WO1992016562A1 - Humanised antibodies having modified allotypic determinants - Google Patents

Humanised antibodies having modified allotypic determinants Download PDF

Info

Publication number
WO1992016562A1
WO1992016562A1 PCT/GB1992/000445 GB9200445W WO9216562A1 WO 1992016562 A1 WO1992016562 A1 WO 1992016562A1 GB 9200445 W GB9200445 W GB 9200445W WO 9216562 A1 WO9216562 A1 WO 9216562A1
Authority
WO
WIPO (PCT)
Prior art keywords
constant region
amino acid
molecule
allotypic
immunoglobulin
Prior art date
Application number
PCT/GB1992/000445
Other languages
French (fr)
Inventor
Michael Ronald Clark
Original Assignee
Lynxvale Limited
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 Lynxvale Limited filed Critical Lynxvale Limited
Priority to JP4505594A priority Critical patent/JPH06510659A/en
Publication of WO1992016562A1 publication Critical patent/WO1992016562A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies

Definitions

  • the present invention relates to binding molecules .
  • it relates to recombinantly produced antibodies .
  • Monoclonal antibodies are made by establishing an immortal cell line which is derived from a single immunoglobulin producing cell secreting one form of a biologically functional antibody molecule with a particular specificity.
  • the simplest antibody comprises four polypeptide chains, two heavy (H) chains and two light (L) chains inter-connected by disulphide bonds (see figure 1).
  • the light chains are of two types, either kappa or lambda.
  • Each of the H and L chains has a region of low sequence variability, the constant region (C) giving rise to allotypes and a region of high sequence variability, the variable region (V) giving rise to idiotypes.
  • the antibody has a tail region (the Fc region) which comprises the C regions of the two H chains.
  • the antibody also has two arms (the Fab region) each of which has a V L and a V H region associated with each other. It is this pair of V regions (V L and V H ) that differ from one antibody to another, and which together are responsible for recognising the antigen.
  • each V region is made up from three complementarity determining regions (CDR) separated by four framework regions (FR).
  • CDRs are the most variable part of the variable regions, and they perform the critical antigen binding function.
  • the CDR regions are derived from many potential germ line sequences via a complex process involving recombination, mutation and selection. It has been shown that the function of binding antigens can be performed by fragments of a whole antibody. Binding fragments are the Fv fragment which comprises the V L and V H of a single heavy chain variable domain (V H ).
  • the Fc tail of a non-human antibody is exchanged for that of a human antibody.
  • the FRs of the non-human antibody are exchanged for human FRs. This process is carried out at the DNA level using recombinant techniques.
  • these humanised immunoglobulins do not solve all the problems, because an immune response may still be mounted against the treatment antibody even when a patient is treated with a human antibody, as it may show certain sequence differences in the V (ie idiotypic differences) and C (ie allotypic differences) regions when compared with the patients own equivalent antibodies.
  • IgG immunoglobulin G
  • IgM immunoglobulin A
  • IgD immunoglobulin D
  • IgE immunoglobulin E
  • the allotypes represent alternative amino acid substitutions found at discrete sites in the protein sequence. These different allotypic determinants are found in different combinations within given allelic forms of genes, but not all possible combinations which theoretically might exist are in practice observed.
  • the four different allotypes of IgG1 can be seen (ie distinguished) by the immune system. These are G1m 1, 2, 3 and 17. Alternatively, combinations thereof, such as G1m (1, 17 ) , can also be distinguished. The four different single allotypes are depicted in figure 2.
  • Antisera can be raised in other non-human species which can see the alternative isoallotypes provided that the antibody is purified away from the other human isotypes.
  • Such isoallotypes for which such an antisera exists have been called non-allotypes and given the designation for example, nG1m(1) which is the isoallotype of G1m(1).
  • nG1m(1) which is the isoallotype of G1m(1).
  • the sequence of the human allotype of IgG1 G1m (1, 2, 17) is shown aligned with sequences for the other human IgG, isotype sub-classes in figure 4 (a, b, c and d). It can be seen that the four isotypes are extremely homologous for the domains CH1, CH2 and CH3, and that the major isotypic differences are in the hinge region which varies in both, length and sequence between isotypes.
  • the allotypic residues of IgG1 G1m (1, 2, 17) have been marked in figure 4. However, for the purposes of clarity the sequences around the allotypic sites G1m (1) (2) and (17) are shown below for each isotype.
  • IgG1 may exist as several allotypes depending on whether aspartic acid or glutamic acid at position 356, or leucine or methionine at position 358 are present.
  • IgG1 may exist as either of two allotypes depending on whether glycine or alanine is present at position 431.
  • IgG1 may exist as either of two allotypes depending on whether lysine or arginine is present.
  • the allotypes (17) and (3) cannot co-exist as they represent alternative substitutions at the same position.
  • the alternative alleles of G1m (1) and (2) do not provoke a human allotype response because of the homology of these alleles with the other IgG sub-classes in this region. These alleles are therefore called isoallotypes because they are only recognisable by xenoantisera (antisera from a different species) and only when the isotype is purified away from the other sub-classes.
  • the present applicants propose the creation of a new IgG1 allele by site-directed mutagenesis of the gene, for example, an existing CAMPATH-1H monoclonal antibody gene described below, so that the new allele consists entirely of isoallotypic determinants.
  • the preparation of IgG1 mutants according to the teaching provided by the present applicants is shown schematically in figure 3.
  • IgG1 is specifically directed to IgG1 and in particular, the CAMPATH-1H monoclonal antibody
  • the same approach can be used to create isoallotypes of the other human isotypes such as IgG2, IgG3 and kappa.
  • the present invention provides a first binding molecule derivable from a second binding molecule; which second binding molecule is an immunoglobulin, or a derivative, structural or functional analogue thereof, a member of a family of homologous molecules, and has one or more sites which are structurally distinctive from equivalent sites in the other family members;
  • said first binding molecule is more closely homologous to the other family members than to said second binding molecule, at at least one of said one or more sites.
  • the first binding molecule may also be an immunoglobulin or a derivative, structural or functional analogue thereof.
  • the one or more sites which are structurally distinctive from the equivalent sites in the other family members may be in the constant region giving rise to an allotypic difference.
  • the first binding molecule may comprise entirely isoallotypic determinants.
  • the second binding molecule may be selected from the group consisting of IgG1, IgG2, IgG3, lgA2, IgE, kappa light chains or derivatives, structural or functional analogues thereof.
  • the allotypic differences may be present at one or more of sites (1) (2) (3) or (17) as described herein.
  • the second binding molecule is IgG2
  • the allotypic difference may be present at site (23).
  • the second binding molecule is IgG3
  • the allotypic differences may be present at one or more of the sites (11) (5) (13) (14)
  • the allotypic differences may be present at one or more of the sites (1) and (2).
  • the second binding molecule is kappa light chain, the allotypic differences may be present at one or more of the sites (1) (2) or (3).
  • the present invention also provides pharmaceutical preparations comprising a first binding molecule as defined above or described herein together with one or more excipients.
  • the pharmaceutical preparation may comprise a cocktail of said first binding molecules.
  • Also provided by the present invention are methods for making a first binding molecule as defined above or described herein.
  • the first binding molecule may be made by providing a gene sequence encoding the second binding molecule and altering those parts of the gene sequence encoding said one or more sites.
  • the gene sequence may be altered by site directed mutagenesis using oligonucleotide primers.
  • the altered gene sequence may be incorporated into a cloning vector or expression vector.
  • the expression vector may be used to transform a cell. The cell may be induced to express the altered gene sequence.
  • the present invention therefore provides cloning vectors and expression vectors incorporating the altered gene sequence. Also provided are cells transformed by expression vectors defined above. Also provided are cell cultures and products of cell cultures containing the first binding molecules. Also provided are recombinantly produced said first binding molecules.
  • the present invention provides a molecule which comprises an amino acid sequence derivable from part or all of the constant region of an immunoglobulin heavy chain which constant regions are of a particular isotype and have one or more allotypic determinants
  • amino acid sequence is substantially homologous to the amino acid sequence of said constant region, but has been altered so that it is without at least one of said allotypic determinants, by making the amino acid residues at the site of an allotypic determinant identical to the amino acid residues from the corresponding position in another equivalent immunoglobulin constant region of a different isotype.
  • the molecule may comprise an amino acid sequence derivable from part or all of a human immunoglobulin constant region.
  • the molecule may also comprise one or more polypeptides together with said amino acid sequence.
  • the polypeptide may comprise a functional biological domain.
  • the domain may be such that it mediates any biological function.
  • the functional biological domain may comprise a binding domain.
  • the binding domain will have an ability to interact with another polypeptide. The interaction may be non-specific or specific.
  • the polypeptide, biological domain, binding domain and immunoglobulin-like binding domain may derive from the same source or a different source to the constant region.
  • the constant region may be from an immunoglobulin of the isotype IgG.
  • the isotype subclass may be IgG1 and the molecule may no longer have one or more of the allotypic determinants 1,2,3 and 17.
  • the isotype subclass may be IgG2 and the molecule may no longer have the allotypic determinant 23.
  • the isotype subclass may be IgG3 and the molecule may no longer have one or more of the allotypic determinants 11,5,13,14,10,6,24,21,15, 16, 26 and 27.
  • the constant region may be from an immunoglobulin of the isotype IgA2 and the molecule may no longer have either or both of the allotypic determinants 1 and 2.
  • the present invention also provides a pharmaceutical preparation which comprises a molecule as defined.
  • the present invention also provides a reagent which comprises a molecule as defined.
  • the present invention also provides a nucleotide sequence encoding a molecule as defined.
  • the present invention also provides cloning and expression vectors comprising a nucleotide sequence as delivered above.
  • the present invention also provides host cells comprising a cloning or expression vector as defined above.
  • the present invention also provides a method of preparing a molecule as defined above which comprises the steps of:
  • the present invention also provides a method of treating a patient which comprises administering a pharmaceutical preparation as defined above.
  • Genes encoding therapeutically useful antibodies are generally available in one of several different forms. They may be available as a cloned variable region DNA sequence with restriction sites at each end, suitable for recloning along with a chosen cloned constant region DNA sequence into a suitable expression vector. This is the strategy described herein for the constructs TF57-19, MTF121 and MTF123. Alternatively, they may be available as complete immunoglobulin DNA sequences including the V and C regions together, e.g. a cDNA clone of a complete humanised or human antibody.
  • the next step is to predict the amino acid sequence of the constant region from the DNA sequence.
  • the DNA sequence can be obtained using a variety of strategies familiar to molecular biologists.
  • the predicted amino acid sequence would then be checked for the amino acids known to vary as allotypes. Any isoallotypes present within the sequence can be left unaltered. Any allotypes present can be mutated.
  • the next step is to decide what amino acid sequence to mutate the allotype to, in order to imitate an isoallotype. This is done by lining up the sequence with the corresponding region of the other immunoglobulin isotypes. For all known allotypes, it has been found that one or more of the other isotypes have invariant sequences for the homologous region. One of these sequences can then be chosen to form the basis for the changes to be made in the allotype in question. Having predicted the new amino acid sequence for the constant region, it is necessary to alter the existing DNA clone or to create a new DNA clone which will encode this sequence. Again there are several strategies available to molecular biologists in order to achieve this.
  • the gamma-1 constant region was cloned in an M13TG131 single stranded phage vector. Mutagenic oligonucleotides were synthesised which were largely homologous to the single strand, but which contained base changes necessary to alter the codons for the critical amino acids. The mutagenesis was carried out using a commercial kit from Amersham International, High Wycombe, Bucks. Alternatively it would be possible to synthesise a complete artificial gene which encodes the predicted sequence.
  • the gene is ready for expression.
  • expression vectors There are many different expression vectors available. Some of these are more suitable for expression in restricted cell types. Again it is within the standard technical expertise of one skilled in this field to choose and adapt expression vectors for this purpose.
  • modifications of the pSVgpt and pSVneo vectors have been used. These vectors have convenient cloning sites for the immunoglobulin variable and constant region, encoding DNA fragments adjacent to suitable promoter and enhancer sequences to allow expression in lymphoid cells.
  • the vector allows the easy independent replacement of variable or constant region encoding DNA fragments.
  • any suitable variable region can be subcloned into the vector, to give rise to a new specificity, or the variable region can be kept and the constant region changed to give rise to a new isotype or allotype.
  • Alternative vector systems are readily available.
  • the most common kappa light chain allotype is Km(3) in the general population. Therefore it may be sufficient to utilise this common kappa light chain allotype, as relatively few members of the population would see it as foreign.
  • the allotype Km(1,2) could first be mutated to the allotype Km(1).
  • the light chain allotype Km(1) is often only weakly recognized in combination with certain heavy chain classes and subclasses, and so may not cause a major problem in therapeutic use.
  • figure 1 illustrates the structure of an IgG antibody
  • FIG 2 shows the allotypes for the IgG1 antibody CAMPATH-1H
  • FIG. 3 shows schematically the preparation of IgG1 mutants
  • figure 4 shows the IgG1 G1m (1,2,17) allotype sequence aligned to single allotypic examples of IgG2, 3 and 4 (none of these other subclasses have allotypic residues which cover the same residues as for the IgG1 allotypes);
  • figure 5 shows the M13TG131 cloning vector containing the human gamma-1 constant region, showing cloning sites and modified polylinker;
  • figure 6 shows the original Hu4vH HuG1 pSVgpt expression vector and its modified version
  • figure 7 shows the result of an ELISA assay testing different dilutions of the antibodies of mutants 1, 2 and wild type CAMPATH-1H for IgG1 specificity
  • figure 8 shows the result of an autologous complement mediated lysis test using human peripheral blood lymphocytes
  • FIG 9 illustrates an antibody-dependent cell- mediated cytotoxicity assay (ADCC) using CD3 activated interleukin-2 expanded human blastocytes cell effectors (E) and targets (T).
  • ADCC antibody-dependent cell- mediated cytotoxicity assay
  • the starting antibody used for site-directed mutagenesis was CAMPATH-1H, a monoclonal antibody with a kappa light chain containing the human constant region sequence for IgG1 which carries the G1m (1, 17) allelic determinants.
  • the whole IgG1 encoding region exists as approximately 2.3 kb HindIII-SphI restriction fragment cloned in an M13 vector.
  • the M13TG131 cloning vector containing the human gamma-1 constant region showing cloning sites and modified polylinker is shown in figure 5.
  • the IgG1 encoding region is entered in the EMBL Sequence Database under the code number HS1GCC4.
  • the accession number is AC J00228 (the printout from the database is provided herein as Appendix 1).
  • This sequence is for the G1m (1, 17) allotype. It covers 2009 bases from the 5' Hindlll site (A)AGCTT including all of the coding region. It does not however, include some of the 3' non-coding region up to the SphI site.
  • the sequence provided by the EMBL Database is that of the upper strand of DNA.
  • the CH1 domain starts at nucleotide 210 and ends at nucleotide 503.
  • the mutagenic oligonucleotides MO1 and MO4 hybridise to nucleotides 486 to 510.
  • the hinge region starts at nucleotide 892 and ends at nucleotide 936.
  • the CH2 domain starts at nucleotide 1481 and ends at nucleotide 1803.
  • the mutagenic oligonucleotide MO2 hybridises to nucleotides 1515 to 1543.
  • the essential signal for the poly A tail is provided by nucleotides 1902 to 1908.
  • the IgG1 coding region exists as a 2260 nucleotide fragment, of which the final 251 nucleotides are non-coding and therefore, inessential. Therefore, an embodiment of the invention could be carried out using the sequence information provided by the EMBL Sequence Database. It should be noted however, that the Sph1 restriction site referred to above, is present in the 3' end inessential non-coding region. Therefore, if the sequence data as provided by the EMBL database were being used, alternative restriction sites would have to be utilised.
  • mutagenic oligonucleotides were:
  • oligonucleotides were synthesised and then purified using an automated synthesizer and oligo purification columns supplied by Applied Biosystems (Applied Biosystems, 850 Lincoln Drive, Foster City, California, 94404 USA) following the manufacturers recommended protocols. Mutations were checked by Sanger Dideoxy sequencing (Sanger, F.S., Nicklen, S., and Coulson, A.R., (1977) Proc. Natl. Acad. Sci., USA, 74, 5463) using standard protocols and kits. As this newly formed allotype sequence is found in all humans, there should be no immunological response to this alternative form of G1m (1). Additionally and similarly, the lysine residue responsible for the G1m (17) allotypic determinant was converted to an arginine residue corresponding to the G1m allele (Lys 214-Arg; mutant 1).
  • mutant 1 The gene for this new constant region of mutant 1 carrying these three changes has been sequenced, incorporated into an expression vector containing the CAMPATH-1H V-region and expressed together with the CAMPATH-1H light chain which had been introduced by co-transfection.
  • a further mutant has been made by replacing the critical arginine residue associated with G1m (3) of mutant 1 with a threonine residue, to produce a heavy chain which is the equivalent of IgG2 and which should fail to react with both anti-G1m (17) and anti-G1m (3) antisera (mutant 2).
  • Mutant 2 has also been sequenced, re-cloned in an expression vector containing the CAMPATH-1H light chain.
  • the supernatants of growing cultures containing either of the two mutants were subsequently assayed for the expression of a human IgG1 kappa product.
  • the mutations were introduced with the oligonucleotides listed above.
  • the modified Hu4vHG1pSVgpt vector shown in figure 6 was used to simplify the subcloning of all the new mutants into the expression vector, owing to the possibility of use of two different "sticky ends" Bam HI and NotI.
  • the expression vectors and V H region sequences and expression, along with the light chains, in YO rat plasmacytoma cells are all as described in Riechmann L., Clark, M.R. Waldman H., Winter G. (1988) Nature 332, 323-327.
  • Example 1 From the positive cultures, the producers of the largest amount of the IgG1 product were selected to obtain purified antibody for biological assays to determine their allotypes and biological effector functions.
  • Example 1 From the positive cultures, the producers of the largest amount of the IgG1 product were selected to obtain purified antibody for biological assays to determine their allotypes and biological effector functions.
  • Human peripheral blood mononuclear cells from a healthy donor were isolated from 60 ml defibrinated blood on a Lymphoprep gradient (Nyeggard & Co., AS, Oslo, Norway). The cell pellet was washed in IMDM (Iscove's Modification of Dulbecco's Medium, Flow Laboratories, Scotland), and the cells were labelled with 51 Cr.
  • the starting dilution of antibodies used in the test was 50 ⁇ g/ml in PBS, 10 ⁇ g/ml BSA (dilution 1). Dilution 1 was further diluted 8 times two-fold to a final dilution of 1/128. Wild type antibody diluted in the same manner was used as a control.
  • ADCC antibody-dependent cell-mediated cytotoxicity
  • Unlabelled blastocytes (2 ⁇ 10 6 ) were diluted 100:1 and 30:1 in IMDM 1% BSA medium.
  • the ratios 100:1 and 30:1 refer to the final absolute ratios of effectors to 5 1 Cr labelled targets in the assay.
  • Assays were performed in microtitre plates with a total volume of 200 ⁇ l per assay well. Thus 100 ⁇ l of targets at a concentration of 2 ⁇ 10 4 were put in each well ie 2 x 10 3 total cells.
  • E:T of 100:1 100 ⁇ l of effectors at 2 x 10 4 were plated per well ie 2 ⁇ 10 5 .
  • For E:T of 30:1 100 ⁇ l of effectors at 6 ⁇ 10 5 were put into each well ie 6 x 10 4 total cells.
  • cpm radioactive counts per minute as measured on a counter.
  • FIG. 9 The result is shown in figure 9. The figure shows that all of the antibodies tested released chromium. Wild type TF 57-19 and mutant 2 (MTF 123) released at about equal levels, whereas mutant 1 (MTF 121) shows a slightly higher release.
  • the antibodies were tested in an assay specific for their G1m (3) allotypes reactivity using a monoclonal reagent from Oxoid (WHO/IVISS recognised agent, Study
  • Microtiter plates were coated with the anti-CAMPATH idiotype YID 13.9.4 antibody captive, and divided into three arrays of 4 ⁇ 4 wells. Into each of the three arrays, 4 ⁇ 5 fold dilutions of the antibody TF 57-19, MTF 121 and MTF 123 (50 ⁇ g/ml) in PBS 1% BSA and a control solution of PBS/BSA each were added.
  • the microtiter plate was developed with streptavidin horseradish peroxidase.
  • the result is illustrated in Table 1.
  • the numbers in the results represent the optical density (O.D) as measured in an ELISA plate reader multiplied by 100 ie 12 represents an O.D of 0.12 and 70 an O.D of 0.70.
  • the antibodies were tested in a conventional allotyping experiment using inhibition of red cell agglutination.
  • the experiment was carried out using reagents supplied by the Central Laboratory of the Netherlands Red Cross, Blood Transfusion Service (PO Box 9190, 1006 AD Amsterdam, Netherlands).
  • coated red blood cells were washed four times with PBS and stored at 4°C in preservation fluid (although coated red blood cells can be stored at 4°C in PBS for one week). These coated red blood cells were then agglutinated with four antisera to the IgG1 allotypes as follows using the recommended dilution for each antiserum.
  • anti-G1m(z) anti-G1m(17) 1 in 30 dilution
  • anti-G1m(a) anti-G1m(l) 1 in 30 dilution
  • anti-G1m(f) anti-G1m(3) 1 in 30 dilution
  • CAMPATH-1H constructs (MTF 121, MTF 123) with the altered gamma-1 constant regions were then tested for their abilities to inhibit the agglutination of the red cells by the above antisera.
  • the inhibiting antibodies were tried at concentrations of 0.5mg/ml, 0.25mg/ml and 0.125mg/ml in phosphate buffered saline containing 5% foetal bovine serum.
  • Control sera containing IgG1 of allotype G1m(zax) or G1m(f) [G1m(1,2,17) or G1m(3)] were also included in the experiment and were used at dilutions of 1 in 10,20 and 40.
  • Allotype CAMPATH-1H constructs Control sera TF57-19 MTF121 MTF123 G1m(1,2,17) G1m(3) G1m(1) + - - + - G1m(2) - - - +
  • binding molecules hereby provided to make pharmaceuticals according to standard techniques.
  • pharmaceuticals can be used in accordance with standard practices.

Abstract

The invention relates to molecules which have an amino acid sequence derivable from part or all of the constant region of an immunoglobulin heavy chain. The constant regions are of a particular isotype and have one or more allotypic determinants. The amino acid sequence is substantially homologous to the amino acid sequence of the constant region. However, it has been altered so that it is without at least one of said allotypic determinants by making its sequence the site for an allotypic determinant identical to the amino acid sequence from the corresponding position in another equivalent immunoglobulin constant region of a different isotype. The invention provides synthetic immunoglobulins with reduced allotypic differences as compared to a given wild-type immunoglobulin.

Description

Humani sed antibodies havi ng modified a llotypic determinants
The present invention relates to binding molecules . In particular, it relates to recombinantly produced antibodies .
Owing to their high specificity for a given antigen, antibodies and particularly monoclonal antibodies ( Kohler , G . and Milstein C. , 1975 Nature 256 : 495 ) represented a significant technical break-through with important consequences scientifically, commercially and therapeutically.
Monoclonal antibodies are made by establishing an immortal cell line which is derived from a single immunoglobulin producing cell secreting one form of a biologically functional antibody molecule with a particular specificity.
Owing to their specificity , the therapeutic applications of monoclonal antibodies hold great promise for the treatment of a wide range of diseases ( Clinical Applications of Monoclonal Antibodies, edited by E. S . Lennox . British . Medical Bulletin 1984 , publishers Churchill Livingstone ) . Antibodies are generally raised in animals , particularly rodents , and therefore the immunoglobulins produced bear characteristic features specific to that species . The repeated administration of these foreign rodent proteins for therapeutic purposes to human patients can lead to harmful hypersensitivity reactions. In the main therefore, these rodent-derived monoclonal antibodies have limited therapeutic use. A further problem with these rodent derived antibodies, is that they are relatively ineffective at the depletion of cells in vivo, although the rat IgG2b antibody CAMPATH-1G is an exception to this rule.
Thus, there is a need for therapeutic antibodies which have characteristic features specific to human proteins. Unfortunately, immortal human antibody- producing cell lines are very difficult to establish and they give low yields of antibody (approximately 1 μg/ml). In contrast, equivalent rodent cell lines yield high amounts of antibody (approximately 100 μg/ml). Furthermore, where one wants to produce a human antibody with a particular specificity it is not practically or ethically feasible to immunise humans with an immunogen bearing the epitope of interest.
In part, this problem has been overcome in recent years by using the techniques of recombinant DNA technology to 'humanise' non-human antibodies. Structurally, the simplest antibody (IgG) comprises four polypeptide chains, two heavy (H) chains and two light (L) chains inter-connected by disulphide bonds (see figure 1). The light chains are of two types, either kappa or lambda. Each of the H and L chains has a region of low sequence variability, the constant region (C) giving rise to allotypes and a region of high sequence variability, the variable region (V) giving rise to idiotypes. The antibody has a tail region (the Fc region) which comprises the C regions of the two H chains. The antibody also has two arms (the Fab region) each of which has a VL and a VH region associated with each other. It is this pair of V regions (VL and VH) that differ from one antibody to another, and which together are responsible for recognising the antigen. In even more detail, each V region is made up from three complementarity determining regions (CDR) separated by four framework regions (FR). The CDRs are the most variable part of the variable regions, and they perform the critical antigen binding function. The CDR regions are derived from many potential germ line sequences via a complex process involving recombination, mutation and selection. It has been shown that the function of binding antigens can be performed by fragments of a whole antibody. Binding fragments are the Fv fragment which comprises the VL and VH of a single heavy chain variable domain (VH).
In creating "humanised" immunoglobulins, the Fc tail of a non-human antibody is exchanged for that of a human antibody. For a more complete humanisation, the FRs of the non-human antibody are exchanged for human FRs. This process is carried out at the DNA level using recombinant techniques. However, these humanised immunoglobulins do not solve all the problems, because an immune response may still be mounted against the treatment antibody even when a patient is treated with a human antibody, as it may show certain sequence differences in the V (ie idiotypic differences) and C (ie allotypic differences) regions when compared with the patients own equivalent antibodies. This is a particular problem where the patient's immune system has already seen, and therefore been primed against, antibodies having these sequence differences (eg a patient may have received a prior blood transfusion which contained allotypically different immunoglobulins). A model system of injecting "mouseised human antibodies" into mice indicated that the allotype matching could critically affect the anti-idiotype response (Bruggemann M., Winter G., Waldmann H., Neuberger M.S., (1989) J. Exp. Med. 170, 2153-2157).
The present applicants have realised that one way around this problem is to eliminate the allotypic variation from the constant region.
There are a range of different immunoglobulins IgG, IgM, IgA, IgD, IgE, known as isotypes, of which IgG is most commonly used therapeutically. It exists as isotypic sub-classes IgG1, IgG2, IgG3 and IgG4.
There are 24 recognised allotypes of human immunoglobulin distributed between the different isotypes as follows:
IgG1 × 4
IgG2 × 1
IgG3 × 13
IgA2 × 2
IgE × 1
Kappa × 3
The allotypes represent alternative amino acid substitutions found at discrete sites in the protein sequence. These different allotypic determinants are found in different combinations within given allelic forms of genes, but not all possible combinations which theoretically might exist are in practice observed.
For example, the four different allotypes of IgG1 can be seen (ie distinguished) by the immune system. These are G1m 1, 2, 3 and 17. Alternatively, combinations thereof, such as G1m (1, 17 ) , can also be distinguished. The four different single allotypes are depicted in figure 2.
Antisera can be raised in other non-human species which can see the alternative isoallotypes provided that the antibody is purified away from the other human isotypes. Such isoallotypes for which such an antisera exists have been called non-allotypes and given the designation for example, nG1m(1) which is the isoallotype of G1m(1). Thus, although a human isoallotype should not be immunogenic in humans, it can still potentially be recognized in a different species.
Of the above mentioned different allotypes of IgG1, three common allelic forms of human IgG1 occur with different frequencies within different racial groups, namely G1m (3), G1m (1, 17), and G1m (1, 2, 17) based upon their reactivities with human antisera directed against the determinants G1m 1, 2, 3 and 17. At some point in the future, it is likely that a patient with an existing anti-allotype response to one or more of these determinants will need treatment with a humanised antibody. The obvious solution and one which has been proposed in a letter to the Journal Nature (Mage, R.G., Nature (1988) 333, 807-808), is to make all the different allelic forms of an antibody and to allotype match each patient for therapy. The present applicants have realised that commercially this is not a good proposal because of increased production costs and the need to process several reagents in parallel through the regulatory requirements. Additionally, each patient would have to be tested for the response to different allotypes.
Thus, the present applicants propose eliminating the allotypes altogether from each therapeutic antibody. The sequence of the human allotype of IgG1 G1m (1, 2, 17) is shown aligned with sequences for the other human IgG, isotype sub-classes in figure 4 (a, b, c and d). It can be seen that the four isotypes are extremely homologous for the domains CH1, CH2 and CH3, and that the major isotypic differences are in the hinge region which varies in both, length and sequence between isotypes. The allotypic residues of IgG1 G1m (1, 2, 17) have been marked in figure 4. However, for the purposes of clarity the sequences around the allotypic sites G1m (1) (2) and (17) are shown below for each isotype.
Site (1)
355 356 357 358
Arg Asp or G1u G1u Leu or Met IgG1
Arg G1u G1u Met IgG2 Arg G1u G1u Met IgG3
Gin G1u G1u Met IgG4
Thus, at site (1), IgG1 may exist as several allotypes depending on whether aspartic acid or glutamic acid at position 356, or leucine or methionine at position 358 are present.
Site 2
430 431 432
G1u G1y or Ala Leu IgG1
G1u Ala Leu IgG2
G1u Ala Leu IgG3
G1u Ala Leu IgG4 Thus, at site (2), IgG1 may exist as either of two allotypes depending on whether glycine or alanine is present at position 431.
Site (17)/(3)
Sites (3) and (17) are alternative substitutions at the same site.
213 214 215
Lys Lys or Arg Val IgG1
Lys Thr Val IgG2
Lys Arg Val IgG3
Lys Arg Val IgG4
Thus, at site (17)/(3), IgG1 may exist as either of two allotypes depending on whether lysine or arginine is present. The allotypes (17) and (3) cannot co-exist as they represent alternative substitutions at the same position.
The alternative alleles of G1m (1) and (2) do not provoke a human allotype response because of the homology of these alleles with the other IgG sub-classes in this region. These alleles are therefore called isoallotypes because they are only recognisable by xenoantisera (antisera from a different species) and only when the isotype is purified away from the other sub-classes.
Therefore, the present applicants propose the creation of a new IgG1 allele by site-directed mutagenesis of the gene, for example, an existing CAMPATH-1H monoclonal antibody gene described below, so that the new allele consists entirely of isoallotypic determinants. The preparation of IgG1 mutants according to the teaching provided by the present applicants is shown schematically in figure 3.
For G1m (1) and G1m (2), the changes comprise simple substitution by the alternative isoallotypic residues. However, in the case of G1m (17) the conversion of lysine to arginine would in some cases merely change the allotype to an allotype that is recognised by certain individuals as a G1m (3) allotype despite the fact that this residue is homologous with IgG3 and IgG4. This apparent contradiction is thought to be because this arginine is seen in a tertiary epitope in the context of the other IgG1 specific residues in close proximity in the CH1 domain or hinge region. This indicates that in addition to changing lysine, other residues in CH1 or the hinge will need to be changed in order to create a new isoallotype.
Although the above and ensuing description is specifically directed to IgG1 and in particular, the CAMPATH-1H monoclonal antibody, the same approach can be used to create isoallotypes of the other human isotypes such as IgG2, IgG3 and kappa.
Thus, the present invention provides a first binding molecule derivable from a second binding molecule; which second binding molecule is an immunoglobulin, or a derivative, structural or functional analogue thereof, a member of a family of homologous molecules, and has one or more sites which are structurally distinctive from equivalent sites in the other family members;
wherein said first binding molecule is more closely homologous to the other family members than to said second binding molecule, at at least one of said one or more sites.
The first binding molecule may also be an immunoglobulin or a derivative, structural or functional analogue thereof. The one or more sites which are structurally distinctive from the equivalent sites in the other family members may be in the constant region giving rise to an allotypic difference. The first binding molecule may comprise entirely isoallotypic determinants.
The second binding molecule may be selected from the group consisting of IgG1, IgG2, IgG3, lgA2, IgE, kappa light chains or derivatives, structural or functional analogues thereof. Where the second binding molecule is IgG1, the allotypic differences may be present at one or more of sites (1) (2) (3) or (17) as described herein. Where the second binding molecule is IgG2, the allotypic difference may be present at site (23). Where the second binding molecule is IgG3, the allotypic differences may be present at one or more of the sites (11) (5) (13) (14)
(10) (6) (24) (21) (15) (16) (26) or (27). Where the second binding molecule is IgA2, the allotypic differences may be present at one or more of the sites (1) and (2). Where the second binding molecule is kappa light chain, the allotypic differences may be present at one or more of the sites (1) (2) or (3). The sites referred to above are well documented in the literature
(see e.g. Eur. J. Immunol. 1976.6:599-601. Review of the notation for the allotypic and related marks of human immunoglobulins).
The present invention also provides pharmaceutical preparations comprising a first binding molecule as defined above or described herein together with one or more excipients. The pharmaceutical preparation may comprise a cocktail of said first binding molecules.
Also provided by the present invention are methods for making a first binding molecule as defined above or described herein.
These methods comprise the steps of: a) identifying in said second binding molecule, one or more sites which are structurally distinctive from the equivalent sites in the other family members; b) making said first binding molecule whereby it is more closely homologous to the other family members than to said second binding molecule at at least one of said one or more sites. The first binding molecule may be made by providing a gene sequence encoding the second binding molecule and altering those parts of the gene sequence encoding said one or more sites. The gene sequence may be altered by site directed mutagenesis using oligonucleotide primers. The altered gene sequence may be incorporated into a cloning vector or expression vector. The expression vector may be used to transform a cell. The cell may be induced to express the altered gene sequence.
The present invention therefore provides cloning vectors and expression vectors incorporating the altered gene sequence. Also provided are cells transformed by expression vectors defined above. Also provided are cell cultures and products of cell cultures containing the first binding molecules. Also provided are recombinantly produced said first binding molecules.
Thus the present invention provides a molecule which comprises an amino acid sequence derivable from part or all of the constant region of an immunoglobulin heavy chain which constant regions are of a particular isotype and have one or more allotypic determinants
wherein said amino acid sequence is substantially homologous to the amino acid sequence of said constant region, but has been altered so that it is without at least one of said allotypic determinants, by making the amino acid residues at the site of an allotypic determinant identical to the amino acid residues from the corresponding position in another equivalent immunoglobulin constant region of a different isotype.
The molecule may comprise an amino acid sequence derivable from part or all of a human immunoglobulin constant region.
The molecule may also comprise one or more polypeptides together with said amino acid sequence.
The polypeptide may comprise a functional biological domain. The domain may be such that it mediates any biological function. The functional biological domain may comprise a binding domain. The binding domain will have an ability to interact with another polypeptide. The interaction may be non-specific or specific.
The polypeptide, biological domain, binding domain and immunoglobulin-like binding domain may derive from the same source or a different source to the constant region.
The constant region may be from an immunoglobulin of the isotype IgG. The isotype subclass may be IgG1 and the molecule may no longer have one or more of the allotypic determinants 1,2,3 and 17. The isotype subclass may be IgG2 and the molecule may no longer have the allotypic determinant 23. The isotype subclass may be IgG3 and the molecule may no longer have one or more of the allotypic determinants 11,5,13,14,10,6,24,21,15, 16, 26 and 27.
The constant region may be from an immunoglobulin of the isotype IgA2 and the molecule may no longer have either or both of the allotypic determinants 1 and 2.
The present invention also provides a pharmaceutical preparation which comprises a molecule as defined.
The present invention also provides a reagent which comprises a molecule as defined.
The present invention also provides a nucleotide sequence encoding a molecule as defined.
The present invention also provides cloning and expression vectors comprising a nucleotide sequence as delivered above.
The present invention also provides host cells comprising a cloning or expression vector as defined above.
The present invention also provides a method of preparing a molecule as defined above which comprises the steps of:
(a) identifying a constant region of an immunoglobulin heavy chain;
(b) comparing the identified constant region with constant regions from immunoglobulin heavy chains of the same isotype to locate allotypic determinants in the identified constant region;
(c) obtaining the coding sequence for the identified constant region having allotypic determinants;
(d) altering the coding sequence so that it codess for a molecule without at least one of said allotypic determinants and by making the amino acid residues at the site for an allotypic determinant identical to the amino acid residues from the corresponding position in an equivalent immunoglobulin constant region of an isotype different to that of said identified constant region;
(e) using said altered coding sequence in an expression system to produce a said molecule.
The present invention also provides a method of treating a patient which comprises administering a pharmaceutical preparation as defined above.
Of course, there are a number of different strategies which could be used in order to make the molecules with fewer allotypic determinants.
Genes encoding therapeutically useful antibodies are generally available in one of several different forms. They may be available as a cloned variable region DNA sequence with restriction sites at each end, suitable for recloning along with a chosen cloned constant region DNA sequence into a suitable expression vector. This is the strategy described herein for the constructs TF57-19, MTF121 and MTF123. Alternatively, they may be available as complete immunoglobulin DNA sequences including the V and C regions together, e.g. a cDNA clone of a complete humanised or human antibody.
Whatever the form in which the cloned imrπunoglobulion gene is obtained, the next step is to predict the amino acid sequence of the constant region from the DNA sequence. The DNA sequence can be obtained using a variety of strategies familiar to molecular biologists. The predicted amino acid sequence would then be checked for the amino acids known to vary as allotypes. Any isoallotypes present within the sequence can be left unaltered. Any allotypes present can be mutated.
The next step, is to decide what amino acid sequence to mutate the allotype to, in order to imitate an isoallotype. This is done by lining up the sequence with the corresponding region of the other immunoglobulin isotypes. For all known allotypes, it has been found that one or more of the other isotypes have invariant sequences for the homologous region. One of these sequences can then be chosen to form the basis for the changes to be made in the allotype in question. Having predicted the new amino acid sequence for the constant region, it is necessary to alter the existing DNA clone or to create a new DNA clone which will encode this sequence. Again there are several strategies available to molecular biologists in order to achieve this. In the case of the example CAMPATH-1H constructs described herein, the gamma-1 constant region was cloned in an M13TG131 single stranded phage vector. Mutagenic oligonucleotides were synthesised which were largely homologous to the single strand, but which contained base changes necessary to alter the codons for the critical amino acids. The mutagenesis was carried out using a commercial kit from Amersham International, High Wycombe, Bucks. Alternatively it would be possible to synthesise a complete artificial gene which encodes the predicted sequence.
Once mutated or newly synthesised, the gene is ready for expression. There are many different expression vectors available. Some of these are more suitable for expression in restricted cell types. Again it is within the standard technical expertise of one skilled in this field to choose and adapt expression vectors for this purpose. In the case of the CAMPATH-1H constructs described herein, modifications of the pSVgpt and pSVneo vectors have been used. These vectors have convenient cloning sites for the immunoglobulin variable and constant region, encoding DNA fragments adjacent to suitable promoter and enhancer sequences to allow expression in lymphoid cells. The vector allows the easy independent replacement of variable or constant region encoding DNA fragments. Thus, any suitable variable region can be subcloned into the vector, to give rise to a new specificity, or the variable region can be kept and the constant region changed to give rise to a new isotype or allotype. Alternative vector systems are readily available.
Having removed allotypes from heavy chain constant regions by mutating them all to isoallotypes, it may still be desirable to consider the light chain effect in stimulating an immune response.
The most common kappa light chain allotype is Km(3) in the general population. Therefore it may be sufficient to utilise this common kappa light chain allotype, as relatively few members of the population would see it as foreign.
Alternatively there are no lambda light chain allotypes. Therefore they could be used in combination with the de-allotyped molecules derivable from heavy chain constant regions.
Where one utilises the kappa light chain, the allotype Km(1,2) could first be mutated to the allotype Km(1). The light chain allotype Km(1) is often only weakly recognized in combination with certain heavy chain classes and subclasses, and so may not cause a major problem in therapeutic use.
In order that the present invention is more fully understood embodiments will now be described in more detail, by way of example only, and not by way of limitation. Reference will be made (and has already been made in the text above) to the following figures in which:
figure 1 illustrates the structure of an IgG antibody;
figure 2 shows the allotypes for the IgG1 antibody CAMPATH-1H;
figure 3 shows schematically the preparation of IgG1 mutants;
figure 4 shows the IgG1 G1m (1,2,17) allotype sequence aligned to single allotypic examples of IgG2, 3 and 4 (none of these other subclasses have allotypic residues which cover the same residues as for the IgG1 allotypes);
figure 5 shows the M13TG131 cloning vector containing the human gamma-1 constant region, showing cloning sites and modified polylinker;
figure 6 shows the original Hu4vH HuG1 pSVgpt expression vector and its modified version;
figure 7 shows the result of an ELISA assay testing different dilutions of the antibodies of mutants 1, 2 and wild type CAMPATH-1H for IgG1 specificity;
figure 8 shows the result of an autologous complement mediated lysis test using human peripheral blood lymphocytes; and
figure 9 illustrates an antibody-dependent cell- mediated cytotoxicity assay (ADCC) using CD3 activated interleukin-2 expanded human blastocytes cell effectors (E) and targets (T).
The starting antibody used for site-directed mutagenesis was CAMPATH-1H, a monoclonal antibody with a kappa light chain containing the human constant region sequence for IgG1 which carries the G1m (1, 17) allelic determinants. The whole IgG1 encoding region exists as approximately 2.3 kb HindIII-SphI restriction fragment cloned in an M13 vector. The M13TG131 cloning vector containing the human gamma-1 constant region showing cloning sites and modified polylinker is shown in figure 5.
The IgG1 encoding region is entered in the EMBL Sequence Database under the code number HS1GCC4. The accession number is AC J00228 (the printout from the database is provided herein as Appendix 1). This sequence is for the G1m (1, 17) allotype. It covers 2009 bases from the 5' Hindlll site (A)AGCTT including all of the coding region. It does not however, include some of the 3' non-coding region up to the SphI site. The sequence provided by the EMBL Database is that of the upper strand of DNA. The CH1 domain starts at nucleotide 210 and ends at nucleotide 503. The mutagenic oligonucleotides MO1 and MO4 hybridise to nucleotides 486 to 510. The hinge region starts at nucleotide 892 and ends at nucleotide 936. The CH2 domain starts at nucleotide 1481 and ends at nucleotide 1803. The mutagenic oligonucleotide MO2 hybridises to nucleotides 1515 to 1543. The essential signal for the poly A tail is provided by nucleotides 1902 to 1908.
In M13TG131, the IgG1 coding region exists as a 2260 nucleotide fragment, of which the final 251 nucleotides are non-coding and therefore, inessential. Therefore, an embodiment of the invention could be carried out using the sequence information provided by the EMBL Sequence Database. It should be noted however, that the Sph1 restriction site referred to above, is present in the 3' end inessential non-coding region. Therefore, if the sequence data as provided by the EMBL database were being used, alternative restriction sites would have to be utilised.
Using site-directed mutagenesis, (carried out using protocols and reagents as supplied in kit form, Amersham code RPN. 1523, Amersham International Pic, Amersham, UK) the sequence corresponding to the G1m ( 1 ) allele was converted to the corresponding sequence found in the other sub-classes for IgG (Asp G1u Leu to G1u G1u Met at positions 356-358 in the CH3 domain).
The mutagenic oligonucleotides used were:
a) MOl (to convert G1m (17) to G1m (3))
5' CTC TCA CCA ACT CTC TTG TCC ACC T 3 '; b) MO2 (to convert G1m (1) to its isoallotype nG1m (1)) 5' GGT TCT TGG TCA TCT CCT CCC GGG ATG GG 3'; and c) M04 (to eliminate G1m(3) by changing Lys to Thr in the CH1 region)
5' CTC TCA CCA ACA GTC TTG TCC ACC T 3'.
The oligonucleotides were synthesised and then purified using an automated synthesizer and oligo purification columns supplied by Applied Biosystems (Applied Biosystems, 850 Lincoln Drive, Foster City, California, 94404 USA) following the manufacturers recommended protocols. Mutations were checked by Sanger Dideoxy sequencing (Sanger, F.S., Nicklen, S., and Coulson, A.R., (1977) Proc. Natl. Acad. Sci., USA, 74, 5463) using standard protocols and kits. As this newly formed allotype sequence is found in all humans, there should be no immunological response to this alternative form of G1m (1). Additionally and similarly, the lysine residue responsible for the G1m (17) allotypic determinant was converted to an arginine residue corresponding to the G1m allele (Lys 214-Arg; mutant 1).
The gene for this new constant region of mutant 1 carrying these three changes has been sequenced, incorporated into an expression vector containing the CAMPATH-1H V-region and expressed together with the CAMPATH-1H light chain which had been introduced by co-transfection. A further mutant has been made by replacing the critical arginine residue associated with G1m (3) of mutant 1 with a threonine residue, to produce a heavy chain which is the equivalent of IgG2 and which should fail to react with both anti-G1m (17) and anti-G1m (3) antisera (mutant 2).
Mutant 2 has also been sequenced, re-cloned in an expression vector containing the CAMPATH-1H light chain.
The supernatants of growing cultures containing either of the two mutants were subsequently assayed for the expression of a human IgG1 kappa product.
The mutations were introduced with the oligonucleotides listed above. The modified Hu4vHG1pSVgpt vector shown in figure 6 was used to simplify the subcloning of all the new mutants into the expression vector, owing to the possibility of use of two different "sticky ends" Bam HI and NotI. The expression vectors and VH region sequences and expression, along with the light chains, in YO rat plasmacytoma cells are all as described in Riechmann L., Clark, M.R. Waldman H., Winter G. (1988) Nature 332, 323-327.
From the positive cultures, the producers of the largest amount of the IgG1 product were selected to obtain purified antibody for biological assays to determine their allotypes and biological effector functions. Example 1
An Enzyme-linked Immuno Sorbent Assay (ELISA) was performed to verify that an IgG1 type antibody was produced by the mutants. This was tested with microtiter plates coated with anti-CAMPATH-idiotype antibody (YID 13.9). Wild type CAMPATH-1H antibody served as control. The bound antibody was detected with biotin-labelled anti-human kappa reagents or anti-human IgG reagent (monoclonals NH3/41 and NH3/130 respectively although other suitable reagents are commonly available) and subsequent development with streptavidin horseradish peroxidase. Figure 7 illustrates the results obtained for:
TF 57-19 ("wild type" CAMPATH-1H antibody, 0) MTF 121 (mutant 1,Δ )
MTF 123 (mutant 2,□)
and the wild type CAMPATH-1H (▼) in a known amount as standard. The concentrations had been estimated, and the starting dilutions adjusted to 50 μg/ml in PBS/10 mg/ml BSA. The starting dilution was used to prepare 8 twofold dilutions.
The slope of the graph shows clearly that the CAMPATH-idiotype antibodies recognises mutants 1 and 2 to an extent equivalent to that of the wild type CAMPATH-1H, and that all three antibodies tested are present in similar concentrations as the standard. Example 2
The mutants' capability of autologous complement mediated lysis of human peripheral blood lymphocytes was tested.
Human peripheral blood mononuclear cells from a healthy donor were isolated from 60 ml defibrinated blood on a Lymphoprep gradient (Nyeggard & Co., AS, Oslo, Norway). The cell pellet was washed in IMDM (Iscove's Modification of Dulbecco's Medium, Flow Laboratories, Scotland), and the cells were labelled with 51Cr. The starting dilution of antibodies used in the test was 50 μg/ml in PBS, 10 μg/ml BSA (dilution 1). Dilution 1 was further diluted 8 times two-fold to a final dilution of 1/128. Wild type antibody diluted in the same manner was used as a control.
The result is illustrated in figure 8. As can be seen, both antibody mutants show a very similar result in lysing the blood mononuclear cells as the wild type. The efficiency of the mutants is almost identical.
Example 3
Experiments were conducted to investigate whether or not, the mutant antibodies were capable of antibody-dependent cell-mediated cytotoxicity (ADCC) using CD3 activated interleukin-2 expanded human blastocytes as effectors (E) and targets (T). Cells were generated and used as both effectors and targets essentially as described in Riechmann L., Clark M.R., Waldmann H., Winter G., 1988, Nature 322, 323-327.
Preparation of Target Cells (T)
5 ml of blastocytes (3 x 104 cells) were labelled with 51Cr for 1 h. After 1 h the cells were washed and transferred in 6 equal aliquots in 100 μl IMDM 1% BSA, to 6 x 10 ml tubes containing 100 μl of the antibodies of mutants 1 and 2, and the control. The tubes were incubated for 1.5 h at room temperature. The cells were then washed with 10 ml IMDM 1% BSA and resuspended in 1.5 ml IMDM 1% BSA.
Preparation of Effector Cells (E)
Unlabelled blastocytes (2 × 106) were diluted 100:1 and 30:1 in IMDM 1% BSA medium. The ratios 100:1 and 30:1 refer to the final absolute ratios of effectors to 51Cr labelled targets in the assay. Assays were performed in microtitre plates with a total volume of 200 μl per assay well. Thus 100 μl of targets at a concentration of 2 × 104 were put in each well ie 2 x 103 total cells. For E:T of 100:1, 100 μl of effectors at 2 x 104 were plated per well ie 2 × 105. For E:T of 30:1 100 μl of effectors at 6 × 105 were put into each well ie 6 x 104 total cells.
The efficiency percentage of specific 51Cr release was calculated as follows:
% specific 51Cr release = (test release cpm - spontaneous (cpm) × 100
(total cpm - spontaneous cpm)
cpm = radioactive counts per minute as measured on a counter.
The result is shown in figure 9. The figure shows that all of the antibodies tested released chromium. Wild type TF 57-19 and mutant 2 (MTF 123) released at about equal levels, whereas mutant 1 (MTF 121) shows a slightly higher release.
These results clearly show that the mutants have biological activity comparable to the wild type CAMPATH- 1H antibody.
Example 4
The antibodies were tested in an assay specific for their G1m (3) allotypes reactivity using a monoclonal reagent from Oxoid (WHO/IVISS recognised agent, Study
Code No HP 6027). These tests were performed in replicates of two.
Microtiter plates were coated with the anti-CAMPATH idiotype YID 13.9.4 antibody captive, and divided into three arrays of 4 × 4 wells. Into each of the three arrays, 4 × 5 fold dilutions of the antibody TF 57-19, MTF 121 and MTF 123 (50 μg/ml) in PBS 1% BSA and a control solution of PBS/BSA each were added.
After an incubation of 45 minutes at room temperature, the antibody solution was removed, and (i) to the first array was added a 1:500 dilution of biotin-labelled anti-G1m (3);
(ii) to the second array was added a 1:100 dilution of biotin-labelled antibody (NH3/41) specific for the kappa light chain; and
(iii) to the third array was added a 1:1000 dilution of biotin-labelled antibody (NH3/130) specific for human IgG1.
The microtiter plate was developed with streptavidin horseradish peroxidase.
The result is illustrated in Table 1. The numbers in the results represent the optical density (O.D) as measured in an ELISA plate reader multiplied by 100 ie 12 represents an O.D of 0.12 and 70 an O.D of 0.70.
The result clearly shows, that samples 1-3 all react with the antibodies specific for IgG1 (see also Example 1 above) and the kappa light chains. The control is negative. However, in the assay for G1m (3) specificity, only MTF 121 (mutant 1) shows reactivity, whereas the wild type TF 57-19, MTF 123 (mutant 2) and the PBS/BSA control did not show any response.
This result illustrates clearly that the elimination of sites recognised in the allotype response by sitedirected mutagenis of these sites can overcome the problems of allotypic immuno-reactions. Although the examples refer to the mutagenesis of IgG1 only, it will be clear to the person skilled in the art that other immunoglobulin isotypes can be similarly modified.
Example 5
The antibodies were tested in a conventional allotyping experiment using inhibition of red cell agglutination. The experiment was carried out using reagents supplied by the Central Laboratory of the Netherlands Red Cross, Blood Transfusion Service (PO Box 9190, 1006 AD Amsterdam, Netherlands).
Human blood group O Rhesus D red cells were washed and then aliquots separately labelled as described below with one of the following three relevant anti-RhD human sera having antibodies of known allotype.
(1) anti-D G1m(az) = G1m (1,17)
(2) anti-D G1m(x) = G1m (2)
(3) anti-D G1m(f) = G1m (3)
Coating of Red Cells with Anti-Rh Antibodies
One volume of packed washed red blood cells were incubated with 4 volumes anti-Rh serum and 4 volumes (phosphate) buffered saline (PBS) at 37ºC during 60 minutes. Every 15 minutes the cells were mixed by shaking.
After incubation the coated cells were washed four times with PBS and stored at 4°C in preservation fluid (although coated red blood cells can be stored at 4°C in PBS for one week). These coated red blood cells were then agglutinated with four antisera to the IgG1 allotypes as follows using the recommended dilution for each antiserum.
(1) anti-G1m(z) = anti-G1m(17) 1 in 30 dilution (2) anti-G1m(a) = anti-G1m(l) 1 in 30 dilution
(3) anti-G1m(x) = anti-G1m(2) 1 in 20 dilution
(4) anti-G1m(f) = anti-G1m(3) 1 in 30 dilution The wild-type CAMPATH-1H TF57-19 or the different
CAMPATH-1H constructs (MTF 121, MTF 123) with the altered gamma-1 constant regions were then tested for their abilities to inhibit the agglutination of the red cells by the above antisera. The inhibiting antibodies were tried at concentrations of 0.5mg/ml, 0.25mg/ml and 0.125mg/ml in phosphate buffered saline containing 5% foetal bovine serum. Control sera containing IgG1 of allotype G1m(zax) or G1m(f) [G1m(1,2,17) or G1m(3)] were also included in the experiment and were used at dilutions of 1 in 10,20 and 40. Where it occurred the inhibition was most easily seen for the CAMPATH-1H antibodies at the 0.5mg/ml concentration and it was much weaker for 0.25mg/ml and no inhibition was seen at 0.125mg/ml. The control sera inhibited at all three dilutions tested. The results for the highest concentration are shown below.
Allotype CAMPATH-1H constructs Control sera TF57-19 MTF121 MTF123 G1m(1,2,17) G1m(3) G1m(1) + - - + - G1m(2) - - - +
- G1m(3) + - +
- - G1m(17) + - - +
-
The results are therefore consistent with the original wild type CAMPATH-1H antibody TF57-19 having allotype G1m(1,17). The new mutant MTF121 type as allotype G1m(3) whilst the mutant MTF123 fails to allotype for any of the IgG1 allotype markers G1m( 1, 2, 3, 17 ) i.e. it appears not to have an IgG1 allotype.
The skilled man will be able to use the binding molecules hereby provided to make pharmaceuticals according to standard techniques. Similarly the pharmaceuticals can be used in accordance with standard practices.
Figure imgf000034_0001
APPENDIX 1 - Sheet (a)
HSIGCC4 2009 bases
Human ig germline g-e-a region a: gamma-1 constant region
ID HSIGCC4 standard; DNA; PRI; 2009 BP.
AC J00228;
DT 23-APR-1990 (reference update)
DT 18-JUL-1985 (incorporated)
DE Human ig germline g-e-a region a: gamma-1 constant
DE region
KW constant region; gamma-immunoglobulin; germ line;
KW hinge exon; immunoglobulin; immunoglobulin heavy
KW chain.
OS Homo sapiens (human)
OC Eukaryota; Metazoa; Chordata; Vertebrata; Tetrapoda;
OC Mammalia; Eutheria; Primates.
RN [1] (bases 1-2009)
RA Ellison J.W., Berson B.J., Hood L.E.;
RT "The nucleotide sequence of a human immunoglobulin
RT c-gamma-1 gene";
RL Nucleic Acids Res. 10:4071-4079(1982).
RN [2] (bases 469-1070, 1465-1821)
RA Takahashi N., Ueda S., Obata M., Nikaido T.,
RA Nakai S., Honjo T.;
RT "Structure of human immunoglobulin gamma genes:
RT Implications for evolution of a gene family";
RL Cell 29:671-679(1982).
CC [1] and [2] report that nucleotide divergence among
CC the four gamma genes is much greater in the hinge
CC regions than anywhere else. [2] also reports the
CC hinge regions of gamma-2, gamma-3, gamma-4, a gamma
CC pseudogene, and the 5' flanking, ch2, and ch3
CC domains of the gamma genes.
CC
CC this entry is part of a multigene region (region a)
CC containing the gamma-3, gamma-1, pseudo-epsilon, and
CC alpha-1 genes. see segment 1 for more comments.
Key Location/Qualifiers
FT CDS 210..503
FT /note= " Ig gaπuna-1 heavy chain APPENDIX 1 - cont. Sheet (b)
FT c-region chl domain (aa at 212)"
FT conflict 563..563
FT /citation=([1],[2])
FT /note="T in [1]; c in [2]"
FT conflict 593..593
FT /citation=([1],[2])
FT /note="C in [1]; t in [2]"
FT conflict 614..614
FT /citation=([1],[2])
FT /note="G in [1]; a in [2]"
FT conflict 633.-633
FT /citation= ([1],[2])
FT /note="G in [1]; gg in [2]"
FT conflict 643..643
FT /citation=([1],[2])
FT /note="G in [1]; a in [2]"
FT conflict 654..654
FT /citation=( [1] , [2] )
FT /note="G in [1]; a in [2]"
FT conflict 684..684
FT /citation=([1],[2])
FT /note="C in [1]; cc in [2]"
FT conflict 692..692
FT /citation=([1],[2])
FT /note="G in [1]; a in [2]"
FT conflict 765..766
FT /citation=([1],[2])
FT /note="Aa in [1]; a in [2]"
FT CDS 892..936
FT /note="Ig gamma-1 heavy chain
FT c-region hinge"
FT CDS 1055..1384
FT /note="Ig gamma-1 heavy chain
FT c-region ch2 domain"
FT conflict 1475..1475
FT /citation=([1],[2])
FT /note="C in [1]; cc in [2]"
FT CDS 1481..1803
FT /note="Ig gamma-1 heavy chain
FT c-region ch3 domain"
FT conflict 1578..1578
FT /citation---([1],[2])
FT /note="T in [1]; c in [2]"
SQ Sequence 2009 BP; 418 A; 698 C; 566 G; 327 T; O
SQ Other; PPENDIX 1 - cont. Sheet (c)
10 20 30 40 50 60
1 AGCTTTCTGG GGCAGGCCAG GCCTGACCTT GGCTTTGGGG CAGGGAGGGG GCTAAGGTGA
61 GGCAGGTGGC GCCAGCAGGT GCACACCCAA TGCCCATGAG CCCAGACACT GGACGCTGAA
121 CCTCGCGGAC AGTTAAGAAC CCAGGGGCCT CTGCGCCTGG GCCCAGCTCT GTCCCACACC
181 GCGGTCACAT GGCACCACCT CTCTTGCAGC CTCCACCAAG GGCCCATCGG TCTTCCCCCT
241 GGCACCCTCC TCCAAGAGCA CCTCTGGGGG CACAGCGGCC CTGGGCTGCC TGGTCAAGGA
310 320 330 340 350 360
301 CTACTTCCCC GAACCGGTGA CGGTGTCGTG GAACTCAGGC GCCCTGACCA GCGGCGTGCA
361 CACCTTCCCG GCTGTCCTAC AGTCCTCAGG ACTCTACTCC CTCAGCAGCG TGGTGACCGT
421 GCCCTCCAGC AGCTTGGGCA CCCAGACCTA CATCTGCAAC GTGAATCACA AGCCCAGCAA 481 CACCAAGGTG GACAAGAAAG TTGGTGAGAG GCCAGCACAG GGAGGGAGGG TGTCTGCTGG
541 AAGCAGGCTC AGCGCTCCTG CCTGGACGCA TCCCGGCTAT GCAGCCCCAG TCCAGGGCAG
610 620 630 640 650 660 601 CAAGGCAGGC CCCGTCTGCC TCTTCACCCG GAGCCTCTGC CCGCCCCACT CATGCTCAGG 661 GAGAGGGTCT TCTGGCTTTT TCCCAGGCTC TGGGCAGGCA CAGGCTAGGT GCCCCTAACC 721 CAGGCCCTGC ACACAAAGGG GCAGGTGCTG GGCTCAGACC TGCCAAGAGC CATATCCGGG 781 AGGACCCTGC CCCTGACCTA AGCCCACCCC AAAGGCCAAA CTCTCCACTC CCTCAGCTCG 841 GACACCTTCT CTCCTCCCAG ATTCCAGTAA CTCCCAATCT TCTCTCTGCA GAGCCCAAAT
910 920 930 940 950 960
901 CTTGTGACAA AACTCACACA TGCCCACCGT GCCCAGGTAA GCCAGCCCAG GCCTCGCCCT
961 CCAGCTCAAG GCGGGACAGG TGCCCTAGAG TAGCCTGCAT CCAGGGACAG GCCCCAGCCG
1021 GGTGCTGACA CGTCCACCTC CATCTCTTCC TCAGCACCTG AACTCCTGGG GGGACCGTCA
1081 GTCTTCCTCT TCCCCCCAAA ACCCAAGGAC ACCCTCATGA TCTCCCGGAC CCCTGAGGTC
1141 ACATGCGTGG TGGTGGACGT GAGCCACGAA GACCCTGAGG TCAAGTTCAA CTGGTACGTG
APPENDIX 1 - cont. Sheet (d)
1210 1220 1230 1240 1250 1260 1201 GACGGCGTGG AGGTGCATAA TGCCAAGACA AAGCCGCGGG AGGAGCAGTA CAACAGCACG
1261 TACCGGGTGG TCAGCGTCCT CACCGTCCTG CACCAGGACT GGCTGAATGG CAAGGAGTAC
1321 AAGTGCAAGG TCTCCAACAA AGCCCTCCCA GCCCCCATCG AGAAAACCAT CTCCAAAGCC 1381 AAAGGTGGGA CCCGTGGGGT GCGAGGGCCA CATGGACAGA GGCCGGCTCG GCCCACCCTC 1441 TGCCCTGAGA GTGACCGCTG TACCAACCTC TGTCCTACAG GGCAGCCCCG AGAACCACAG
1510 1520 1530 1540 1550 1560 1501 GTGTACACCC TGCCCCCATC CCGGGATGAG CTGACCAAGA ACCAGGTCAG CCTGACCTGC 1561 CTGGTCAAAG GCTTCTATCC CAGCGACATC GCCGTGGAGT GGGAGAGCAA TGGGCAGCCG 1621 GAGAACAACT ACAAGACCAC GCCTCCCGTG CTGGACTCCG ACGGCTCCTT CTTCCTCTAC 1681 AGCAAGCTCA CCGTGGACAA GAGCAGGTGG CAGCAGGGGA ACGTCTTCTC ATGCTCCGTG 1741 ATGCATGAGG CTCTGCACAA CCACTACACG CAGAAGAGCC TCTCCCTGTC TCCGGGTAAA 1810 1820 1830 1840 1850 1860 1801 TGAGTGCGAC GGCCGGCAAG CCCCGCTCCC CGGGCTCTCG CGGTCGCACG AGGATGCTTG 1861 GCACGTACCC CCTGTACATA CTTCCCGGGC GCCCAGCATG GAAATAAAGC ACCCAGCGCT 1921 GCCCTGGGCC CCTGCGAGAC TGTGATGGTT CTTTCCACGG GTCAGGCCGA GTCTGAGGCC 1981 TGAGTGGCAT GAGGGAGGCA GAGCGGGTC

Claims

1. A molecule which comprises an amino acid sequence derivable from part or all of the constant region of an immunoglobulin heavy chain which constant regions are of a particular isotype and have one or more allotypic determinants
wherein said amino acid sequence is substantially homologous to the amino acid sequence of said constant region, but has been altered so that it is without at least one of said allotypic determinants, by making the amino acid residues at the site of an allotypic determinant identical to the amino acid residues from the corresponding position in another equivalent immunoglobulin constant region of a different isotype.
2. A molecule according to claim 1 which comprises an amino acid sequence derivable from part or all of a human immunoglobulin constant region.
3. A molecule according to claim 1 or claim 2 which comprises one or more polypeptides together with said amino acid sequence.
4. A molecule according to claim 3 wherein the polypeptide comprises a functional biological domain.
5. A molecule according to claim 4 wherein the functional biological domain comprises a binding domain.
6. A molecule according to claim 5 wherein the binding domain is an immunoglobulin-like binding domain.
7. A molecule according to claim 6 in which said immunoglobulin-like binding domain and said amino acid sequence are derivable from the same or different sources.
8. A molecule according to any one of claims 1 to 7 wherein the constant region is from an immunoglobulin of the isotype IgG.
9. A molecule according to claim 8 wherein the isotype subclass is IgG1 and the molecule no longer has one or more of the allotypic determinants 1,2,3 and 17.
10. A molecule according to claim 8 wherein the isotype subclass is IgG2 and the molecule no longer has the allotypic determinant 23.
11. A molecule according to claim 8 wherein the isotype subclass is IgG3 and the molecule no longer has one or more of the allotypic determinants 11,5,13,14,10,6,24,21, 15,16,26 and 27.
12. A molecule according to any one of claims 1 to 7 wherein the constant region is from an immunoglobulin of the isotype IgA2 and the molecule no longer has either or both of the allotypic determinants 1 and 2.
13. A pharmaceutical preparation which comprises a molecule according to any one of claims 1 to 12.
14. A reagent which comprises a molecule according to any one of claims 1 to 12.
15. A nucleotide sequence encoding a molecule according to any one of claims 1 to 12.
16. A cloning or expression vector comprising a nucleotide sequence according to claim 15.
17. A host cell comprising a cloning or expression vector according to claim 16.
18. A method of preparing a molecule according to any one of claims 1 to 12 which comprises the steps of:
(a) identifying a constant region of an immunoglobulin heavy chain;
(b) comparing the identified constant region with constant regions from immunoglobulin heavy chains of the same isotype to locate allotypic determinants in the identified constant region;
(c) obtaining the coding sequence for the identified constant region having allotypic determinants;
(d) altering the coding sequence so that it codes for a molecule without at least one of said allotypic determinants and by making the amino acid residues at the site for an allotypic determinant identical to the amino acid residues from the corresponding position in an equivalent immunoglobulin constant region of an isotype different to that of said identified constant region;
(e) using said altered coding sequence in an expression system to produce a said molecule.
19. A method of treating a patient which comprises administering a pharmaceutical preparation according to claim 13.
PCT/GB1992/000445 1991-03-12 1992-03-12 Humanised antibodies having modified allotypic determinants WO1992016562A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
JP4505594A JPH06510659A (en) 1991-03-12 1992-03-12 Humanized antibodies with altered allotypic determinants

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
GB9105245.6 1991-03-12
GB919105245A GB9105245D0 (en) 1991-03-12 1991-03-12 Binding molecules

Publications (1)

Publication Number Publication Date
WO1992016562A1 true WO1992016562A1 (en) 1992-10-01

Family

ID=10691455

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/GB1992/000445 WO1992016562A1 (en) 1991-03-12 1992-03-12 Humanised antibodies having modified allotypic determinants

Country Status (5)

Country Link
EP (1) EP0575407A1 (en)
JP (1) JPH06510659A (en)
CA (1) CA2105980A1 (en)
GB (1) GB9105245D0 (en)
WO (1) WO1992016562A1 (en)

Cited By (74)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1995005468A1 (en) * 1993-08-16 1995-02-23 Lynxvale Limited Binding molecules containing at least an immunoglobulin constant domain with modified allotypic determinant
US5821337A (en) * 1991-06-14 1998-10-13 Genentech, Inc. Immunoglobulin variants
WO1999058572A1 (en) 1998-05-08 1999-11-18 Cambridge University Technical Services Limited Binding molecules derived from immunoglobulins which do not trigger complement mediated lysis
US6054297A (en) * 1991-06-14 2000-04-25 Genentech, Inc. Humanized antibodies and methods for making them
WO2001030852A1 (en) * 1999-10-28 2001-05-03 Cambridge University Technical Services Limited Production of immunoglobulins which binds heterologous fcr with modified affinity
US6617135B1 (en) 1999-08-09 2003-09-09 Emd Lexigen Research Center Corp. Multiple cytokine protein complexes
US6800738B1 (en) 1991-06-14 2004-10-05 Genentech, Inc. Method for making humanized antibodies
WO2005070963A1 (en) * 2004-01-12 2005-08-04 Applied Molecular Evolution, Inc Fc region variants
EP1724282A2 (en) 1997-05-21 2006-11-22 Biovation Limited Method for the production of non-immunogenic proteins
US7355008B2 (en) 2003-01-09 2008-04-08 Macrogenics, Inc. Identification and engineering of antibodies with variant Fc regions and methods of using same
US7432357B2 (en) 2004-01-22 2008-10-07 Merck Patent Gmbh Anti-cancer antibodies with reduced complement fixation
US7632497B2 (en) 2004-11-10 2009-12-15 Macrogenics, Inc. Engineering Fc Antibody regions to confer effector function
US7662925B2 (en) 2002-03-01 2010-02-16 Xencor, Inc. Optimized Fc variants and methods for their generation
US7740847B2 (en) 2004-08-04 2010-06-22 Applied Molecular Evolution, Inc. Variant Fc regions
US7786270B2 (en) 2006-05-26 2010-08-31 Macrogenics, Inc. Humanized FcγRIIB-specific antibodies and methods of use thereof
US20110070230A1 (en) * 2009-09-24 2011-03-24 Xbiotech, Inc. Method and devices for identifying and treating a subject who has developed an anti-antibody response
US7973136B2 (en) 2005-10-06 2011-07-05 Xencor, Inc. Optimized anti-CD30 antibodies
US8039592B2 (en) 2002-09-27 2011-10-18 Xencor, Inc. Optimized Fc variants and methods for their generation
US8084582B2 (en) 2003-03-03 2011-12-27 Xencor, Inc. Optimized anti-CD20 monoclonal antibodies having Fc variants
US8101720B2 (en) 2004-10-21 2012-01-24 Xencor, Inc. Immunoglobulin insertions, deletions and substitutions
US8124731B2 (en) 2002-03-01 2012-02-28 Xencor, Inc. Optimized Fc variants and methods for their generation
US8133982B2 (en) 2004-05-10 2012-03-13 Macrogenics, Inc. FcγRIIB specific antibodies and methods of use thereof
US8188231B2 (en) 2002-09-27 2012-05-29 Xencor, Inc. Optimized FC variants
US8187593B2 (en) 2002-08-14 2012-05-29 Macrogenics, Inc. FcγRIIB specific antibodies and methods of use thereof
US8318907B2 (en) 2004-11-12 2012-11-27 Xencor, Inc. Fc variants with altered binding to FcRn
US8388955B2 (en) 2003-03-03 2013-03-05 Xencor, Inc. Fc variants
US8394374B2 (en) 2006-09-18 2013-03-12 Xencor, Inc. Optimized antibodies that target HM1.24
US20130129739A1 (en) * 2009-12-10 2013-05-23 Bayer Intellectual Property Gmbh Neutralizing prolactin receptor antibodies and their therapeutic use
US8524867B2 (en) 2006-08-14 2013-09-03 Xencor, Inc. Optimized antibodies that target CD19
US8530627B2 (en) 2002-08-14 2013-09-10 Macrogenics, Inc. FcγRIIB specific antibodies and methods of use thereof
US8546543B2 (en) 2004-11-12 2013-10-01 Xencor, Inc. Fc variants that extend antibody half-life
US8691952B2 (en) 2005-12-30 2014-04-08 Merck Patent Gmbh Anti-CD19 antibodies with reduced immunogenicity
US8697071B2 (en) 2005-08-10 2014-04-15 Macrogenics, Inc. Identification and engineering of antibodies with variant Fc regions and methods of using same
US8795667B2 (en) 2007-12-19 2014-08-05 Macrogenics, Inc. Compositions for the prevention and treatment of smallpox
US8802820B2 (en) 2004-11-12 2014-08-12 Xencor, Inc. Fc variants with altered binding to FcRn
US8907066B2 (en) 2009-04-22 2014-12-09 Merck Patent Gmbh Antibody fusion proteins with a modified FcRn binding site
US8926973B2 (en) 2001-03-30 2015-01-06 Merck Patent Gmbh Reducing the immunogenicity of fusion proteins
US8946387B2 (en) 2002-08-14 2015-02-03 Macrogenics, Inc. FcγRIIB specific antibodies and methods of use thereof
US8951517B2 (en) 2003-01-09 2015-02-10 Macrogenics, Inc. Identification and engineering of antibodies with variant Fc regions and methods of using same
US8968730B2 (en) 2002-08-14 2015-03-03 Macrogenics Inc. FcγRIIB specific antibodies and methods of use thereof
US8993730B2 (en) 2008-04-02 2015-03-31 Macrogenics, Inc. BCR-complex-specific antibodies and methods of using same
US9029330B2 (en) 2005-12-30 2015-05-12 Merck Patent Gmbh Methods of treating cancer using interleukin-12p40 variants having improved stability
US9040041B2 (en) 2005-10-03 2015-05-26 Xencor, Inc. Modified FC molecules
US9051373B2 (en) 2003-05-02 2015-06-09 Xencor, Inc. Optimized Fc variants
US9096877B2 (en) 2009-10-07 2015-08-04 Macrogenics, Inc. Fc region-containing polypeptides that exhibit improved effector function due to alterations of the extent of fucosylation, and methods for their use
US9150656B2 (en) 2010-03-04 2015-10-06 Macrogenics, Inc. Antibodies reactive with B7-H3, immunologically active fragments thereof and uses thereof
US9200079B2 (en) 2004-11-12 2015-12-01 Xencor, Inc. Fc variants with altered binding to FcRn
US9243069B2 (en) 2008-04-02 2016-01-26 Macrogenics, Inc. HER2/neu-specific antibodies and methods of using the same
US9284375B2 (en) 2005-04-15 2016-03-15 Macrogenics, Inc. Covalent diabodies and uses thereof
US9296816B2 (en) 2005-04-15 2016-03-29 Macrogenics, Inc. Covalent diabodies and uses thereof
US9376495B2 (en) 2011-05-21 2016-06-28 Macrogenics, Inc. Deimmunized serum-binding domains and their use in extending serum half-life
US9441049B2 (en) 2010-03-04 2016-09-13 Macrogenics, Inc. Antibodies reactive with B7-H3 and uses thereof
US9475881B2 (en) 2010-01-19 2016-10-25 Xencor, Inc. Antibody variants with enhanced complement activity
US9487587B2 (en) 2013-03-05 2016-11-08 Macrogenics, Inc. Bispecific molecules that are immunoreactive with immune effector cells of a companion animal that express an activating receptor and cells that express B7-H3 and uses thereof
US9657106B2 (en) 2003-03-03 2017-05-23 Xencor, Inc. Optimized Fc variants
US9695233B2 (en) 2012-07-13 2017-07-04 Roche Glycart Ag Bispecific anti-VEGF/anti-ANG-2 antibodies and their use in the treatment of ocular vascular diseases
US9708408B2 (en) 2006-12-08 2017-07-18 Macrogenics, Inc. Methods for the treatment of disease using immunoglobulins having Fc Regions with altered affinities for FcγRactivating and FcγRinhibiting
US9714282B2 (en) 2003-09-26 2017-07-25 Xencor, Inc. Optimized Fc variants and methods for their generation
US9737599B2 (en) 2006-06-26 2017-08-22 Macrogenics, Inc. Combination of FcγRIIB-specific antibodies and CD20-specific antibodies and methods of use thereof
US9822181B2 (en) 2013-08-23 2017-11-21 Macrogenics, Inc. Bi-specific monovalent diabodies that are capable of binding CD123 and CD3, and uses thereof
US9889197B2 (en) 2005-04-15 2018-02-13 Macrogenics, Inc. Covalently-associated diabody complexes that possess charged coil domains and that are capable of enhanced binding to serum albumin
US9908938B2 (en) 2013-03-14 2018-03-06 Macrogenics, Inc. Bispecific molecules that are immunoreactive with immune effector cells that express an activating receptor and an antigen expressed by a cell infected by a virus and uses thereof
US9932400B2 (en) 2013-08-23 2018-04-03 Macrogenics, Inc. Bi-specific monovalent diabodies that are capable of binding to gpA33 and CD3, and uses thereof
US9963510B2 (en) 2005-04-15 2018-05-08 Macrogenics, Inc. Covalent diabodies and uses thereof
US10100116B2 (en) 2006-06-26 2018-10-16 Macrogenics, Inc. FcγRIIB-specific antibodies and methods of use thereof
US10344092B2 (en) 2013-08-09 2019-07-09 Macrogenics, Inc. Bi-specific monovalent Fc diabodies that are capable of binding CD32B and CD79b and uses thereof
US10717778B2 (en) 2014-09-29 2020-07-21 Duke University Bispecific molecules comprising an HIV-1 envelope targeting arm
WO2020154475A1 (en) 2019-01-23 2020-07-30 Molecular Templates, Inc. Proteins comprising modified immunoglobulin variable light chains
US10961311B2 (en) 2016-04-15 2021-03-30 Macrogenics, Inc. B7-H3 binding molecules, antibody drug conjugates thereof and methods of use thereof
US11254748B2 (en) 2005-04-15 2022-02-22 Macrogenics, Inc. Covalent diabodies and uses thereof
US11384149B2 (en) 2013-08-09 2022-07-12 Macrogenics, Inc. Bi-specific monovalent Fc diabodies that are capable of binding CD32B and CD79b and uses thereof
US11401348B2 (en) 2009-09-02 2022-08-02 Xencor, Inc. Heterodimeric Fc variants
US11820830B2 (en) 2004-07-20 2023-11-21 Xencor, Inc. Optimized Fc variants
US11932685B2 (en) 2007-10-31 2024-03-19 Xencor, Inc. Fc variants with altered binding to FcRn

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0328404A1 (en) * 1988-02-12 1989-08-16 Btg International Limited Modified antibodies

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0328404A1 (en) * 1988-02-12 1989-08-16 Btg International Limited Modified antibodies

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
NATURE, vol. 333, 30 June 1988, (London, GB), R.G. MAGE: "Designing antibodies for human therapies", pages 807-808, see the article (cited in the application) *
The Journal of Immunology, vol. 143, no. 11, 1 December 1989, (Baltimore, MD, US), G. HEINRICH et al.: "Characterization of a human T cell-specific antibody (CD7) with human constant and mouse variable regions", pages 3589-3597, see the whole document, especially front page abstract; page 3591, left-hand column: "Cloning of human IgG1 genes"; page 3591, right-hand column, lines 9-13; page 3592, right-hand column, lines 13-19; page 3593, line 34 - page 3594, line 4 *
W.E. PAUL, M.D.: "Fundamental Immunology", 1984, Raven Press, New York, US; chapter 9: J.B. FLEICHMAN et al.: "Immunoglobulins: Allotypes and Idotypes", see the whole document *

Cited By (142)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6800738B1 (en) 1991-06-14 2004-10-05 Genentech, Inc. Method for making humanized antibodies
US5821337A (en) * 1991-06-14 1998-10-13 Genentech, Inc. Immunoglobulin variants
US8075890B2 (en) 1991-06-14 2011-12-13 Genentech, Inc. Method for making humanized antibodies
US6054297A (en) * 1991-06-14 2000-04-25 Genentech, Inc. Humanized antibodies and methods for making them
US6407213B1 (en) 1991-06-14 2002-06-18 Genentech, Inc. Method for making humanized antibodies
US6639055B1 (en) 1991-06-14 2003-10-28 Genentech, Inc. Method for making humanized antibodies
US6719971B1 (en) 1991-06-14 2004-04-13 Genentech, Inc. Method for making humanized antibodies
WO1995005468A1 (en) * 1993-08-16 1995-02-23 Lynxvale Limited Binding molecules containing at least an immunoglobulin constant domain with modified allotypic determinant
EP1724282A2 (en) 1997-05-21 2006-11-22 Biovation Limited Method for the production of non-immunogenic proteins
US7597889B1 (en) 1998-05-08 2009-10-06 Cambridge Enterprise Limited Binding molecules derived from immunoglobulins which do not trigger complement mediated lysis
WO1999058572A1 (en) 1998-05-08 1999-11-18 Cambridge University Technical Services Limited Binding molecules derived from immunoglobulins which do not trigger complement mediated lysis
US6617135B1 (en) 1999-08-09 2003-09-09 Emd Lexigen Research Center Corp. Multiple cytokine protein complexes
WO2001030852A1 (en) * 1999-10-28 2001-05-03 Cambridge University Technical Services Limited Production of immunoglobulins which binds heterologous fcr with modified affinity
US8926973B2 (en) 2001-03-30 2015-01-06 Merck Patent Gmbh Reducing the immunogenicity of fusion proteins
US7662925B2 (en) 2002-03-01 2010-02-16 Xencor, Inc. Optimized Fc variants and methods for their generation
US8124731B2 (en) 2002-03-01 2012-02-28 Xencor, Inc. Optimized Fc variants and methods for their generation
US8734791B2 (en) 2002-03-01 2014-05-27 Xencor, Inc. Optimized fc variants and methods for their generation
US8093357B2 (en) 2002-03-01 2012-01-10 Xencor, Inc. Optimized Fc variants and methods for their generation
US8530627B2 (en) 2002-08-14 2013-09-10 Macrogenics, Inc. FcγRIIB specific antibodies and methods of use thereof
US8187593B2 (en) 2002-08-14 2012-05-29 Macrogenics, Inc. FcγRIIB specific antibodies and methods of use thereof
US8968730B2 (en) 2002-08-14 2015-03-03 Macrogenics Inc. FcγRIIB specific antibodies and methods of use thereof
US8946387B2 (en) 2002-08-14 2015-02-03 Macrogenics, Inc. FcγRIIB specific antibodies and methods of use thereof
US8039592B2 (en) 2002-09-27 2011-10-18 Xencor, Inc. Optimized Fc variants and methods for their generation
US8383109B2 (en) 2002-09-27 2013-02-26 Xencor, Inc. Optimized Fc variants and methods for their generation
US10184000B2 (en) 2002-09-27 2019-01-22 Xencor, Inc. Optimized Fc variants and methods for their generation
US10183999B2 (en) 2002-09-27 2019-01-22 Xencor, Inc. Optimized Fc variants and methods for their generation
US8093359B2 (en) 2002-09-27 2012-01-10 Xencor, Inc. Optimized Fc variants and methods for their generation
US9353187B2 (en) 2002-09-27 2016-05-31 Xencor, Inc. Optimized FC variants and methods for their generation
US8809503B2 (en) 2002-09-27 2014-08-19 Xencor, Inc. Optimized Fc variants and methods for their generation
US9193798B2 (en) 2002-09-27 2015-11-24 Xencor, Inc. Optimized Fc variants and methods for their generation
US8188231B2 (en) 2002-09-27 2012-05-29 Xencor, Inc. Optimized FC variants
US8858937B2 (en) 2002-09-27 2014-10-14 Xencor, Inc. Optimized Fc variants and methods for their generation
US8951517B2 (en) 2003-01-09 2015-02-10 Macrogenics, Inc. Identification and engineering of antibodies with variant Fc regions and methods of using same
US9028815B2 (en) 2003-01-09 2015-05-12 Macrogenics, Inc. Identification and engineering of antibodies with variant FC regions and methods of using same
US7355008B2 (en) 2003-01-09 2008-04-08 Macrogenics, Inc. Identification and engineering of antibodies with variant Fc regions and methods of using same
US10584176B2 (en) 2003-03-03 2020-03-10 Xencor, Inc. Fc variants with increased affinity for FcγRIIc
US8735545B2 (en) 2003-03-03 2014-05-27 Xencor, Inc. Fc variants having increased affinity for fcyrllc
US10113001B2 (en) 2003-03-03 2018-10-30 Xencor, Inc. Fc variants with increased affinity for FcyRIIc
US9663582B2 (en) 2003-03-03 2017-05-30 Xencor, Inc. Optimized Fc variants
US8388955B2 (en) 2003-03-03 2013-03-05 Xencor, Inc. Fc variants
US9657106B2 (en) 2003-03-03 2017-05-23 Xencor, Inc. Optimized Fc variants
US8084582B2 (en) 2003-03-03 2011-12-27 Xencor, Inc. Optimized anti-CD20 monoclonal antibodies having Fc variants
US9051373B2 (en) 2003-05-02 2015-06-09 Xencor, Inc. Optimized Fc variants
US9714282B2 (en) 2003-09-26 2017-07-25 Xencor, Inc. Optimized Fc variants and methods for their generation
EA009746B1 (en) * 2004-01-12 2008-04-28 Эпплайд Молекьюлар Эволюшн, Инк. Fc region variants
WO2005070963A1 (en) * 2004-01-12 2005-08-04 Applied Molecular Evolution, Inc Fc region variants
US8835606B2 (en) 2004-01-22 2014-09-16 Merck Patent Gmbh Anti-cancer antibodies with reduced complement fixation
US10017579B2 (en) 2004-01-22 2018-07-10 Meck Patent Gmbh Anti-cancer antibodies with reduced complement fixation
US10633452B2 (en) 2004-01-22 2020-04-28 Merck Patent Gmbh Anti-cancer antibodies with reduced complement fixation
US9617349B2 (en) 2004-01-22 2017-04-11 Merck Patent Gmbh Anti-cancer antibodies with reduced complement fixation
US7432357B2 (en) 2004-01-22 2008-10-07 Merck Patent Gmbh Anti-cancer antibodies with reduced complement fixation
US8133982B2 (en) 2004-05-10 2012-03-13 Macrogenics, Inc. FcγRIIB specific antibodies and methods of use thereof
US8784808B2 (en) 2004-05-10 2014-07-22 Macrogenics, Inc. FcγRIIB specific antibodies and methods of use thereof
US11820830B2 (en) 2004-07-20 2023-11-21 Xencor, Inc. Optimized Fc variants
US7740847B2 (en) 2004-08-04 2010-06-22 Applied Molecular Evolution, Inc. Variant Fc regions
US8101720B2 (en) 2004-10-21 2012-01-24 Xencor, Inc. Immunoglobulin insertions, deletions and substitutions
US7632497B2 (en) 2004-11-10 2009-12-15 Macrogenics, Inc. Engineering Fc Antibody regions to confer effector function
US8216574B2 (en) 2004-11-10 2012-07-10 Macrogenics, Inc. Engineering Fc antibody regions to confer effector function
US8318907B2 (en) 2004-11-12 2012-11-27 Xencor, Inc. Fc variants with altered binding to FcRn
US8883973B2 (en) 2004-11-12 2014-11-11 Xencor, Inc. Fc variants with altered binding to FcRn
US8852586B2 (en) 2004-11-12 2014-10-07 Xencor, Inc. Fc variants with altered binding to FcRn
US8802820B2 (en) 2004-11-12 2014-08-12 Xencor, Inc. Fc variants with altered binding to FcRn
US9803023B2 (en) 2004-11-12 2017-10-31 Xencor, Inc. Fc variants with altered binding to FcRn
US8546543B2 (en) 2004-11-12 2013-10-01 Xencor, Inc. Fc variants that extend antibody half-life
US10336818B2 (en) 2004-11-12 2019-07-02 Xencor, Inc. Fc variants with altered binding to FcRn
US8367805B2 (en) 2004-11-12 2013-02-05 Xencor, Inc. Fc variants with altered binding to FcRn
US8338574B2 (en) 2004-11-12 2012-12-25 Xencor, Inc. FC variants with altered binding to FCRN
US8324351B2 (en) 2004-11-12 2012-12-04 Xencor, Inc. Fc variants with altered binding to FcRn
US9200079B2 (en) 2004-11-12 2015-12-01 Xencor, Inc. Fc variants with altered binding to FcRn
US11198739B2 (en) 2004-11-12 2021-12-14 Xencor, Inc. Fc variants with altered binding to FcRn
US10093739B2 (en) 2005-04-15 2018-10-09 Macrogenics, Inc. Covalent diabodies and uses thereof
US10093738B2 (en) 2005-04-15 2018-10-09 Macrogenics, Inc. Covalent diabodies and uses thereof
US9889197B2 (en) 2005-04-15 2018-02-13 Macrogenics, Inc. Covalently-associated diabody complexes that possess charged coil domains and that are capable of enhanced binding to serum albumin
US9284375B2 (en) 2005-04-15 2016-03-15 Macrogenics, Inc. Covalent diabodies and uses thereof
US9296816B2 (en) 2005-04-15 2016-03-29 Macrogenics, Inc. Covalent diabodies and uses thereof
US9963510B2 (en) 2005-04-15 2018-05-08 Macrogenics, Inc. Covalent diabodies and uses thereof
US11254747B2 (en) 2005-04-15 2022-02-22 Macrogenics, Inc. Covalent diabodies and uses thereof
US11254748B2 (en) 2005-04-15 2022-02-22 Macrogenics, Inc. Covalent diabodies and uses thereof
US8697071B2 (en) 2005-08-10 2014-04-15 Macrogenics, Inc. Identification and engineering of antibodies with variant Fc regions and methods of using same
US9040041B2 (en) 2005-10-03 2015-05-26 Xencor, Inc. Modified FC molecules
US9574006B2 (en) 2005-10-06 2017-02-21 Xencor, Inc. Optimized anti-CD30 antibodies
US7973136B2 (en) 2005-10-06 2011-07-05 Xencor, Inc. Optimized anti-CD30 antibodies
US9029330B2 (en) 2005-12-30 2015-05-12 Merck Patent Gmbh Methods of treating cancer using interleukin-12p40 variants having improved stability
US8691952B2 (en) 2005-12-30 2014-04-08 Merck Patent Gmbh Anti-CD19 antibodies with reduced immunogenicity
US11208496B2 (en) 2005-12-30 2021-12-28 Cancer Research Technology Ltd. Anti-CD19 antibodies with reduced immunogenicity
US10072092B2 (en) 2005-12-30 2018-09-11 Merck Patent Gmbh Methods of use of anti-CD19 antibodies with reduced immunogenicity
US8957195B2 (en) 2005-12-30 2015-02-17 Merck Patent Gmbh Anti-CD19 antibodies with reduced immunogenicity
US7786270B2 (en) 2006-05-26 2010-08-31 Macrogenics, Inc. Humanized FcγRIIB-specific antibodies and methods of use thereof
US10100116B2 (en) 2006-06-26 2018-10-16 Macrogenics, Inc. FcγRIIB-specific antibodies and methods of use thereof
US11098125B2 (en) 2006-06-26 2021-08-24 Macrogenics, Inc. FcγRIIB-specific antibodies and methods of use thereof
US9737599B2 (en) 2006-06-26 2017-08-22 Macrogenics, Inc. Combination of FcγRIIB-specific antibodies and CD20-specific antibodies and methods of use thereof
US9803020B2 (en) 2006-08-14 2017-10-31 Xencor, Inc. Optimized antibodies that target CD19
US8524867B2 (en) 2006-08-14 2013-09-03 Xencor, Inc. Optimized antibodies that target CD19
US10626182B2 (en) 2006-08-14 2020-04-21 Xencor, Inc. Optimized antibodies that target CD19
US11618788B2 (en) 2006-08-14 2023-04-04 Xencor, Inc. Optimized antibodies that target CD19
US8394374B2 (en) 2006-09-18 2013-03-12 Xencor, Inc. Optimized antibodies that target HM1.24
US9040042B2 (en) 2006-09-18 2015-05-26 Xencor, Inc. Optimized antibodies that target HM1.24
US9708408B2 (en) 2006-12-08 2017-07-18 Macrogenics, Inc. Methods for the treatment of disease using immunoglobulins having Fc Regions with altered affinities for FcγRactivating and FcγRinhibiting
US11787871B2 (en) 2006-12-08 2023-10-17 Macrogenics, Inc. Methods for the treatment of disease using immunoglobulins having fc regions with altered affinities for FcgammaRactivating and FegammaRinhibiting
US10711069B2 (en) 2006-12-08 2020-07-14 Macrogenics, Inc. Methods for the treatment of disease using immunoglobulins having Fc regions with altered affinities for FcγRactivating and FcγRinhibiting
US11932685B2 (en) 2007-10-31 2024-03-19 Xencor, Inc. Fc variants with altered binding to FcRn
US8795667B2 (en) 2007-12-19 2014-08-05 Macrogenics, Inc. Compositions for the prevention and treatment of smallpox
US9695236B2 (en) 2008-04-02 2017-07-04 Macrogenics, Inc. BCR-complex-specific antibodies and methods of using same
US9243069B2 (en) 2008-04-02 2016-01-26 Macrogenics, Inc. HER2/neu-specific antibodies and methods of using the same
US9469692B2 (en) 2008-04-02 2016-10-18 Macrogenics, Inc. HER2/neu-specific antibodies and methods of using same
US11028183B2 (en) 2008-04-02 2021-06-08 Macrogenics, Inc. HER2/neu-specific antibodies and methods of using same
US8993730B2 (en) 2008-04-02 2015-03-31 Macrogenics, Inc. BCR-complex-specific antibodies and methods of using same
US10131713B2 (en) 2008-04-02 2018-11-20 Macrogenics, Inc. HER2/neu-specific antibodies and methods of using same
US10479831B2 (en) 2008-04-02 2019-11-19 Macrogenics, Inc BCR-complex-specific antibodies and methods of using same
US8907066B2 (en) 2009-04-22 2014-12-09 Merck Patent Gmbh Antibody fusion proteins with a modified FcRn binding site
US11401348B2 (en) 2009-09-02 2022-08-02 Xencor, Inc. Heterodimeric Fc variants
US20110070230A1 (en) * 2009-09-24 2011-03-24 Xbiotech, Inc. Method and devices for identifying and treating a subject who has developed an anti-antibody response
EP2480252A4 (en) * 2009-09-24 2014-04-30 Xbiotech Inc Methods, compositions, and kits for reducing anti-antibody responses
EP2480252A1 (en) * 2009-09-24 2012-08-01 XBiotech, Inc Methods, compositions, and kits for reducing anti-antibody responses
US9096877B2 (en) 2009-10-07 2015-08-04 Macrogenics, Inc. Fc region-containing polypeptides that exhibit improved effector function due to alterations of the extent of fucosylation, and methods for their use
US9241989B2 (en) * 2009-12-10 2016-01-26 Bayer Intellectual Property Gmbh Neutralizing prolactin receptor antibodies and their therapeutic use
US20130129739A1 (en) * 2009-12-10 2013-05-23 Bayer Intellectual Property Gmbh Neutralizing prolactin receptor antibodies and their therapeutic use
US9475881B2 (en) 2010-01-19 2016-10-25 Xencor, Inc. Antibody variants with enhanced complement activity
US9441049B2 (en) 2010-03-04 2016-09-13 Macrogenics, Inc. Antibodies reactive with B7-H3 and uses thereof
US9714296B2 (en) 2010-03-04 2017-07-25 Macrogenics, Inc. Antibodies reactive with B7-H3, immunologically active fragments thereof and uses thereof
US9714295B2 (en) 2010-03-04 2017-07-25 Macrogenics, Inc. Antibodies reactive with B7-H3, immunologically active fragments thereof and uses thereof
US10730945B2 (en) 2010-03-04 2020-08-04 Macrogenics, Inc. Antibodies reactive with B7-H3 and users thereof
US10683364B2 (en) 2010-03-04 2020-06-16 Macrogenics, Inc. Antibodies reactive with B7-H3, immunologically active fragments thereof and uses thereof
US9896508B2 (en) 2010-03-04 2018-02-20 Macrogenics, Inc. Antibodies reactive with B7-H3 and uses thereof
US9150656B2 (en) 2010-03-04 2015-10-06 Macrogenics, Inc. Antibodies reactive with B7-H3, immunologically active fragments thereof and uses thereof
US9376495B2 (en) 2011-05-21 2016-06-28 Macrogenics, Inc. Deimmunized serum-binding domains and their use in extending serum half-life
US9695233B2 (en) 2012-07-13 2017-07-04 Roche Glycart Ag Bispecific anti-VEGF/anti-ANG-2 antibodies and their use in the treatment of ocular vascular diseases
US10683345B2 (en) 2012-07-13 2020-06-16 Roche Glycart Ag Bispecific anti-VEGF/anti-ANG-2 antibodies and their use in the treatment of ocular vascular diseases
US9487587B2 (en) 2013-03-05 2016-11-08 Macrogenics, Inc. Bispecific molecules that are immunoreactive with immune effector cells of a companion animal that express an activating receptor and cells that express B7-H3 and uses thereof
US10730947B2 (en) 2013-03-14 2020-08-04 Macrogenics, Inc. Bispecific molecules that are immunoreactive with immune effector cells that express an activating receptor and an antigen expressed by a cell infected by a virus and uses thereof
US9908938B2 (en) 2013-03-14 2018-03-06 Macrogenics, Inc. Bispecific molecules that are immunoreactive with immune effector cells that express an activating receptor and an antigen expressed by a cell infected by a virus and uses thereof
US11421031B2 (en) 2013-03-14 2022-08-23 Macrogenics, Inc. Bispecific molecules that are immunoreactive with immune effector cells that express an activating receptor and an antigen expressed by a cell infected by a virus and uses thereof
US11384149B2 (en) 2013-08-09 2022-07-12 Macrogenics, Inc. Bi-specific monovalent Fc diabodies that are capable of binding CD32B and CD79b and uses thereof
US10344092B2 (en) 2013-08-09 2019-07-09 Macrogenics, Inc. Bi-specific monovalent Fc diabodies that are capable of binding CD32B and CD79b and uses thereof
US9822181B2 (en) 2013-08-23 2017-11-21 Macrogenics, Inc. Bi-specific monovalent diabodies that are capable of binding CD123 and CD3, and uses thereof
US10858430B2 (en) 2013-08-23 2020-12-08 Macrogenics, Inc. Bi-specific monovalent diabodies that are capable of binding to gpA33 and CD3, and uses thereof
US10787521B2 (en) 2013-08-23 2020-09-29 Macrogenics, Inc. Bi-specific monovalent diabodies that are capable of binding CD123 and CD3, and uses thereof
US9932400B2 (en) 2013-08-23 2018-04-03 Macrogenics, Inc. Bi-specific monovalent diabodies that are capable of binding to gpA33 and CD3, and uses thereof
US10717778B2 (en) 2014-09-29 2020-07-21 Duke University Bispecific molecules comprising an HIV-1 envelope targeting arm
US10961311B2 (en) 2016-04-15 2021-03-30 Macrogenics, Inc. B7-H3 binding molecules, antibody drug conjugates thereof and methods of use thereof
US11591400B2 (en) 2016-04-15 2023-02-28 Macrogenics, Inc. B7-H3 directed antibody drug conjugates
WO2020154475A1 (en) 2019-01-23 2020-07-30 Molecular Templates, Inc. Proteins comprising modified immunoglobulin variable light chains

Also Published As

Publication number Publication date
CA2105980A1 (en) 1992-09-13
GB9105245D0 (en) 1991-04-24
JPH06510659A (en) 1994-12-01
EP0575407A1 (en) 1993-12-29

Similar Documents

Publication Publication Date Title
WO1992016562A1 (en) Humanised antibodies having modified allotypic determinants
AU662752B2 (en) Production of antibodies
KR101378302B1 (en) Antibodies and methods for making and using them
KR101385886B1 (en) Human antibodies and proteins
Pascual et al. Nucleotide sequence analysis of the V regions of two IgM cold agglutinins. Evidence that the VH4-21 gene segment is responsible for the major cross-reactive idiotype.
CA2501984C (en) Erythropoietin receptor binding antibodies
US20040175379A1 (en) Erythropoietin receptor binding antibodies
US11059907B2 (en) Antibodies that bind Notum Pectinacetylesterase
JP2002522063A (en) Generation of modified molecules with increased serum half-life
JP2004073210A (en) Modified antibody and product and process relating thereto
CZ69896A3 (en) Recombinant il4 antibodies usable for treating diseases mediated by il4
CA2106719C (en) Reshaped monoclonal antibodies against an immunoglobulin isotype
Parhami-Seren et al. Clustered H chain somatic mutations shared by anti-p-azophenylarsonate antibodies confer enhanced affinity and ablate the cross-reactive idiotype.
Coney et al. Apoptotic cell death induced by a mouse‐human anti‐APO‐1 chimeric antibody leads to tumor regression
JP2001510329A (en) Human monoclonal antibody
Demaison et al. Analysis of variable region genes encoding anti-Sm and anti-cardiolipin antibodies from a systemic lupus erythematosus patient.
Chapman et al. Pattern of usage of the VH4–21 gene by B lymphocytes in a patient with EBV infection indicates ongoing mutation and class switching
US6641999B1 (en) Probing method for identifying antibodies specific for selected antigens
Hall et al. Establishment, molecular rescue, and expression of 123AV16-1, a tumor-reactive human monoclonal antibody
Perlmutter The molecular genetics of phosphocholine-binding antibodies
Léger et al. Isolation and characterization of a monoclonal rheumatoid factor specific for the hinge region of rat IgG2b
RO116809B1 (en) Recombinant monclonal antibody, pharmaceutical combination and treating method employing the same

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): CA JP US

AL Designated countries for regional patents

Kind code of ref document: A1

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

DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
WWE Wipo information: entry into national phase

Ref document number: 1992906335

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 2105980

Country of ref document: CA

ENP Entry into the national phase

Ref document number: 1993 119044

Country of ref document: US

Date of ref document: 19931108

Kind code of ref document: A

WWP Wipo information: published in national office

Ref document number: 1992906335

Country of ref document: EP

WWW Wipo information: withdrawn in national office

Ref document number: 1992906335

Country of ref document: EP