CA2105980A1 - Humanised antibodies having modified allotypic determinants - Google Patents

Humanised antibodies having modified allotypic determinants

Info

Publication number
CA2105980A1
CA2105980A1 CA 2105980 CA2105980A CA2105980A1 CA 2105980 A1 CA2105980 A1 CA 2105980A1 CA 2105980 CA2105980 CA 2105980 CA 2105980 A CA2105980 A CA 2105980A CA 2105980 A1 CA2105980 A1 CA 2105980A1
Authority
CA
Canada
Prior art keywords
constant region
molecule
amino acid
allotypic
immunoglobulin
Prior art date
Legal status (The legal status 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 status listed.)
Abandoned
Application number
CA 2105980
Other languages
French (fr)
Inventor
Michael R. Clark
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Lynxvale Ltd
Original Assignee
Individual
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 Individual filed Critical Individual
Publication of CA2105980A1 publication Critical patent/CA2105980A1/en
Abandoned legal-status Critical Current

Links

Classifications

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

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

~VO 92/16562 ~ 8 l~ PCT/GB92/OQ445 Human1sPd antlbod1es hav~ng mod1f~ed allotypic determlnants 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 particular~y monoclonal antibodies (Kohler, G. and Milstein CO, 1975 Nature 256:495) represented a significant ~echnical break-throush with important consequences scientifically, commercially and therapeutically.
Monoclonal antibodies are made by establishing an immortal cell line whi~h is derived from a single immunoglobulin producing cell secreting one form of a biologically functional antibody molecule with a particular specificity.
Owing to their sp~cificity, the therapeutic applications of monoclonal an~ibodies 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 ~1059~
W092/]6562 " i PCT/GB92/0~5 human patients can lead to harmful hypersensitivity raactions. 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-lG
is an exception to this rule.
Thus, there is a need for therapeutic antibodies which have characteristic features speciic to human proteins. Unfortunately, immortal human antibody-producing cell lines are very difficult to establish and they give low yields of antibody (approximately 1 ,ug/ml).
In contrast, equivalent rodent cell lines yield high amounts o~ antibody (approximately 100 ~g/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 l). The light chains are of two types, either kappa or la~bda. Each of the H and L chains has a region ,: ~

:

~092/16~62 ~1 Q ~ PCT/GB92/0~5 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 th~! 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 :.

~ 1 ~3 ~ U
WO92/1fi~62 PCT/Gs92/0~45 -~

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 dif~erences 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 r antibodies having these sequence differences (eg a patient may have received a prior blood transfusion which containèd allotypically di~ferent immunoglobulins). A model system of in~ecting "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 IgGl, IgG2, IgG3 and IgG4.
There are 2 A recognised allotypes of human ~092/ ~ 8 ~
16562 PCT/GB92/0~i5 immunoglobulin distributed between the different isotypes as follows: -IgG1 x 4 IgG2 x IgG3 x 13 IgA2 x 2 IgE x .
Kappa x 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 Glm 1, 2, 3 and 17. Alternatively, combinations thereof, such as Glm (1, 17), can also be distinguished. The four different single allotypes are depicted in fisure 2.
Antisera can be raised in other non-human species whlch 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, nGlm(1) which is the isoallotype 2 1 0 ~ 0 WO92/16~62 PCT/GB92/0~i4s _ of Glm(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 IgGl, three common allelic forms of human IgGl occur with different irequencies within different racial ~roups, namely Glm (3~, Glm (1, 17), and Glm (1, 2, 17) based upon their reactivities with human antisera directed against the determinants Glm 1, 2, 3 and 17. At some 10 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 patlent 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 IgGl Glm (1, 2, 17) is ." ~ ..";. ..

~092/16562 ~1- Q ~ 9 8 0 PCT/GB92/0~45 shown aligned with sequences for the other human IgG, iso~ype sub-classes in figure 4 (a, b, c and d). It can be seen that the four isotypes are extrsmely homologous for the domains CHl, 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 IgGl Glm (1, 2, 17) have been marked in figure 4. However, for the purposes of clarity the sequences around the allotypic sites Glm (1) (2) and (17) are shown below for each isotype.
Site (l) Arg Asp or Glu Glu Leu or Met IgGl Arg Glu Glu Met IgG2 lS Arg Glu Glu Met IgG3 Gln Glu Glu Met IgG4 Thus, at site (l), IgGl may exist as several allotypes depending on whether aspartic acid or glutamic r acid at position 356, or leucine or methionine at 20 position 358 are present.
Site 2 Glu Gly or Ala Leu IgGl Glu Ala Leu IgG2 25 Glu Ala Leu IgG3 Glu Ala Leu IgG4 9 ~ 0 WO92/1656~ PCT/GB92/0~5 -~

Thus, at site (2), IgGl may exist as either of two allotypes depending on whether glycine or alanine is present at position 431.
Site (17)/~3) ~ , .
5 Sites (3) and (17) are alternative substitutions at the ~
same site. `
213 Z14 2l5 Lys Lys or Arg Val IgGl Lys Thr Val IgG2 10 Lys Arg Val IgG3 Lys Arg Val IgG4 Thus, at site (17)/(3), IgGl 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 Glm (l) 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 IyGl allele by site-directed mutagenesis of the gene, for example, an existing ~092/16562 ~ PCT/GB9t/0~45 CAMPATH-lH monoclonal antibody gena described below, so that the new allele consists entirely of isoallotypic determinants. The preparation of IgGl mutants according to the teaching provided by the present applicants is shown schematically in figure 3.
For Glm (1) and Glm (2), the changes comprise simple substitution by the alternative isoallotypic residues.
However, in the case of Glm (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 Glm (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 IgGl specific residues in close proximity in the CHl domain or hinge region. This indicates that in addition to changing lysine, other residues in CHl 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 IgGl and in particular, the CAMPATH-lH monoclonaI 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 ~inding molecule der:ivable from a second binding molecule;

2105~
WO92/16562 PCT/GB92/0~45 -which second binding molecule is an immunoglobulin, or a derivative, stru~tural 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 blnding molecule may comprise entirely isoallotypic determinants.
The second binding molecule may be selected from the group consisting of IgGl, IgG2, IgG3, IgA2, IgE, kappa light chains or-derivatives, structural or functional analogues thereof. Where the second binding molecule is IgGl, 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 ~0 92/lfi~62 ~ .1 n 5 ~ ~ Q PCT~GB92/0~45 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.

.. . . . . . . .. ...

~10~80 W092/16~62 PCT/GB92/O~S

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 incorporatin~ 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 `', .' . ' .
;' ' v092/]6562 ~1 Q~ PCT/GB92/0~45 determinant identlcal to the amino acid r~sidues fro~ 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 Aave the allotypic determinant 23. The isotype subclass may be IgG3 and the molecule may no longer have one or ~ore of the allotypic determinants 11,5,13,14,10,6,24,21,15, .. . . . .

~lO~Q
WO92/16562 pcT/Gs92fo~4 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 ,! , . -. ' . ' . ' .:, ' ' ~ ' ' ` ', ! ' . ' ' ' ' '; ' ' ' ` ~ ' ' `

~092/16562 ~1 Q 5 ~ ~ O PiCT/GB~2/0~5 -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 miake 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 - : ~ .. : :.. :: : . .

~10~) 98 0 WO 92/16562 PCr/GB92/00~5 humanised or human antibody.
Whatever the f orm in which the cloned immunoglobulion gene is obtained, the next step is to predict the amino acid sequence of the constant region 5 from the DNA sequence. The DNA sequence can be obtained using a variety of strategies familiar to molecular biologists. The predicted amino acid segiuence would then be checked for the amino acids known to vary as allotypes. Any isoallotypes present within the seguence lO 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 wlth 15 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 ...
20 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 Dl~1A clone which will encode this sequence. Again there are several strategies available 25 to molecular biologists in order to achieve this. In the ~ ` , case of the example CAMPATH-lH constructs described `;

"., ' ` ' .'. . ', ` , ' :~ i '' ' i ' ':~ . ' ' . . ` i ' i , ,,-. `

W092~16562 ? t O ~ PCT/GB92/0~45 herein, the gamma-l constant region was cloned in an Ml3TGl31 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-lH 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 - , . . : - - . . ~ , "

210a98~

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 ara 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 combinatlon 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(l). The light chain allotype Km(l) 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 W092~16~62 ~ ~ 5 ~ ~ o PCT/GB92/0~45 - 1~
limitation. Reference will be made (and has already been made in the text above) to the following figures in which:
figure 1 illustratas the structure of an IgG
antibody;
figure 2 shows the allotypes for the IgG1 antibody CAMPATH-lH;
figure 3 shows schematically the preparation of IgG1 mutants;
figure 4 shows the IgG1 Glm (1,2,17) allotype seguence 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 IgGl allotypes);
figure 5 shows the M13TGl31 cloning vector contai~ing the human gamma-l constant region, showing cloning sites and modified polylinker;
figure 6 shows the original Hu4vH HuGl pSVgpt expression vector and its modified version;
figure 7 shows the result of an ELISA assay testing different dilu-tions of the antibodies of mutants 1, 2 and wild type CAMPATH-lH 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-~10 2 ~ 8 0 W092/l6562 PCT/GB92/0~5 20mediated 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-lH, a monoclonal antibody with a kappa light chain containing the human constant region sequence for IgG1 which carries the Glm (1, 17) allelic determinants. The whole IgGl 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-l constant region showing cloning sites and modified polylinker is shown in figure 5.
The IgGl encoding region is entered in the EMBL
Sequence Database under the code number HSlGCC4. The accession number is AC J00228 (the printout from the database is provided herein as Appendix 1). This sequence is for the Glm (1, 17) allotype. It covers 2009 bases from the 5' HindIII 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 pro~ided 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 M01 and M04 hybridise to nucleotides 4B6 to 510. The hinge region starts at nucleotide 892 and ~092J16562 ~ 3 0 ' ~ ,~ n PCT/GB92/0~45 ends at nucleotide 936. The CH2 domain starts at nucleotide 1481 and ends at nucleotide 1803. The mutagenic oligonucleotide M02 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 fragmant, 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 E~L Sequence Database. It should be noted however, that the Sphl 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 sltes 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 Plc, Amersham, UK) the sPquence corresponding to the Glm (1~ allele was converted to the corresponding sequence found in the other sub-classes for IgG (Asp Glu Leu to Glu Glu Met at - positions 356-358 in the CH3 domain).
The mutagenic oligonucleotides used were-a) M01 (to convert Glm (17) to Glm (3)) 5' CTC TCA CCA ACT CTC TTG TCC ACC T 3';

' ~ `, 2~0~t381) W092/~6~62 PCT/GB92/0~45 -b) M02 (to convert Glm (1) to its isoallotype nGlm (1)) 5 ' GGT TCT TGG TCA TCT CCT CCC GGG ATG GG 3'; and c) M04 (to eliminate Glm(3) by chan~ing Lys to Thr in the C~1 ragion) 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 sequenclng (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 ~ormed allotype sequence is found in all humans, there should be no immunological response to this alternative form of Glm (1). Additionally and similarly, the lysine residue responsible for the Glm (17) allotypic determinant was converted to an arginine residue corresponding to the Glm allele (Lys 214-Arg; mutant 1).
The gene for this new constant region of mutant 1 carrying these three changes has been se~uenced, incorporated into an expression vector containing the CAMPATH-lH V-region and expressed together with the CAMPATH-lH light chain which had been introduced by co-transfection.

~/O 92/16562 2 ~ O ~ ~ 8 ~ PCl`/GB92/00445 A further mutant has been made by replacing the critical arginine residue associated with Glm (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-Glm (17) and anti-Glm (3) antisera (mutant 2).
Mutant 2 has also bsen sequenced, re-cloned in an expression vector containing the CAMPATH-lH 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 Hu4vHGlpSVgpt 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 Notl. The expression vectors and VH region sequences and expression, along with the light chains, in Y0 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 laryest amount of the IgGl product were selected to obtain purified antibody for biological assays to determine their allotypes and biological effector functions.

: ' ~, .

2~0.~9~0 W092/]6562 PCT/GB92/0~4~ -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 coatPd with anti-CAMPATH-idiotype antibody (YID
13.9). Wild type CAMPATH-lH 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:
~F 57-19 ("wild type" CAMPATH-lH antibody, 0) MTF 121 (mutant 1,~) MTF 123 (mutant 2, a ) and the wild type CAMPATH-lH (~) in a known amount as standard. The concentrations had been estimated, and the starting dilutions adjusted to 50 ,ug/ml in PBS/lO mg/ml BSA. The starting dilution was used to prepare 8 two-fold 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-lH, and that all three antibodies tested are present in similar concentrations as the standard.

~092/l6~62 PCT~GB92/O~S
0~9~) 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). ~he cell pellet was washed in IMDM (Iscove's Modification of Dulbecco's Medium, Flow Laboratories, Scotland), and the cells were labelled with 5lCr. 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 dllution o~
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.
Exam~le 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 ~lastocytes as effectors (E) and targets (T). Cells were generated and - used as both effectors and targets essentially as '.,':.;' , :' ' ' W092/16~62 PCT/GB92/0~5 described in Riechmann L., Clark M.R., Waldmann H., Winter G., lsa8, Nature 322, 323-327.
PreDaration of Target Cells ( T ) 5 ml of blastocytes (3 x 106 cells) were labelled with 51cr for 1 h. After 1 h the cells were washed and transferred in 6 equal aliquots in 100 ~1 IMDM 1~ BSA, to 6 x 10 ml tubes containing 100 ~1 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.
Pre~aration of Effector Cells (E) Unlabelled blastocytes (2 x 106) were diluted 100:1 and 30:1 in IMDM 1~ ~SA 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 ,ul per assay well. Thus 100 ~1 of tar~ets at a concentration of 2 x 104 were put in each well ie 2 x 103 total cells. For E:T of 100:1, 100 ~1 of effectors at 2 x 106 were plated per well ie 2 x 105. For E:T of 30:1 100 ~1 of effectors at 6 x 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 =

~092tl6~62 2 ~ 8 n PCT/GB92/0~5 (test release cpm - spontaneous (cpm) x 100 (total cpm - spontaneous cpm) cpm = radioactive counts per minute as measured on a counter.
The result is shown in fisure 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 ~he wild type CAMPATH-lH antibody.
Example 4 The antibodies were tes~ed in an assay specific for their Glm ~3) allotypes reactivlty 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 x 4 wells. Into each of the three arrays, 4 x 5 fold dilutions of the antibody TF 57-l9, MTF 121 and MTF 123 (50 ~g/ml~ in PBS l~ BSA and a control solution of PBS/BSA each were added.
After an incubation of 45 minutes at room temperature, th~ antibody solution was removed, and , ~ .

210:?~80 W092/]6~62 ~ PCT/~B92/0~5 (i) to the first~ array was added a 1:~00 dilution o biotin-labelled anti-Glm (3~;
(ii) to the second array was added a 1:100 dilution of biotin-labelled antibody (NH3/41) specific 5 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 IgGl (see also Example 1 above) and the kappa light chains. The control i5 negative. However, in the assay for Glm (3) specificity, only MTF 121 (mutant 1) shows reactivity, whereas the 20 wild type TF 57-19, MTF 123 (mutant 2) and the P~S/BSA
control did not show any response.
This result illustrates clearly that the elimination of sites recognised in the allotype response by site-directed mutagenis of these sites can overcome the problems of allotypic immuno-reactions. Although the examples refer to the mutagenesis of IgGl only, it will - . . ; ,. , , - : :

~092/16562 ~1 n ~ 9 8 0 PCT/GB92/0~45 be clear to ~he person skilled in the art that other immunoglobulin isotypes can be similarly modified.
Example 5 The antibodies were t:ested 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 Glm(az) = Glm (1,17) (2) anti-D Glm(x) = Glm (2) (3) anti-D Glm(f) = Glm (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 volwnes (phosphate) buffered saline (PBS) at 37C during 60 minutes. Every 15 minutes the cells were mixed by shakin~.
After incubation the coated cells were washed four times with PBS and stored at 4C in preservation fluid (although coated red blood cells can be stored at ~C in PBS for one week).

'~10~80 ~092/16562 PCT/GB92/O~

These coated red blood cells were then ag~lutina~ed with four antisera to the IgGl allotypes as follows using the recommended dilution for each antiserum.
( 1 ) anti-Glm( z ) = antl--Glm( 17 ) 1 in 30 dilution (2) anti-Glm(a) = anti-Glm(l) 1 in 30 dilution (3) anti-Glm(x) = anti-Glm(2) 1 in 20 diluti~n (4) anti-Glm(f) = anti-Glm(3) 1 in 30 dilution - The wild-type CAMPATH-lH TF57-19 or the different CAMPATH-lH 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 IgGl of allotype Glm(zax) or Glm(f) ~Glm(1,2,17) or Glm(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-lH
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-lH constructs Control sera ~r -~

~092/16562 ~ ~ PCT/GB92/0~5 TF57-19 MTFlZl MTF123 Glm(1,2,17) Glmt3) Glm(l) + - _ +
Glm(2) - - - + _ Glm(3) - + _ _ +
5 Glm(17) + - _ +
~', The results are therefore consistent with the ~ ;
oriyinal wild type CAMPATH-lH antibody TF57-19 having allotype Glm(l, 17). The new mutant MTF121 type as allotype Glm(3) whilst the mutant MTF123 fails to allotype for any of the IgGl allotype markers Glm(1,2,3,17) i.e. it appears not to have an IgGl 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.

21~8~
wo 92/16~62 PCI`/~B92/00445 _ ~ ~ ' ' -:' Z- O ~ . `~ `
-_ .. In U~ . .`
,~ ~O a) O ~
H ~
~ . _ .__ ~ .' .
C O ~

Z O ~ 0 0 0 ~able 1 _ :~ _ _ . ~

,:: o ~ D ,:, _l ~ . ' '.,.
. _ . . . , O <I~ :.' _ . .
~ O ~ ~
. - ~ ~ ~
~ N ~ ~ tO
~ - ~ ~ :;,:,~ ' _/ N C t` IJ~
~ : .
~ ,:
--I N
t~ ~3 _ ~ ' ... N
C~ O c) O
C~ .-1 ~,~ Ul _I v V
U~ Cl~
~ E ~ ~ 3 E~
Cl C: U~ ' I E~ , _ ~

._ ._ ._ SUB~i~3 ~ E ~EET `
`~

~092/16562 21 0 :~ 9 8 ~ PCT/GB92/0~45 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-l 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., ~erson B.J., Hood L.E.;
RT "The nucleotide sequence of a human immunoglobulin RT c-gamma-l 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-l, pseudo-epsilon, and CC alpha-l genes. see segment 1 for more comments.
Key Location/Qualifiers FT CDS 210............... 503 ;
FT ~note="Ig gamma-1 heavy chain .:

210 ~98~
WO92/16562 PCT/GB92/0~5 APPENDIX 1 - COIlt . Sheet (b) FT c-region chl domain (aa at 212)"
FT conflict 563... 563 FT /citation=([1],[2]) : :
FT /note="T in [l]; 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 [l]; 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 [l]; 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=([l]~t2]) ~, FT /note="Aa in [1]; a in [2]"
FT CDS 892... 936 FT /note="Ig gamma-1 heavy chain FT c-region hinge" r . .
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 /not-e="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, 0 SQ Other;

, ~,..

- . , .

WO 92/16562 2~ :~ Q ~PCr/GB92/oo~

o ~ O ~ U ~O O O ~O ~ D C U U ~ ~

~ o u u; ~ U ~ 5, e ~ ~;
o ~ ~ ~ y . yO y ~ y ,I y ~: u b U ~ '~~ ~ y ~ y ~ j a a o c~ ~., o .c U, ~ o U ~ y o 5 U a ¢ ~ , ¢ ~ J i ~ 1 o ~ rO ~D ~ es:) ~ ~ ~ ~ ~

~lQ~8~
WO 92/16562 PCI~/GB92/00445 -I U a ~ U

5 ~ ~ U ~
O ~ E~ ~ o ~ C~ o ~ ~ ~ V ~ E~
o ~ E~ ~ ~ ~ o ~ E~ o U ~ U

O j ~ ~ ~ u a U ~ ¢ ~ ¢ ~ U ¢ U: ~ ¢ ~ ¢ U

U t~ h ~ ¢ ~ U c~ u ~ c~

U ~ ~ ~ E - ~ ¢ ~ ~ U ¢ _,, ~ U ~g U
~ U E~ ~ V E~ ~ ¢ ~ ~ U U C~
¢ ~U ~ ~ U E~ E~ ¢ V h ~ ~ u ~
u h E~ t3 u ~ ¢ ~: h ~ ~ h O ~D ~ CO ~ O ~ r O ~ ~ CO

SUE3ST~T~ EFT

.
. ~ . .

Claims (13)

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.
CA 2105980 1991-03-12 1992-03-12 Humanised antibodies having modified allotypic determinants Abandoned CA2105980A1 (en)

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
CA2105980A1 true CA2105980A1 (en) 1992-09-13

Family

ID=10691455

Family Applications (1)

Application Number Title Priority Date Filing Date
CA 2105980 Abandoned CA2105980A1 (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)

Families Citing this family (74)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE122004000008I1 (en) 1991-06-14 2005-06-09 Genentech Inc Humanized heregulin antibody.
US6800738B1 (en) 1991-06-14 2004-10-05 Genentech, Inc. Method for making humanized antibodies
WO1994004679A1 (en) * 1991-06-14 1994-03-03 Genentech, Inc. Method for making humanized antibodies
GB9316989D0 (en) * 1993-08-16 1993-09-29 Lynxvale Ltd Binding molecules
EP0983303B1 (en) 1997-05-21 2006-03-08 Biovation Limited Method for the production of non-immunogenic proteins
GB9809951D0 (en) 1998-05-08 1998-07-08 Univ Cambridge Tech Binding molecules
US6617135B1 (en) 1999-08-09 2003-09-09 Emd Lexigen Research Center Corp. Multiple cytokine protein complexes
GB9925490D0 (en) * 1999-10-28 1999-12-29 Univ Cambridge Tech Binding molecules and treatment and screening methods
WO2002079415A2 (en) 2001-03-30 2002-10-10 Lexigen Pharmaceuticals Corp. Reducing the immunogenicity of fusion proteins
US7317091B2 (en) 2002-03-01 2008-01-08 Xencor, Inc. Optimized Fc variants
US7662925B2 (en) 2002-03-01 2010-02-16 Xencor, Inc. Optimized Fc variants and methods for their generation
US8188231B2 (en) 2002-09-27 2012-05-29 Xencor, Inc. Optimized FC variants
US20040132101A1 (en) 2002-09-27 2004-07-08 Xencor Optimized Fc variants and methods for their generation
US8946387B2 (en) 2002-08-14 2015-02-03 Macrogenics, Inc. FcγRIIB specific antibodies and methods of use thereof
US8530627B2 (en) 2002-08-14 2013-09-10 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
US8187593B2 (en) 2002-08-14 2012-05-29 Macrogenics, Inc. FcγRIIB specific antibodies and methods of use thereof
US7960512B2 (en) 2003-01-09 2011-06-14 Macrogenics, Inc. Identification and engineering of antibodies with variant Fc regions and methods of using same
AU2004204494B2 (en) 2003-01-09 2011-09-29 Macrogenics, Inc. Identification and engineering of antibodies with variant Fc regions and methods of using same
US8388955B2 (en) 2003-03-03 2013-03-05 Xencor, Inc. Fc variants
US20090010920A1 (en) 2003-03-03 2009-01-08 Xencor, Inc. Fc Variants Having Decreased Affinity for FcyRIIb
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
US8101720B2 (en) 2004-10-21 2012-01-24 Xencor, Inc. Immunoglobulin insertions, deletions and substitutions
JP4762156B2 (en) * 2004-01-12 2011-08-31 アプライド モレキュラー エボリューション,インコーポレイテッド Fc region variant
EP1706428B1 (en) 2004-01-22 2009-09-23 MERCK PATENT GmbH Anti-cancer antibodies with reduced complement fixation
US7521542B2 (en) 2004-05-10 2009-04-21 Macrogenics, Inc. Humanized FcγRIIB-specific antibodies and methods of use thereof
US20150010550A1 (en) 2004-07-15 2015-01-08 Xencor, Inc. OPTIMIZED Fc VARIANTS
ES2426817T3 (en) 2004-08-04 2013-10-25 Mentrik Biotech, Llc Fc regions variants
AU2005335714B2 (en) 2004-11-10 2012-07-26 Macrogenics, Inc. Engineering Fc antibody regions to confer effector function
US8802820B2 (en) 2004-11-12 2014-08-12 Xencor, Inc. Fc variants with altered binding to FcRn
EP2325207B1 (en) 2004-11-12 2017-03-15 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
US8367805B2 (en) 2004-11-12 2013-02-05 Xencor, Inc. Fc variants with altered binding to FcRn
US11254748B2 (en) 2005-04-15 2022-02-22 Macrogenics, Inc. Covalent diabodies and uses thereof
US9963510B2 (en) 2005-04-15 2018-05-08 Macrogenics, Inc. Covalent diabodies and uses thereof
US9284375B2 (en) 2005-04-15 2016-03-15 Macrogenics, Inc. Covalent diabodies and uses thereof
ES2707152T3 (en) 2005-04-15 2019-04-02 Macrogenics Inc Covalent diabodies and uses thereof
DK2573114T3 (en) 2005-08-10 2016-07-04 Macrogenics Inc The identification and production of antibodies with variant Fc regions, and methods of using same
DK1931709T3 (en) 2005-10-03 2017-03-13 Xencor Inc FC VARIETIES WITH OPTIMIZED FC RECEPTOR BINDING PROPERTIES
US7973136B2 (en) 2005-10-06 2011-07-05 Xencor, Inc. Optimized anti-CD30 antibodies
JP2009521912A (en) 2005-12-30 2009-06-11 メルク パテント ゲゼルシャフト ミット ベシュレンクテル ハフツング Anti-CD19 antibody with reduced immunogenicity
ATE555125T1 (en) 2005-12-30 2012-05-15 Merck Patent Gmbh INTERLEUKIN 12P40 VARIANTS WITH IMPROVED STABILITY
EP2021029B1 (en) 2006-05-26 2014-06-11 MacroGenics, Inc. Humanized fc gamma riib-specific antibodies and methods of use thereof
ES2599319T3 (en) 2006-06-26 2017-02-01 Macrogenics, Inc. Fc RIIB specific antibodies and their methods of use
EP2032159B1 (en) 2006-06-26 2015-01-07 MacroGenics, Inc. Combination of fcgammariib antibodies and cd20-specific antibodies and methods of use thereof
ME01786B (en) 2006-08-14 2014-09-20 Xencor Inc Optimized antibodies that target cd19
US8394374B2 (en) 2006-09-18 2013-03-12 Xencor, Inc. Optimized antibodies that target HM1.24
WO2008140603A2 (en) 2006-12-08 2008-11-20 Macrogenics, Inc. METHODS FOR THE TREATMENT OF DISEASE USING IMMUNOGLOBULINS HAVING FC REGIONS WITH ALTERED AFFINITIES FOR FCγR ACTIVATING AND FCγR INHIBITING
US8795667B2 (en) 2007-12-19 2014-08-05 Macrogenics, Inc. Compositions for the prevention and treatment of smallpox
HUE024903T2 (en) 2007-12-26 2016-02-29 Xencor Inc Fc variants with altered binding to fcrn
EP3045475B1 (en) 2008-04-02 2017-10-04 MacroGenics, Inc. Bcr-complex-specific antibodies and methods of using same
US8802093B2 (en) 2008-04-02 2014-08-12 Macrogenics, Inc. HER2/neu-specific antibodies and methods of using same
AU2010238858A1 (en) 2009-04-22 2011-12-08 Merck Patent Gmbh Antibody fusion proteins with modified FcRn binding sites
WO2011028952A1 (en) 2009-09-02 2011-03-10 Xencor, Inc. Compositions and methods for simultaneous bivalent and monovalent co-engagement of antigens
JP2013505938A (en) * 2009-09-24 2013-02-21 エックスバイオテク,インコーポレイテッド Methods, compositions and kits for reducing anti-antibody responses
JP5898082B2 (en) 2009-10-07 2016-04-06 マクロジェニクス,インコーポレーテッド Fc region-containing polypeptide exhibiting improved effector function by changing the degree of fucosylation and use thereof
EP2332995A1 (en) * 2009-12-10 2011-06-15 Bayer Schering Pharma Aktiengesellschaft Neutralizing prolactin receptor antibodies and their therapeutic use
US8362210B2 (en) 2010-01-19 2013-01-29 Xencor, Inc. Antibody variants with enhanced complement activity
US8802091B2 (en) 2010-03-04 2014-08-12 Macrogenics, Inc. Antibodies reactive with B7-H3 and uses thereof
PE20130479A1 (en) 2010-03-04 2013-05-12 Macrogenics Inc ANTIBODIES REACTIVE WITH B7-H3, IMMULOGICALLY ACTIVE FRAGMENTS OF THE SAME AND USES OF THE SAME
CN103154025B (en) 2010-08-02 2015-07-01 宏观基因有限公司 Covalent diabodies and uses thereof
MX347818B (en) 2011-05-21 2017-05-15 Macrogenics Inc Deimmunized serum-binding domains and their use for extending serum half-life.
HRP20211641T1 (en) 2012-07-13 2022-02-04 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
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
UA116479C2 (en) 2013-08-09 2018-03-26 Макродженікс, Інк. Bi-specific monovalent fc diabodies that are capable of binding cd32b and cd79b 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
EP2840091A1 (en) 2013-08-23 2015-02-25 MacroGenics, Inc. Bi-specific diabodies that are capable of binding gpA33 and CD3 and uses thereof
EP2839842A1 (en) 2013-08-23 2015-02-25 MacroGenics, Inc. Bi-specific monovalent diabodies that are capable of binding CD123 and CD3 and uses thereof
SG11201702544WA (en) 2014-09-29 2017-04-27 Univ Duke Bispecific molecules comprising an hiv-1 envelope targeting arm
IL262396B2 (en) 2016-04-15 2023-09-01 Macrogenics Inc Novel b7-h3 binding molecules, antibody drug conjugates thereof and methods of use thereof
WO2020154475A1 (en) 2019-01-23 2020-07-30 Molecular Templates, Inc. Proteins comprising modified immunoglobulin variable light chains

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA1339198C (en) * 1988-02-12 1997-08-05 Gregory Paul Winter Antibodies to the antigen campath-1

Also Published As

Publication number Publication date
EP0575407A1 (en) 1993-12-29
WO1992016562A1 (en) 1992-10-01
GB9105245D0 (en) 1991-04-24
JPH06510659A (en) 1994-12-01

Similar Documents

Publication Publication Date Title
CA2105980A1 (en) Humanised antibodies having modified allotypic determinants
CA2212750C (en) Humanized antibodies against cd3
AU694926B2 (en) Humanised antibodies
US5712120A (en) Method for obtaining modified immunoglobulins with reduced immunogenicity of murine antibody variable domains, compositions containing them
US6797492B2 (en) Method for reducing the immunogenicity of antibody variable domains
KR101276596B1 (en) Humanized anti-cd4 antibody with immunosuppressive properties
JP4804357B2 (en) Modified anti-CD52 antibody
KR100362340B1 (en) Recombinant IL4 Antibody Useful for the Treatment of Interleukin-4 (IL4) Mediated Disease
JP2004073210A (en) Modified antibody and product and process relating thereto
Kabat Origins of antibody complementarity and specificity--hypervariable regions and minigene hypothesis.
JP4574750B2 (en) Antibody variant
JPH05502384A (en) Preparation of antibodies
JP2005538706A (en) Super humanized antibody
JPH10509876A (en) Anthropomorphic antibody against CD38
GB2216126A (en) Antibodies to Campath-1
Waisman et al. The pathogenic human monoclonal anti-DNA that induces experimental systemic lupus erythematosus in mice is encoded by a VH4 gene segment
McKean et al. Mechanisms of antibody diversity: Multiple genes encode structurally related mouse κ variable regions
CA2106719C (en) Reshaped monoclonal antibodies against an immunoglobulin isotype
Von Feldt et al. Bioactive peptide design based on antibody structure
Kipps et al. Homologous chromosome recombination generating immunoglobulin allotype and isotype switch variants.
Parhami-Seren et al. Clustered H chain somatic mutations shared by anti-p-azophenylarsonate antibodies confer enhanced affinity and ablate the cross-reactive idiotype.
Thielemans et al. Syngeneic antiidiotypic immune responses to a B cell lymphoma. Comparison between heavy chain hypervariable region peptides and intact Ig as immunogens.
EP0511308B2 (en) Chimeric immunoglobulin for cd4 receptors
JP2001510329A (en) Human monoclonal antibody
AU719369B2 (en) Monoclonal antiidiotypic antibodies (AB2) and their uses

Legal Events

Date Code Title Description
FZDE Dead