating proteins
US6146631A (en) * 1991-11-04 2000-11-14 Xoma Corporation Immunotoxins comprising ribosome-inactivating proteins
US6649742B1 (en) 1991-11-04 2003-11-18 Xoma Technology Ltd. Immunotoxins comprising ribosome-inactivating proteins
US6376217B1 (en) 1991-11-04 2002-04-23 Xoma Technology Ltd. Fusion proteins and polynucleotides encoding gelonin sequences
US5756699A (en) * 1991-11-04 1998-05-26 Xoma Corporation Immunotoxins comprising ribosome-inactivating proteins
US5766886A (en) * 1991-12-13 1998-06-16 Xoma Corporation Modified antibody variable domains
US5869619A (en) * 1991-12-13 1999-02-09 Xoma Corporation Modified antibody variable domains
US5770196A (en) * 1991-12-13 1998-06-23 Xoma Corporation Modified antibody variable domains and therapeutic uses thereof
US5821123A (en) * 1991-12-13 1998-10-13 Xoma Corporation Modified antibody variable domains
WO1997017374A1 (en) * 1995-11-08 1997-05-15 Medac Gesellschaft Für Klinische Spezialpräparate Gmbh Recombinant ligands for the human cell membrane antigen cd30
WO1997028828A1 (en) 1996-02-09 1997-08-14 Amgen Boulder Inc. Composition comprising interleukin-1 inhibitor and controlled release polymer
US6013256A (en) * 1996-09-24 2000-01-11 Protein Design Labs, Inc. Method of preventing acute rejection following solid organ transplantation
EP2002846A2 (en) 1996-12-06 2008-12-17 Amgen Inc. Combination therapy using an IL-1 inhibitor for treating IL-1 mediated diseases
EP1724282A2 (en) 1997-05-21 2006-11-22 Biovation Limited Method for the production of non-immunogenic proteins
WO2000006604A3 (en) * 1998-07-27 2000-06-15 Novartis Ag Use of cd25 binding molecules in the treatment of rheumatoid arthritis or skin diseases
WO2000006604A2 (en) * 1998-07-27 2000-02-10 Novartis Ag Use of cd25 binding molecules in the treatment of rheumatoid arthritis or skin diseases
WO2000069459A1 (en) 1999-05-14 2000-11-23 Imclone Systems Incorporated Treatment of refractory human tumors with epidermal growth factor receptor antagonists
EP2042194A2 (en) 1999-05-14 2009-04-01 Imclone Systems, Inc. Treatment of refractory human tumors with epidermal growth factor receptor antagonists
EP2309275A1 (en) 1999-07-05 2011-04-13 K.U.Leuven Research & Development Detection of Von-Willebrand factor (vwf) activity
WO2001002853A2 (en) 1999-07-05 2001-01-11 K.U. Leuven Research & Development DETECTION OF VON-WILLEBRANDFACTOR (vWF) ACTIVITY
US7740841B1 (en) 2000-01-28 2010-06-22 Sunnybrook Health Science Center Therapeutic method for reducing angiogenesis
EP2301579A1 (en) 2000-01-28 2011-03-30 Sunnybrook Health Science Centre Therapeutic method for reducing angiogenesis
US7071319B2 (en) 2000-05-16 2006-07-04 Thomas Jefferson University Recombinant antibodies, and compositions and methods for making and using the same
US6890532B2 (en) 2000-05-16 2005-05-10 Thomas Jefferson University Rabies virus-specific neutralizing human monoclonal antibodies and nucleic acids and related methods
EP1998266A2 (en) 2001-02-19 2008-12-03 Merck Patent GmbH Method for identification of T-cell epitopes and use for preparing molecules with reduced immunogenicity
US7575742B2 (en) 2002-06-28 2009-08-18 The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services Method of treating autoimmune diseases with interferon-beta and IL-2R antagonist
EP3269377A1 (en) * 2002-06-28 2018-01-17 The Government of the U.S.A. as represented by The Secretary of the dept. of Health & Human Services Method for the treatment of multiple sclerosis
EP1539200A2 (en) * 2002-06-28 2005-06-15 The Government of the United States of America, as Represented by the Secretary, Department of Health and Human Services Method for the treatment of multiple sclerosis
US7258859B2 (en) 2002-06-28 2007-08-21 The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services Method for the treatment of multiple sclerosis
US8298525B2 (en) 2002-06-28 2012-10-30 The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services Method of treating multiple sclerosis with interferon-beta and an IL-2R antagonist
US8454965B2 (en) 2002-06-28 2013-06-04 The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services Method for the treatment of multiple sclerosis
EP1539200A4 (en) * 2002-06-28 2007-11-07 Us Gov Health & Human Serv Method for the treatment of multiple sclerosis
WO2004002421A2 (en) 2002-06-28 2004-01-08 The Government Of The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services Method for the treatment of multiple sclerosis
EP2314627A3 (en) * 2002-06-28 2011-05-11 The Government of the U.S.A. as represented by The Secretary of the dept. of Health & Human Services Method for the treatment of multiple sclerosis
EP2305301A2 (en) 2002-07-19 2011-04-06 Beth Israel Deaconess Medical Center Methods of diagnosing and treating pre-eclampsia or eclampsia
EP2308507A2 (en) 2002-07-19 2011-04-13 Beth Israel Deaconess Medical Center Methods of diagnosing and treating pre-eclampsia or eclampsia
EP2363410A1 (en) 2002-11-27 2011-09-07 Minerva Biotechnologies Corporation Isoforms of MUC1
EP2365001A2 (en) 2003-05-01 2011-09-14 Imclone LLC Fully human antibodies directed against the human insulin-like growth factor-1 receptor
WO2005042581A2 (en) 2003-11-01 2005-05-12 Biovation Ltd. Modified anti-cd52 antibody
EP2070947A1 (en) 2003-12-05 2009-06-17 multimmune GmbH Therapeutic and diagnostic anti-Hsp70 antibodies
EP2172220A1 (en) 2004-02-04 2010-04-07 Beth Israel Deaconess Medical Center Methods of diagnosing and treating pre-eclampsia or eclampsia
EP2332990A1 (en) 2004-03-19 2011-06-15 Imclone LLC Human anti-epidermal growth factor receptor antibody
EP2287339A1 (en) 2004-05-18 2011-02-23 Georg Dewald Methods and kits to detect hereditary angioedema type III
EP3056571A1 (en) 2004-05-18 2016-08-17 Georg Dewald Methods and kits to detect hereditary angioedema type iii
WO2005116657A2 (en) 2004-05-24 2005-12-08 Universität Zu Köln Identification of ergothioneine transporter and therapeutic uses thereof
EP1923702A2 (en) 2004-06-04 2008-05-21 University of Geneva Novel means and methods for the treatment of hearing loss and phantom hearing
WO2006021893A2 (en) 2004-08-26 2006-03-02 The University Of Western Ontario Pharmaceutical compositions comprising inhibitors of iron transport, and method of identifying iron transport inhibitors, fn staphylococcus aureus
US7655765B2 (en) 2004-09-24 2010-02-02 Amgen Inc. Modified Fc molecules
US7655764B2 (en) 2004-09-24 2010-02-02 Amgen Inc. Modified Fc molecules
US7645861B2 (en) 2004-09-24 2010-01-12 Amgen Inc. Modified Fc molecules
WO2006034507A2 (en) 2004-09-24 2006-03-30 Beth Israel - Deaconess Medical Center Methods of diagnosing and treating complications of pregnancy
EP2347765A1 (en) 2004-09-24 2011-07-27 Beth Israel Deaconess Medical Center Methods of diagnosing and treating complications of pregnancy
US7442778B2 (en) 2004-09-24 2008-10-28 Amgen Inc. Modified Fc molecules
US7662931B2 (en) 2004-09-24 2010-02-16 Amgen Inc. Modified Fc molecules
US8143025B2 (en) 2004-11-18 2012-03-27 Imclone Llc Antibodies against vascular endothelial growth factor receptor-1
EP2377555A2 (en) 2004-11-18 2011-10-19 Imclone LLC Antibodies against vascular endothelial growth factor receptor-1
WO2006083355A2 (en) 2004-11-19 2006-08-10 Cornell Research Foundation, Inc. Use of vascular endothelial growth factor receptor 1+ cells in treating and monitoring cancer and in screening for chemotherapeutics
US8859495B2 (en) 2005-03-30 2014-10-14 Minerva Biotechnologies Corporation Methods for stimulating or enhancing proliferation of non-tumorous cells expressing MUC1 receptors
WO2006105448A2 (en) 2005-03-30 2006-10-05 Minerva Biotechnologies Corporation Proliferation of muc1 expressing cells
EP2527365A2 (en) 2005-03-30 2012-11-28 Minerva Biotechnologies Corporation Proliferation of MUC1 expressing cells
EP2526957A2 (en) 2005-03-30 2012-11-28 Minerva Biotechnologies Corporation Proliferation of MUC1 expressing cells
US10703821B2 (en) 2005-03-30 2020-07-07 Minerva Biotechnologies Corporation Method for stimulating or enhancing proliferation of cells by activating the mucin 1 (MUC1) receptor
US8008007B2 (en) 2005-04-14 2011-08-30 Rwth Aachen S-adenosyl-L-methionine analogs with extended activated groups for transfer by methyltransferases
EP2100618A2 (en) 2005-06-17 2009-09-16 Imclone LLC PDGFR-alpha antagonists for treatment of metastatic bone cancer
EP2100614A2 (en) 2005-06-17 2009-09-16 Imclone LLC PDGFR-alpha antagonists for treatment of metastatic bone cancer
EP2505205A1 (en) 2005-06-17 2012-10-03 Imclone LLC Anti-PDGFR Alpha Antibodies
US10188740B2 (en) 2005-08-12 2019-01-29 Amgen Inc. Modified Fc molecules
US9114175B2 (en) 2005-08-12 2015-08-25 Amgen Inc. Modified Fc molecules
US11266744B2 (en) 2005-08-12 2022-03-08 Amgen Inc. Modified Fc molecules
WO2007027751A2 (en) 2005-08-30 2007-03-08 University Of Miami Immunomodulating tumor necrosis factor receptor 25 (tnfr25) agonists, antagonists and immunotoxins
USRE47123E1 (en) 2006-07-18 2018-11-13 Sanofi EPHA2 receptor antagonist antibodies
US8460667B2 (en) 2006-07-18 2013-06-11 Sanofi EPHA2 receptor antagonist antibodies
WO2008054603A2 (en) 2006-10-02 2008-05-08 Amgen Inc. Il-17 receptor a antigen binding proteins
US11180564B2 (en) 2006-10-02 2021-11-23 Amgen K-A, Inc. IL-17 Receptor A antigen binding proteins
US11858999B2 (en) 2006-10-02 2024-01-02 Amgen K-A, Inc. IL-17 receptor A antigen binding proteins
US10208122B2 (en) 2006-10-02 2019-02-19 Amgen K-A, Inc. IL-17 receptor A antigen binding proteins
EP3165539A1 (en) 2006-10-02 2017-05-10 Kirin-Amgen, Inc. Il-17 receptor a antigen binding proteins
EP3909980A1 (en) 2006-10-19 2021-11-17 Sanofi Novel anti-cd38 antibodies for the treatment of cancer
EP3498735A1 (en) 2006-10-19 2019-06-19 Sanofi Novel anti-cd38 antibodies for the treatment of leukemia
US8153765B2 (en) 2006-10-19 2012-04-10 Sanof Aventis Anti-CD38 antibodies for the treatment of cancer
EP2433966A1 (en) 2006-11-03 2012-03-28 U3 Pharma GmbH FGFR4 antibodies
EP3239175A1 (en) 2007-01-05 2017-11-01 University of Zurich Method of providing disease-specific binding molecules and targets
EP2426143A2 (en) 2007-01-05 2012-03-07 University of Zurich Method of providing disease-specific binding molecules and targets
EP3239174A1 (en) 2007-01-05 2017-11-01 University of Zurich Anti-beta-amyloid antibody and uses thereof
WO2008081008A1 (en) 2007-01-05 2008-07-10 University Of Zurich Method of providing disease-specific binding molecules and targets
EP2436696A1 (en) 2007-01-05 2012-04-04 University of Zurich Method of providing disease-specific binding molecules and targets
EP2457928A2 (en) 2007-03-13 2012-05-30 Universität Zürich Monoclonal human tumor-specific antibody
WO2008110372A1 (en) 2007-03-13 2008-09-18 University Of Zurich Monoclonal human tumor-specific antibody
EP3159355A1 (en) 2007-03-13 2017-04-26 Universität Zürich Monoclonal human tumor-specific antibody
US8519106B2 (en) 2007-03-13 2013-08-27 University Of Zurich Monoclonal human tumor-specific antibody
WO2008118324A2 (en) 2007-03-26 2008-10-02 Macrogenics, Inc. Composition and method of treating cancer with an anti-uroplakin ib antibody
US9150650B2 (en) 2007-06-13 2015-10-06 Pharmabcine Inc. Human monoclonal antibody neutralizing vascular endothelial growth factor receptor and use thereof
WO2009014835A2 (en) 2007-06-21 2009-01-29 Angelica Therapeutics, Inc. Modified toxins
US8252897B2 (en) 2007-06-21 2012-08-28 Angelica Therapeutics, Inc. Modified toxins
US9815902B2 (en) 2007-08-29 2017-11-14 Sanofi Humanized anti-CXCR5 antibodies, derivatives thereof and their uses
US8980262B2 (en) 2007-08-29 2015-03-17 Sanofi Humanized anti-CXCR5 antibodies, derivatives thereof and their use
US9243067B2 (en) 2007-08-29 2016-01-26 Sanofi Humanized anti-CXCR5 antibodies, derivatives thereof and their use
US9228019B2 (en) 2007-08-29 2016-01-05 Sanofi Humanized anti-CXCR5 antibodies, derivatives thereof and their use
US9175087B2 (en) 2007-08-29 2015-11-03 Sanofi Humanized anti-CXCR5 antibodies, derivatives thereof and their use
US8647622B2 (en) 2007-08-29 2014-02-11 Sanofi Humanized anti-CXCR5 antibodies, derivatives thereof and their use
EP2527369A2 (en) 2007-09-13 2012-11-28 University Of Zurich Prorektorat Forschung Monoclonal amyloid beta (abeta)-specific antibody and uses thereof
EP2573119A1 (en) 2007-10-15 2013-03-27 Sanofi Antibodies that bind IL-4 and/or IL-13 and their uses
EP2574630A1 (en) 2007-10-15 2013-04-03 Sanofi Antibodies that bind il-4 and/or il-13 and their uses
EP2573117A1 (en) 2007-10-15 2013-03-27 Sanofi Antibodies that bind IL-4 and/or IL-13 and their uses
EP2573118A1 (en) 2007-10-15 2013-03-27 Sanofi Antibodies that bind IL-4 and/or IL-13 and their uses
US9732162B2 (en) 2007-10-15 2017-08-15 Sanofi Antibodies that bind IL-4 and/or IL-13 and their uses
EP2573115A1 (en) 2007-10-15 2013-03-27 Sanofi Antibodies that bind IL-4 and/or IL-13 and their uses
EP2573116A1 (en) 2007-10-15 2013-03-27 Sanofi Antibodies that bind IL-4 and/or IL-13 and their uses
EP2574629A1 (en) 2007-10-15 2013-04-03 Sanofi Antibodies that bind il-4 and/or il-13 and their uses
EP2574626A1 (en) 2007-10-15 2013-04-03 Sanofi Antibodies that bind IL-4 and/or IL-13 and their uses
EP2573121A1 (en) 2007-10-15 2013-03-27 Sanofi Antibodies that bind il-4 and/or il-13 and their uses
US10759871B2 (en) 2007-10-15 2020-09-01 Sanofi Antibodies that bind IL-4 and/or IL-13 and their uses
EP3686220A1 (en) 2007-10-15 2020-07-29 Sanofi Antibodies that bind il-4 and/or il-13 and their uses
US11453727B2 (en) 2007-10-15 2022-09-27 Sanofi Antibodies that bind IL-4 and/or IL-13 and their uses
US9738728B2 (en) 2007-10-15 2017-08-22 Sanofi Antibodies that bind IL-4 and/or IL-13 and their uses
US8388965B2 (en) 2007-10-15 2013-03-05 Sanofi Antibodies that bind IL-4 and/or IL-13 and their uses
EP2918605A1 (en) 2007-11-12 2015-09-16 U3 Pharma GmbH Axl antibodies
US7951370B2 (en) 2008-03-12 2011-05-31 Imclone Llc Anti-TYRP1 antibodies
US8071099B2 (en) 2008-05-30 2011-12-06 ImClone, LLC Anti-FLT3 antibodies
WO2010027364A1 (en) 2008-09-07 2010-03-11 Glyconex Inc. Anti-extended type i glycosphingolipid antibody, derivatives thereof and use
US11898160B2 (en) 2008-10-09 2024-02-13 Minerva Biotechnologies Corporation Method for maintaining pluripotency in cells
EP3521309A1 (en) 2008-12-19 2019-08-07 Biogen International Neuroscience GmbH Human anti-alpha-synuclein antibodies
EP2949666A1 (en) 2008-12-19 2015-12-02 Biogen Idec International Neuroscience GmbH Human anti-alpha-synuclein antibodies
WO2010069603A1 (en) 2008-12-19 2010-06-24 Neurimmune Therapeutics Ag Human anti-alpha-synuclein autoantibodies
WO2011015379A1 (en) 2009-08-05 2011-02-10 Nexigen Gbmh Human hcv-interacting proteins and methods of use
US9163086B2 (en) 2009-08-18 2015-10-20 President And Fellows Of Harvard College Methods and compositions for the treatment of proliferative and pathogenic diseases
WO2011024114A1 (en) 2009-08-25 2011-03-03 Ecole Polytechnique Federale De Lausanne (Epfl) Targeting extracellular matrix molecules for the treatment of cancer
EP2305285A1 (en) 2009-09-29 2011-04-06 Julius-Maximilians-Universität Würzburg Means and methods for treating ischemic conditions
WO2011039282A1 (en) 2009-09-29 2011-04-07 Julius-Maximilians-Universität Würzburg Means and methods for treating ischemic conditions
US8668910B2 (en) 2009-10-02 2014-03-11 Sanofi Antibodies that specifically bind to the EphA2 receptor
WO2011039724A1 (en) 2009-10-02 2011-04-07 Sanofi-Aventis Antibodies that specifically bind to the epha2 receptor
US9676864B2 (en) 2009-10-02 2017-06-13 Sanofi Antibodies that specifically bind to the EphA2 receptor
WO2011046958A1 (en) 2009-10-12 2011-04-21 Amgen Inc. Use of il-17 receptor a antigen binding proteins
US10808033B2 (en) 2010-01-15 2020-10-20 Amgen K-A, Inc. IL-17 receptor antibody formulation
US11505612B2 (en) 2010-01-15 2022-11-22 Amgen K-A, Inc. Method of treating diseases using an IL-17 receptor antibody formulation
US10072085B2 (en) 2010-01-15 2018-09-11 Kirin-Amgen, Inc. Method of treating psoriasis using an IL-17 receptor antibody formulation
WO2011100403A1 (en) 2010-02-10 2011-08-18 Immunogen, Inc Cd20 antibodies and uses thereof
WO2011103389A1 (en) 2010-02-19 2011-08-25 Cornell University Method to treat autoimmune demyelinating diseases and other autoimmune or inflammatory diseases
WO2011123381A1 (en) 2010-04-01 2011-10-06 Imclone Llc Antibodies against csf-1r
EP3156419A1 (en) 2010-04-01 2017-04-19 ImClone LLC Antibodies against csf-1r
US8263079B2 (en) 2010-04-01 2012-09-11 ImClone, LLC Antibodies against CSF-1R
WO2011145085A2 (en) 2010-05-21 2011-11-24 Procognia (Israel) Ltd Novel antibodies and methods of use for the treatment and diagnosis of cancer
US9181319B2 (en) 2010-08-06 2015-11-10 Moderna Therapeutics, Inc. Engineered nucleic acids and methods of use thereof
US9447164B2 (en) 2010-08-06 2016-09-20 Moderna Therapeutics, Inc. Engineered nucleic acids and methods of use thereof
US9937233B2 (en) 2010-08-06 2018-04-10 Modernatx, Inc. Engineered nucleic acids and methods of use thereof
WO2012025636A1 (en) 2010-08-27 2012-03-01 University Of Zurich Method for target and drug validation in inflammatory and/or cardiovascular diseases
WO2012028697A1 (en) 2010-09-01 2012-03-08 Eth Zürich, Institute Of Molecular Biology And Biophysics Affinity purification system based on donor strand complementation
US10064959B2 (en) 2010-10-01 2018-09-04 Modernatx, Inc. Modified nucleosides, nucleotides, and nucleic acids, and uses thereof
US9334328B2 (en) 2010-10-01 2016-05-10 Moderna Therapeutics, Inc. Modified nucleosides, nucleotides, and nucleic acids, and uses thereof
US9701965B2 (en) 2010-10-01 2017-07-11 Modernatx, Inc. Engineered nucleic acids and methods of use thereof
US9657295B2 (en) 2010-10-01 2017-05-23 Modernatx, Inc. Modified nucleosides, nucleotides, and nucleic acids, and uses thereof
WO2012049570A1 (en) 2010-10-11 2012-04-19 Panima Pharmaceuticals Ag Human anti-tau antibodies
WO2012052230A1 (en) 2010-10-18 2012-04-26 Mediapharma S.R.L. Erbb3 binding antibody
EP3628689A1 (en) 2010-12-17 2020-04-01 Neurimmune Holding AG Human anti-sod1 antibodies
WO2012080518A1 (en) 2010-12-17 2012-06-21 Neurimmune Holding Ag Human anti-sod1 antibodies
WO2012104824A1 (en) 2011-02-04 2012-08-09 Ecole polytechnique fédérale de Lausanne (EPFL) Therapeutic antibodies targeting app-c99
WO2012113775A1 (en) 2011-02-21 2012-08-30 University Of Zurich Ankyrin g and modulators thereof for the treatment of neurodegenerative disorders
EP3235508A1 (en) 2011-03-16 2017-10-25 Sanofi Compositions comprising a dual v region antibody-like protein
WO2012125775A1 (en) 2011-03-16 2012-09-20 Sanofi Uses of a dual v region antibody-like protein
US11008389B2 (en) 2011-03-16 2021-05-18 Sanofi Uses of a dual V region antibody-like protein
US9950068B2 (en) 2011-03-31 2018-04-24 Modernatx, Inc. Delivery and formulation of engineered nucleic acids
US9533047B2 (en) 2011-03-31 2017-01-03 Modernatx, Inc. Delivery and formulation of engineered nucleic acids
EP2530088A1 (en) 2011-05-30 2012-12-05 Klinikum rechts der Isar der Technischen Universität München Means and methods for diagnosing and treating multiple sclerosis
WO2012163765A1 (en) 2011-05-30 2012-12-06 Klinikum Rechts Der Isar Der Technischen Universität München Means and methods for diagnosing and treating multiple sclerosis
WO2012177972A1 (en) 2011-06-23 2012-12-27 Biogen Idec International Neuroscience Gmbh Anti-alpha synuclein binding molecules
DE202011103324U1 (en) 2011-07-12 2012-01-02 Nekonal S.A.R.L. Therapeutic anti-TIRC7 antibodies for use in immune and other diseases
WO2013016220A1 (en) 2011-07-22 2013-01-31 Amgen Inc. Il-17 receptor a is required for il-17c biology
US10022425B2 (en) 2011-09-12 2018-07-17 Modernatx, Inc. Engineered nucleic acids and methods of use thereof
US10751386B2 (en) 2011-09-12 2020-08-25 Modernatx, Inc. Engineered nucleic acids and methods of use thereof
US9464124B2 (en) 2011-09-12 2016-10-11 Moderna Therapeutics, Inc. Engineered nucleic acids and methods of use thereof
EP3275461A1 (en) 2011-09-19 2018-01-31 Axon Neuroscience SE Protein-based therapy and diagnosis of tau-mediated pathology in alzheimer's disease field
WO2013041962A1 (en) 2011-09-19 2013-03-28 Axon Neuroscience Se Protein-based therapy and diagnosis of tau-mediated pathology in alzheimer's disease
US9428535B2 (en) 2011-10-03 2016-08-30 Moderna Therapeutics, Inc. Modified nucleosides, nucleotides, and nucleic acids, and uses thereof
WO2013050540A1 (en) 2011-10-05 2013-04-11 University Of Bremen Wnt4 and med12 for use in the diagnosis and treatment of tumor diseases
EP2602621A1 (en) 2011-12-08 2013-06-12 Julius-Maximilians-Universität Würzburg LASP-1, a novel urinary marker for transitional cell carcinoma detection
WO2013083690A1 (en) 2011-12-08 2013-06-13 Julius-Maximilians-Universität Würzburg Lasp-1, a novel urinary marker for transitional cell carcinoma detection
US9186372B2 (en) 2011-12-16 2015-11-17 Moderna Therapeutics, Inc. Split dose administration
US9295689B2 (en) 2011-12-16 2016-03-29 Moderna Therapeutics, Inc. Formulation and delivery of PLGA microspheres
US9271996B2 (en) 2011-12-16 2016-03-01 Moderna Therapeutics, Inc. Formulation and delivery of PLGA microspheres
WO2013093122A2 (en) 2011-12-23 2013-06-27 Phenoquest Ag Antibodies for the treatment and diagnosis of affective and anxiety disorders
EP3546481A2 (en) 2011-12-28 2019-10-02 ImmunoQure AG Anti-interleukin 22 (il-22) antibody and uses thereof
WO2013098419A1 (en) 2011-12-28 2013-07-04 Immunoqure Ag Method of providing monoclonal auto-antibodies with desired specificity
US9592289B2 (en) 2012-03-26 2017-03-14 Sanofi Stable IgG4 based binding agent formulations
US10525130B2 (en) 2012-03-26 2020-01-07 Sanofi Stable IGG4 based binding agent formulations
US10023639B2 (en) 2012-04-01 2018-07-17 Carsgen Therapeutics Co., Ltd. Multi-functional antibody polypeptide for cryptic epitope of epidermal growth factor receptor and T cell antigen
WO2013149526A1 (en) 2012-04-01 2013-10-10 上海益杰生物技术有限公司 Multi-functional antibody polypeptide for cryptic epitope of epidermal growth factor receptor and t cell antigen
US9572897B2 (en) 2012-04-02 2017-02-21 Modernatx, Inc. Modified polynucleotides for the production of cytoplasmic and cytoskeletal proteins
US10501512B2 (en) 2012-04-02 2019-12-10 Modernatx, Inc. Modified polynucleotides
US9675668B2 (en) 2012-04-02 2017-06-13 Moderna Therapeutics, Inc. Modified polynucleotides encoding hepatitis A virus cellular receptor 2
US8999380B2 (en) 2012-04-02 2015-04-07 Moderna Therapeutics, Inc. Modified polynucleotides for the production of biologics and proteins associated with human disease
US9782462B2 (en) 2012-04-02 2017-10-10 Modernatx, Inc. Modified polynucleotides for the production of proteins associated with human disease
US9050297B2 (en) 2012-04-02 2015-06-09 Moderna Therapeutics, Inc. Modified polynucleotides encoding aryl hydrocarbon receptor nuclear translocator
US9587003B2 (en) 2012-04-02 2017-03-07 Modernatx, Inc. Modified polynucleotides for the production of oncology-related proteins and peptides
US9061059B2 (en) 2012-04-02 2015-06-23 Moderna Therapeutics, Inc. Modified polynucleotides for treating protein deficiency
US9814760B2 (en) 2012-04-02 2017-11-14 Modernatx, Inc. Modified polynucleotides for the production of biologics and proteins associated with human disease
US9089604B2 (en) 2012-04-02 2015-07-28 Moderna Therapeutics, Inc. Modified polynucleotides for treating galactosylceramidase protein deficiency
US9828416B2 (en) 2012-04-02 2017-11-28 Modernatx, Inc. Modified polynucleotides for the production of secreted proteins
US9827332B2 (en) 2012-04-02 2017-11-28 Modernatx, Inc. Modified polynucleotides for the production of proteins
US9095552B2 (en) 2012-04-02 2015-08-04 Moderna Therapeutics, Inc. Modified polynucleotides encoding copper metabolism (MURR1) domain containing 1
US9107886B2 (en) 2012-04-02 2015-08-18 Moderna Therapeutics, Inc. Modified polynucleotides encoding basic helix-loop-helix family member E41
US9878056B2 (en) 2012-04-02 2018-01-30 Modernatx, Inc. Modified polynucleotides for the production of cosmetic proteins and peptides
US9114113B2 (en) 2012-04-02 2015-08-25 Moderna Therapeutics, Inc. Modified polynucleotides encoding citeD4
US9149506B2 (en) 2012-04-02 2015-10-06 Moderna Therapeutics, Inc. Modified polynucleotides encoding septin-4
US9192651B2 (en) 2012-04-02 2015-11-24 Moderna Therapeutics, Inc. Modified polynucleotides for the production of secreted proteins
US9303079B2 (en) 2012-04-02 2016-04-05 Moderna Therapeutics, Inc. Modified polynucleotides for the production of cytoplasmic and cytoskeletal proteins
US9216205B2 (en) 2012-04-02 2015-12-22 Moderna Therapeutics, Inc. Modified polynucleotides encoding granulysin
US9301993B2 (en) 2012-04-02 2016-04-05 Moderna Therapeutics, Inc. Modified polynucleotides encoding apoptosis inducing factor 1
US9221891B2 (en) 2012-04-02 2015-12-29 Moderna Therapeutics, Inc. In vivo production of proteins
US9283287B2 (en) 2012-04-02 2016-03-15 Moderna Therapeutics, Inc. Modified polynucleotides for the production of nuclear proteins
US9255129B2 (en) 2012-04-02 2016-02-09 Moderna Therapeutics, Inc. Modified polynucleotides encoding SIAH E3 ubiquitin protein ligase 1
US9220792B2 (en) 2012-04-02 2015-12-29 Moderna Therapeutics, Inc. Modified polynucleotides encoding aquaporin-5
US9254311B2 (en) 2012-04-02 2016-02-09 Moderna Therapeutics, Inc. Modified polynucleotides for the production of proteins
US9233141B2 (en) 2012-04-02 2016-01-12 Moderna Therapeutics, Inc. Modified polynucleotides for the production of proteins associated with blood and lymphatic disorders
US9220755B2 (en) 2012-04-02 2015-12-29 Moderna Therapeutics, Inc. Modified polynucleotides for the production of proteins associated with blood and lymphatic disorders
EP3699284A1 (en) 2012-07-05 2020-08-26 The Trustees of the University of Pennsylvania U1 snrnp regulates gene expression and modulates oncogenicity
EP4148135A1 (en) 2012-07-05 2023-03-15 The Trustees of the University of Pennsylvania U1 snrnp regulates gene expression and modulates oncogenicity
US9937205B2 (en) 2012-09-04 2018-04-10 The Trustees Of The University Of Pennsylvania Inhibition of diacylglycerol kinase to augment adoptive T cell transfer
US11931380B2 (en) 2012-09-04 2024-03-19 The Trustees Of The University Of Pennsylvania Inhibition of diacylglycerol kinase to augment adoptive T cell transfer
EP2711016A1 (en) 2012-09-21 2014-03-26 Covagen AG Novel IL-17A binding molecules and medical uses thereof
DE102012020496A1 (en) 2012-10-18 2014-04-24 Charité - Universitätsmedizin Berlin Invitro diagnosis and/or early detection of neurofibromatosis type 1 and/or neurofibromatosis type 1-associated tumor from sample of a patient for treating the patient with the tumor, comprises detecting protein or its fragment in sample
US9597380B2 (en) 2012-11-26 2017-03-21 Modernatx, Inc. Terminally modified RNA
EP3792278A2 (en) 2012-12-21 2021-03-17 Biogen MA Inc. Human anti-tau antibodies
WO2014102399A1 (en) 2012-12-31 2014-07-03 Neurimmune Holding Ag Recombinant human antibodies for therapy and prevention of polyomavirus-related diseases
EP3517545A1 (en) 2012-12-31 2019-07-31 Neurimmune Holding AG Recombinant human antibodies for therapy and prevention of polyomavirus-related diseases
US8980864B2 (en) 2013-03-15 2015-03-17 Moderna Therapeutics, Inc. Compositions and methods of altering cholesterol levels
US10059750B2 (en) 2013-03-15 2018-08-28 Angelica Therapeutics, Inc. Modified toxins
WO2014180306A1 (en) 2013-05-08 2014-11-13 上海益杰生物技术有限公司 Nucleic acid of coded gpc3 chimeric antigen receptor protein and t lymphocyte expressing gpc3 chimeric antigen receptor protein
EP4056591A1 (en) 2013-07-03 2022-09-14 ImmunoQure AG Human anti-ifn-alpha antibodies
US10815291B2 (en) 2013-09-30 2020-10-27 Modernatx, Inc. Polynucleotides encoding immune modulating polypeptides
US10323076B2 (en) 2013-10-03 2019-06-18 Modernatx, Inc. Polynucleotides encoding low density lipoprotein receptor
WO2015106641A1 (en) 2014-01-15 2015-07-23 上海易乐生物技术有限公司 Anti-human probdnf monoclonal antibody, and uses thereof in pains
WO2015153144A1 (en) 2014-03-31 2015-10-08 Kirin-Amgen, Inc. Methods of treating nail and scalp psoriasis
US10345298B2 (en) 2014-04-30 2019-07-09 Klinikum Rechts Der Isar Der Technischen Universität München Diagnosis of multiple sclerosis
US11136388B2 (en) 2014-06-27 2021-10-05 Sanofi Biomarkers for anti-IL4-IL13 bispecific antibodies
WO2015198146A2 (en) 2014-06-27 2015-12-30 Sanofi Anti-il4-il 13 bispecific antibodies
US11198729B2 (en) 2014-07-17 2021-12-14 Cafa Therapeutics Limited Immunologic effector cell of targeted CLD18A2, and preparation method and use thereof
US10377822B2 (en) 2014-07-17 2019-08-13 Carsgen Therapeutics Ltd. Immunologic effector cell of targeted CLD18A2, and preparation method and use thereof
US10183965B2 (en) 2014-08-29 2019-01-22 Katholieke Universiteit Leuven Cofactor analogues for methyltransferases
WO2016050822A2 (en) 2014-09-30 2016-04-07 Neurimmune Holding Ag Human-derived anti-dipeptide repeats (dprs) antibody
WO2016081796A1 (en) 2014-11-21 2016-05-26 Yale University Compositions and methods for modulating salm5 and hvem
WO2016140910A2 (en) 2015-03-04 2016-09-09 University Of Rochester Compositions and methods of using anti-mullerian hormone for treatment of infertility
WO2016146702A1 (en) 2015-03-16 2016-09-22 Helmholtz Zentrum München - Deutsches Forschungszentrum für Gesundheit und Umwelt (GmbH) Trispecific binding molecules for treating hbv infection and associated conditions
WO2016149621A1 (en) 2015-03-18 2016-09-22 The Johns Hopkins University Novel monoclonal antibody inhibitors targeting potassium channel kcnk9
US10889644B2 (en) 2015-11-05 2021-01-12 Biouniversa S.R.L. Humanized anti-BAG3 antibodies
EP3693387A1 (en) 2015-11-05 2020-08-12 Intrepida Bio, Inc. Humanized anti-bag3 antibodies
EP3202788A1 (en) 2016-02-05 2017-08-09 MediaPharma S.r.l. Endosialin-binding antibody
US11318210B2 (en) 2016-02-05 2022-05-03 Fusion Pharmaceuticals Inc. Endosialin-binding antibody
WO2017167967A1 (en) 2016-04-01 2017-10-05 Istituto Biochimico Italiano Giovanni Lorenzini S.P.A. Erbb2-targeting antibody
EP3269740A1 (en) 2016-07-13 2018-01-17 Mabimmune Diagnostics AG Novel anti-fibroblast activation protein (fap) binding agents and uses thereof
US10316037B1 (en) 2016-11-04 2019-06-11 Yale University Compounds and methods for treating cancer
WO2018083282A1 (en) 2016-11-07 2018-05-11 Biouniversa S.R.L. Anti-bag3 antibodies in combination with inhibitors of immune check-point for therapeutic use
WO2018129029A1 (en) 2017-01-04 2018-07-12 Immunogen, Inc. Met antibodies and immunoconjugates and uses thereof
US11773182B2 (en) 2017-01-05 2023-10-03 The Johns Hopkins University Development of new monoclonal antibodies recognizing human prostate-specific membrane antigen (PSMA)
WO2018172465A1 (en) 2017-03-22 2018-09-27 Sanofi Treatment of lupus using humanized anti-cxcr5 antibodies
US10842869B2 (en) 2017-03-22 2020-11-24 Sanofi Method of treating lupus by administering humanized anti-CXCR5 (C-X-C motif chemokine receptor 5) antibodies
US10722589B2 (en) 2017-04-03 2020-07-28 Covagen Ag FGFR3 binding molecules
WO2018185050A1 (en) 2017-04-03 2018-10-11 Covagen Ag Fgfr3 binding molecules
US11351267B2 (en) 2017-04-03 2022-06-07 Cilag Gmbh International FGFR3 binding molecules
WO2019207159A1 (en) 2018-04-27 2019-10-31 Fondazione Ebri Rita Levi-Montalcini Antibody directed against a tau-derived neurotoxic peptide and uses thereof
US11155638B2 (en) 2018-05-08 2021-10-26 Rhode Island Hospital Anti-CHI3L1 antibodies for the detection and/or treatment of nonalcoholic fattly liver disease/nonalcoholic steatonhepatitis and subsequent complications
WO2020014306A1 (en) 2018-07-10 2020-01-16 Immunogen, Inc. Met antibodies and immunoconjugates and uses thereof
WO2020053808A1 (en) 2018-09-12 2020-03-19 Georg Dewald Method of diagnosing vasoregulatory disorders
WO2020099235A1 (en) 2018-11-12 2020-05-22 Mediapharma S.R.L. Bispecific antibodies directed against human 90k and either endosialin or her3
WO2020157122A1 (en) 2019-01-29 2020-08-06 Helmholtz Zentrum München - Deutsches Forschungszentrum für Gesundheit und Umwelt (GmbH) Treating the causative agent in adhesiogenesis
WO2020167912A1 (en) 2019-02-13 2020-08-20 The Brigham And Women's Hospital, Inc. Anti-peripheral lymph node addressin antibodies and uses thereof
WO2020242989A1 (en) 2019-05-24 2020-12-03 Sanofi Methods for treating systemic sclerosis
US11827671B2 (en) 2019-05-24 2023-11-28 Sanofi Methods for treating systemic sclerosis
WO2021140173A1 (en) 2020-01-10 2021-07-15 Biouniversa S.R.L. Methods and uses for treating fibrotic solid tumors with bags inhibitors
WO2021183207A1 (en) 2020-03-10 2021-09-16 Massachusetts Institute Of Technology COMPOSITIONS AND METHODS FOR IMMUNOTHERAPY OF NPM1c-POSITIVE CANCER
WO2022189632A1 (en) 2021-03-12 2022-09-15 Fibrosys S.R.L. Monoclonal antibodies for the treatment of viral infections

Also Published As

Publication number Publication date
AU3544589A (en) 1989-11-03
EP0362371A4 (en) 1990-10-24
KR900700134A (en) 1990-08-11
AU631545B2 (en) 1992-12-03
JPH02503867A (en) 1990-11-15
FI895955A0 (en) 1989-12-13
EP0362371A1 (en) 1990-04-11

Similar Documents

Publication Publication Date Title
AU631545B2 (en) Il-2 receptor-specific chimeric antibodies
CA2006865C (en) Humanized immunoglobulins and their production and use
EP0566647B1 (en) Improved humanized immunoglobulins
US5714350A (en) Increasing antibody affinity by altering glycosylation in the immunoglobulin variable region
US5882644A (en) Monoclonal antibodies specific for the platelet derived growth factor β receptor and methods of use thereof
NZ227968A (en) Hybrid antibody and compositions
IL141158A (en) Humanized immunoglobulin which binds to human von willebrand factor

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AU DK FI JP KR NO

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): AT BE CH DE FR GB IT LU NL SE

WWE Wipo information: entry into national phase

Ref document number: 895955

Country of ref document: FI

WWE Wipo information: entry into national phase

Ref document number: 1989905832

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: 1989905832

Country of ref document: EP

WWW Wipo information: withdrawn in national office

Ref document number: 1989905832

Country of ref document: EP